Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Res ; 32(7): 670-686, 2022 07.
Article in English | MEDLINE | ID: mdl-35296796

ABSTRACT

Diet can impact on gut health and disease by modulating intestinal stem cells (ISCs). However, it is largely unknown if and how the ISC niche responds to diet and influences ISC function. Here, we demonstrate that Lepr+ mesenchymal cells (MCs) surrounding intestinal crypts sense diet change and provide a novel niche signal to maintain ISC and progenitor cell proliferation. The abundance of these MCs increases upon administration of a high-fat diet (HFD) but dramatically decreases upon fasting. Depletion of Lepr+ MCs resulted in fewer intestinal stem/progenitor cells, compromised the architecture of crypt-villus axis and impaired intestinal regeneration. Furthermore, we showed that IGF1 secreted by Lepr+ MCs is an important effector that promotes proliferation of ISCs and progenitor cells in the intestinal crypt. We conclude that Lepr+ MCs sense diet alterations and, in turn, modulate intestinal stem/progenitor cell function via a stromal IGF1-epithelial IGF1R axis. These findings reveal that Lepr+ MCs are important mediators linking systemic diet changes to local ISC function and might serve as a novel therapeutic target for gut diseases.


Subject(s)
Leptin , Mesenchymal Stem Cells , Diet , Intestinal Mucosa , Stem Cells/physiology
2.
Food Funct ; 12(21): 10700-10713, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34605504

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic intestinal inflammation that is currently incurable. Increasing evidence indicates that supplementation with probiotics could improve the symptoms of IBD. It is scientifically significant to identify novel and valid strains for treating IBD. It has been reported that the probiotic Lactobacillus paracasei L9 (L9), which is identified from the gut of healthy centenarians, can modulate host immunity and plays an anti-allergic role. Here, we demonstrated that L9 alleviates the pathological phenotypes of experimental colitis by expanding the abundance of butyrate-producing bacteria. Oral administration of sodium butyrate in experimental colitis recapitulates the L9 anti-inflammatory phenotypes. Mechanistically, sodium butyrate ameliorated the inflammatory responses by inhibiting the IL-6/STAT3 signaling pathway in colitis. Overall, these findings demonstrated that L9 alleviates the DSS-induced colitis development by enhancing the abundance of butyrate-producing bacterial strains that produce butyrate to suppress the IL-6/STAT3 signaling pathway, providing new insight into a promising therapeutic target for the remission of IBD.


Subject(s)
Colitis/chemically induced , Colitis/therapy , Interleukin-6/metabolism , Lacticaseibacillus paracasei , Probiotics/therapeutic use , STAT3 Transcription Factor/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Butyrates , Butyric Acid/administration & dosage , Butyric Acid/pharmacology , Dextran Sulfate/toxicity , Female , Gene Expression Regulation/drug effects , Histamine Antagonists/administration & dosage , Histamine Antagonists/pharmacology , Inflammation/drug therapy , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Random Allocation , STAT3 Transcription Factor/genetics
3.
Front Cell Dev Biol ; 9: 691050, 2021.
Article in English | MEDLINE | ID: mdl-34336839

ABSTRACT

Myoepithelial and luminal cells synergistically expand in the mammary gland during pregnancy, and this process is precisely governed by hormone-related signaling pathways. The bone morphogenetic protein (BMP) signaling pathway is now known to play crucial roles in all organ systems. However, the functions of BMP signaling in the mammary gland remain unclear. Here, we found that BMPR1a is upregulated by hormone-induced Sp1 at pregnancy. Using a doxycycline (Dox)-inducible BMPR1a conditional knockout mouse model, we demonstrated that loss of BMPR1a in myoepithelium results in compromised myoepithelial integrity, reduced mammary stem cells and precocious alveolar differentiation during pregnancy. Mechanistically, BMPR1a regulates the expression of p63 and Slug, two key regulators of myoepithelial maintenance, through pSmad1/5-Smad4 complexes, and consequently activate P-cadherin during pregnancy. Furthermore, we observed that loss of BMPR1a in myoepithelium results in the upregulation of a secreted protein Spp1 that could account for the precocious alveolar differentiation in luminal layer, suggesting a defective basal-to-luminal paracrine signaling mechanism. Collectively, these findings identify a novel role of BMP signaling in maintaining the identity of myoepithelial cells and suppressing precocious alveolar formation.

4.
Oncogene ; 40(29): 4832-4845, 2021 07.
Article in English | MEDLINE | ID: mdl-34155343

ABSTRACT

Metastasis is the main cause of death in breast cancer patients. The initial step of metastasis is invadopodia-mediated extracellular matrix (ECM) degradation, which enables local breast tumor cells to invade surrounding tissues. However, the molecular mechanism underlying invadopodia-mediated metastasis remains largely unknown. Here we found that the RNA-binding protein Musashi1 (Msi1) exhibited elevated expression in invasive breast tumors and promoted lung metastasis of mammary cancer cells. Suppression of Msi1 reduced invadopodia formation in mammary cancer cells. Furthermore, Msi1 deficiency decreased the expression and activity of Mmp9, an important enzyme in ECM degradation. Mechanistically, Msi1 directly suppressed Timp3, an endogenous inhibitor of Mmp9. In clinical breast cancer specimens, TIMP3 and MSI1 levels were significantly inversely correlated both in normal breast tissue and breast cancer tissues and associated with overall survival in breast cancer patients. Taken together, our findings demonstrate that the MSI1-TIMP3-MMP9 cascade is critical for invadopodia-mediated onset of metastasis in breast cancer, providing novel insights into a promising therapeutic strategy for breast cancer metastasis.


Subject(s)
Breast Neoplasms , Humans , Podosomes , Tissue Inhibitor of Metalloproteinase-3
5.
Theranostics ; 11(8): 3607-3623, 2021.
Article in English | MEDLINE | ID: mdl-33664851

ABSTRACT

Background: Brown adipose tissue (BAT) dissipates chemical energy as heat and has the potential to be a protective strategy to prevent obesity. microRNAs (miRNAs) are emerging as important posttranscriptional factors affecting the thermogenic function of BAT. However, the regulatory mechanism underlying miRNA-mediated energy metabolism in BAT is not fully understood. Here, we explored the roles of miR-22 in BAT thermogenesis and energy metabolism. Methods: Using global and conditional knockout mice as in vivo models and primary brown adipocytes as an in vitro system, we investigated the function of miR-22 in BAT thermogenesis in vivo and in vitro. Results: miR-22 expression was upregulated in BAT in response to cold exposure and during brown preadipocyte differentiation. Both global and conditional knockout mice displayed BAT whitening, impaired cold tolerance, and decreased BAT thermogenesis. Moreover, we found that miR-22 deficiency impaired BAT glycolytic capacity, which is critical for thermogenesis. The mechanistic results revealed that miR-22 activated the mTORC1 signaling pathway by directly suppressing Tsc1 and concomitantly directly suppressing Hif1an, an inhibitor of Hif1α, which promotes glycolysis and maintains thermogenesis. Conclusions: Our findings identify miR-22 as a critical regulator in the control of thermogenesis in BAT and as a potential therapeutic target for human metabolic disorders.


Subject(s)
Adipocytes, Brown/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , MicroRNAs/genetics , Thermogenesis/genetics , Adipose Tissue, Brown/metabolism , Animals , Cells, Cultured , Cold Temperature , Diet, High-Fat/adverse effects , Glycolysis , In Vitro Techniques , Insulin Resistance/genetics , Male , Mice , Mice, Knockout , MicroRNAs/metabolism , Mixed Function Oxygenases/metabolism , Precision Medicine , Signal Transduction , Thermogenesis/physiology , Tuberous Sclerosis Complex 1 Protein/metabolism , Up-Regulation
7.
Cell Rep ; 32(4): 107952, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726617

ABSTRACT

A certain number of epithelial cells in intestinal crypts are DNA damage resistant and contribute to regeneration. However, the cellular mechanism underlying intestinal regeneration remains unclear. Using lineage tracing, we show that cells marked by an Msi1 reporter (Msi1+) are right above Lgr5high cells in intestinal crypts and exhibit DNA damage resistance. Single-cell RNA sequencing reveals that the Msi1+ cells are heterogeneous with the majority being intestinal stem cells (ISCs). The DNA damage-resistant subpopulation of Msi1+ cells is characterized by low-to-negative Lgr5 expression and is more rapidly cycling than Lgr5high radiosensitive crypt base columnar stem cells (CBCs). This enables an efficient repopulation of the intestinal epithelium at early stage when Lgr5high cells are not emerging. Furthermore, relative to CBCs, Msi1+ cells preferentially produce Paneth cells during homeostasis and upon radiation repair. Together, we demonstrate that the DNA damage-resistant Msi1+ cells are cycling ISCs that maintain and regenerate the intestinal epithelium.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/physiology , Stem Cells/metabolism , Animals , Cell Lineage/genetics , Female , Homeostasis , Intestinal Mucosa/radiation effects , Intestines/radiation effects , Male , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Paneth Cells/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Radiation Tolerance , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Regeneration/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
8.
Stem Cell Reports ; 15(2): 374-388, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32649903

ABSTRACT

Intestinal regeneration is crucial for functional restoration after injury, and nutritional molecules can play an important role in this process. Here, we found that arachidonic acid (AA) serves as a direct proliferation promoter of intestinal epithelial cells that facilitates small intestinal regeneration in both three-dimensional cultured organoids and mouse models. As shown in the study, during post-irradiation regeneration, AA positively regulates intestinal epithelial cell proliferation by upregulating the expression of Ascl2 and activating WNT signaling, but negatively regulates intestinal epithelial cell differentiation. AA acts as a delicate regulator that efficiently facilitates epithelial tissue repair by activating radiation-resistant Msi1+ cells rather than Lgr5+ cells, which are extensively considered WNT-activated crypt base stem cells. Additionally, short-term AA treatment maintains optimal intestinal epithelial homeostasis under physiological conditions. As a result, AA treatment can be considered a potential therapy for irradiation injury repair and tissue regeneration.


Subject(s)
Arachidonic Acid/pharmacology , Intestine, Small/physiology , Regeneration/drug effects , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/radiation effects , Intestine, Small/cytology , Male , Mice, Inbred C57BL , Organoids/cytology , Radiation, Ionizing , Regeneration/radiation effects , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/radiation effects , Transcriptome/genetics , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/radiation effects
9.
Cell Res ; 30(10): 854-872, 2020 10.
Article in English | MEDLINE | ID: mdl-32457396

ABSTRACT

Mammary and extramammary Paget's Diseases (PD) are a malignant skin cancer characterized by the appearance of Paget cells. Although easily diagnosed, its pathogenesis remains unknown. Here, single-cell RNA-sequencing identified distinct cellular states, novel biomarkers, and signaling pathways - including mTOR, associated with extramammary PD. Interestingly, we identified MSI1 ectopic overexpression in basal epithelial cells of human PD skin, and show that Msi1 overexpression in the epidermal basal layer of mice phenocopies human PD at histopathological, single-cell and molecular levels. Using this mouse model, we identified novel biomarkers of Paget-like cells that translated to human Paget cells. Furthermore, single-cell trajectory, RNA velocity and lineage-tracing analyses revealed a putative keratinocyte-to-Paget-like cell conversion, supporting the in situ transformation theory of disease pathogenesis. Mechanistically, the Msi1-mTOR pathway drives keratinocyte-Paget-like cell conversion, and suppression of mTOR signaling with Rapamycin significantly rescued the Paget-like phenotype in Msi1-overexpressing transgenic mice. Topical Rapamycin treatment improved extramammary PD-associated symptoms in humans, suggesting mTOR inhibition as a novel therapeutic treatment in PD.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Nerve Tissue Proteins/metabolism , Paget Disease, Extramammary/drug therapy , RNA-Binding Proteins/metabolism , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Aged , Animals , Biomarkers/metabolism , Female , Humans , Male , Mice , Middle Aged
10.
EMBO J ; 39(7): e103255, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32128839

ABSTRACT

The Hippo-YAP signaling pathway plays an essential role in epithelial cells during intestinal regeneration and tumorigenesis. However, the molecular mechanism linking stromal signals to YAP-mediated intestinal regeneration and tumorigenesis is poorly defined. Here, we report a stroma-epithelium ISLR-YAP signaling axis essential for stromal cells to modulate epithelial cell growth during intestinal regeneration and tumorigenesis. Specifically, upon inflammation and in cancer, an oncogenic transcription factor ETS1 in stromal cells induces expression of a secreted protein ISLR that can inhibit Hippo signaling and activate YAP in epithelial cells. Deletion of Islr in stromal cells in mice markedly impaired intestinal regeneration and suppressed tumorigenesis in the colon. Moreover, the expression of stromal cell-specific ISLR and ETS1 significantly increased in inflamed mucosa of human IBD patients and in human colorectal adenocarcinoma, accounting for the epithelial YAP hyperactivation. Collectively, our findings provide new insights into the signaling crosstalk between stroma and epithelium during tissue regeneration and tumorigenesis.


Subject(s)
Colorectal Neoplasms/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Inflammatory Bowel Diseases/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Animals , Colorectal Neoplasms/genetics , Disease Models, Animal , Gene Knockout Techniques , HCT116 Cells , HEK293 Cells , HT29 Cells , Hippo Signaling Pathway , Humans , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/metabolism , Male , Mice , Mutation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
11.
Gastroenterology ; 156(8): 2281-2296.e6, 2019 06.
Article in English | MEDLINE | ID: mdl-30779922

ABSTRACT

BACKGROUND & AIMS: Levels of microRNA 31 (MIR31) are increased in intestinal tissues from patients with inflammatory bowel diseases and colitis-associated neoplasias. We investigated the effects of this microRNA on intestinal inflammation by studying mice with colitis. METHODS: We obtained colon biopsy samples from 82 patients with ulcerative colitis (UC), 79 patients with Crohn's disease (CD), and 34 healthy individuals (controls) at Shanghai Tenth People's Hospital. MIR31- knockout mice and mice with conditional disruption of Mir31 specifically in the intestinal epithelium (MIR31 conditional knockouts) were given dextran sulfate sodium (DSS) or 2,4,6-trinitrobenzene sulfonic acid (TNBS) to induce colitis. We performed chromatin immunoprecipitation and luciferase assays to study proteins that regulate expression of MIR31, including STAT3 and p65, in LOVO colorectal cancer cells and organoids derived from mouse colon cells. Partially hydrolyzed alpha-lactalbumin was used to generate peptosome nanoparticles, and MIR31 mimics were loaded onto their surface using electrostatic adsorption. Peptosome-MIR31 mimic particles were encapsulated into oxidized konjac glucomannan (OKGM) microspheres, which were administered by enema into the large intestines of mice with DSS-induced colitis. Intestinal tissues were collected and analyzed by histology and immunohistochemistry. RESULTS: Levels of MIR31 were increased in inflamed mucosa from patients with CD or UC, and from mice with colitis, compared with controls. STAT3 and nuclear factor-κB activated transcription of MIR31 in colorectal cancer cells and organoids in response to tumor necrosis factor and interleukin (IL)6. MIR31-knockout and conditional-knockout mice developed more severe colitis in response to DSS and TNBS, with increased immune responses, compared with control mice. MIR31 bound to 3' untranslated regions of Il17ra and Il7r messenger RNAs (RNAs) (which encode receptors for the inflammatory cytokines IL17 and IL7) and Il6st mRNA (which encodes GP130, a cytokine signaling protein). These mRNAs and proteins were greater in MIR31-knockout mice with colitis, compared with control mice; MIR31 and MIR31 mimics inhibited their expression. MIR31 also promoted epithelial regeneration by regulating the WNT and Hippo signaling pathways. OKGM peptosome-MIR31 mimic microspheres localized to colonic epithelial cells in mice with colitis; they reduced the inflammatory response, increased body weight and colon length, and promoted epithelial cell proliferation. CONCLUSIONS: MIR31, increased in colon tissues from patients with CD or UC, reduces the inflammatory response in colon epithelium of mice by preventing expression of inflammatory cytokine receptors (Il7R and Il17RA) and signaling proteins (GP130). MIR31 also regulates the WNT and Hippo signaling pathways to promote epithelial regeneration following injury. OKGM peptosome-MIR31 microspheres localize to the colon epithelium of mice to reduce features of colitis. Transcript Profiling: GSE123556.


Subject(s)
Biomarkers/metabolism , Colitis, Ulcerative/pathology , Crohn Disease/pathology , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , Regeneration/physiology , Animals , Biopsy, Needle , Case-Control Studies , China , Disease Models, Animal , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Microspheres , RNA, Messenger/metabolism , Random Allocation , Signal Transduction
12.
Nat Commun ; 8(1): 1036, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051494

ABSTRACT

MicroRNA-mediated post-transcriptional regulation plays key roles in stem cell self-renewal and tumorigenesis. However, the in vivo functions of specific microRNAs in controlling mammary stem cell (MaSC) activity and breast cancer formation remain poorly understood. Here we show that miR-31 is highly expressed in MaSC-enriched mammary basal cell population and in mammary tumors, and is regulated by NF-κB signaling. We demonstrate that miR-31 promotes mammary epithelial proliferation and MaSC expansion at the expense of differentiation in vivo. Loss of miR-31 compromises mammary tumor growth, reduces the number of cancer stem cells, as well as decreases tumor-initiating ability and metastasis to the lung, supporting its pro-oncogenic function. MiR-31 modulates multiple signaling pathways, including Prlr/Stat5, TGFß and Wnt/ß-catenin. Particularly, it activates Wnt/ß-catenin signaling by directly targeting Wnt antagonists, including Dkk1. Importantly, Dkk1 overexpression partially rescues miR31-induced mammary defects. Together, these findings identify miR-31 as the key regulator of MaSC activity and breast tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Stem Cells/cytology , Wnt Proteins/genetics , Wnt Signaling Pathway , beta Catenin/metabolism
13.
Elife ; 62017 09 05.
Article in English | MEDLINE | ID: mdl-28870287

ABSTRACT

Intestinal regeneration and tumorigenesis are believed to be driven by intestinal stem cells (ISCs). Elucidating mechanisms underlying ISC activation during regeneration and tumorigenesis can help uncover the underlying principles of intestinal homeostasis and disease including colorectal cancer. Here we show that miR-31 drives ISC proliferation, and protects ISCs against apoptosis, both during homeostasis and regeneration in response to ionizing radiation injury. Furthermore, miR-31 has oncogenic properties, promoting intestinal tumorigenesis. Mechanistically, miR-31 acts to balance input from Wnt, BMP, TGFß signals to coordinate control of intestinal homeostasis, regeneration and tumorigenesis. We further find that miR-31 is regulated by the STAT3 signaling pathway in response to radiation injury. These findings identify miR-31 as a critical modulator of ISC biology, and a potential therapeutic target for a broad range of intestinal regenerative disorders and cancers.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Intestines/pathology , MicroRNAs/metabolism , Regeneration/genetics , Stem Cells/metabolism , Stress, Physiological , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Epithelial Cells/metabolism , Epithelium/metabolism , Gamma Rays , Gene Expression Regulation , HCT116 Cells , Humans , Mice, Transgenic , MicroRNAs/genetics , Models, Biological , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
14.
Talanta ; 170: 406-412, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28501188

ABSTRACT

A new ratiometric fluorescent probe based on a hemicyanine dye was synthesized. Investigation on the response behavior towards amino acids showed that the probe had a capability of rapidly and selectively detecting cysteine over other biothiols such as glutathione and homocysteine with similar structure and reactivity, and it also displayed a high selectivity and rapid response. Moreover, the probe had good water solubility, which authorized it could be applied in fluorescent bioimaging. The laser confocal fluorescence images indicated that the probe could visualize the intracellular cysteine. This work suggested that it could be utilized as a fluorescent indicator to discriminate the endogenous cysteine in living cells.


Subject(s)
Carbocyanines/chemistry , Cysteine/analysis , Fluorescent Dyes/chemistry , Optical Imaging/methods , Colorimetry/methods , Glutathione/analysis , HeLa Cells , Humans , Models, Molecular , Spectrometry, Fluorescence/methods
15.
FASEB J ; 30(10): 3474-3488, 2016 10.
Article in English | MEDLINE | ID: mdl-27383182

ABSTRACT

Mammary epithelium is comprised of an inner layer of luminal epithelial cells and an outer layer of contractile myoepithelial cells with mesenchymal properties. These two compartments interact throughout mammary morphogenesis to form branching ducts during puberty and terminate in secretory alveoli during lactation. It is not known how the myoepithelial cell lineage is specified, nor how signals in myoepithelial cells contribute to lactogenesis. Here, we show that Numb and Numbl are enriched in mammary myoepithelial cells, with their expression peaking during pregnancy. We use conditional Numb- and Numbl-knockout mouse models to demonstrate that loss of Numb/Numbl compromised the myoepithelial layer and expanded the luminal layer, led epithelial cells to undergo epithelial-to-mesenchymal transition, and resulted in lactation failure as a result of abnormal alveolar formation during pregnancy. Numb and Numbl function via repression of the Notch signaling pathway and of the p53-p21 axis during mammary gland development. These findings highlight the importance of Numb and Numbl in the control of myoepithelial cell fate determination, epithelial identity, and lactogenesis.-Zhang Y., Li, F., Song, Y., Sheng, X., Ren, F., Xiong, K., Chen, L., Zhang, H., Liu, D., Lengner, C. J., Xue, L., Yu, Z. Numb and Numbl act to determine mammary myoepithelial cell fate, maintain epithelial identity, and support lactogenesis.


Subject(s)
Cell Differentiation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Breast/metabolism , Cell Lineage , Epithelial Cells/cytology , Epithelium/metabolism , Female , Humans , Mice, Transgenic , Muscle Cells/cytology , Muscle, Smooth/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...