Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 898
Filter
1.
Adv Protein Chem Struct Biol ; 141: 331-360, 2024.
Article in English | MEDLINE | ID: mdl-38960479

ABSTRACT

We recently identified TMEM230 as a master regulator of the endomembrane system of cells. TMEM230 expression is necessary for promoting motor protein dependent intracellular trafficking of metalloproteins for cellular energy production in mitochondria. TMEM230 is also required for transport and secretion of metalloproteinases for autophagy and phagosome dependent clearance of misfolded proteins, defective RNAs and damaged cells, activities that decline with aging. This suggests that aberrant levels of TMEM230 may contribute to aging and regain of proper levels may have therapeutic applications. The components of the endomembrane system include the Golgi complex, other membrane bound organelles, and secreted vesicles and factors. Secreted cellular components modulate immune response and tissue regeneration in aging. Upregulation of intracellular packaging, trafficking and secretion of endosome components while necessary for tissue homeostasis and normal wound healing, also promote secretion of pro-inflammatory and pro-senescence factors. We recently determined that TMEM230 is co-regulated with trafficked cargo of the endomembrane system, including lysosome factors such as RNASET2. Normal tissue regeneration (in aging), repair (following injury) and aberrant destructive tissue remodeling (in cancer or autoimmunity) likely are regulated by TMEM230 activities of the endomembrane system, mitochondria and autophagosomes. The role of TMEM230 in aging is supported by its ability to regulate the pro-inflammatory secretome and senescence-associated secretory phenotype in tissue cells of patients with advanced age and chronic disease. Identifying secreted factors regulated by TMEM230 in young patients and patients of advanced age will facilitate identification of aging associated targets that aberrantly promote, inhibit or reverse aging. Ex situ culture of patient derived cells for identifying secreted factors in tissue regeneration and aging provides opportunities in developing therapeutic and personalized medicine strategies. Identification and validation of human secreted factors in tissue regeneration requires long-term stabile scaffold culture conditions that are different from those previously reported for cell lines used as cell models for aging. We describe a 3 dimensional (3D) platform utilizing non-biogenic and non-labile poly ε-caprolactone scaffolds that supports maintenance of long-term continuous cultures of human stem cells, in vitro generated 3D organoids and patient derived tissue. Combined with animal component free culture media, non-biogenic scaffolds are suitable for proteomic and glycobiological analyses to identify human factors in aging. Applications of electrospun nanofiber technologies in 3D cell culture allow for ex situ screening and the development of patient personalized therapeutic strategies and predicting their effectiveness in mitigating or promoting aging.


Subject(s)
Aging , Organoids , Humans , Organoids/metabolism , Aging/metabolism , Membrane Proteins/metabolism , Cellular Senescence , Female , Tissue Scaffolds/chemistry , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology
2.
PLoS One ; 19(6): e0305421, 2024.
Article in English | MEDLINE | ID: mdl-38870243

ABSTRACT

Human milk is optimal for infant nutrition. However, many mothers cease breastfeeding because of low milk supply (LMS). It is difficult to identify mothers at risk for LMS because its biologic underpinnings are not fully understood. Previously, we demonstrated that milk micro-ribonucleic acids (miRNAs) may be related to LMS. Transforming growth factor beta (TGFß) also plays an important role in mammary involution and may contribute to LMS. We performed a longitudinal cohort study of 139 breastfeeding mothers to test the hypothesis that milk levels of TGFß would identify mothers with LMS. We explored whether TGFß impacts the expression of LMS-related miRNAs in cultured human mammary epithelial cells (HMECs). LMS was defined by maternal report of inadequate milk production, and confirmed by age of formula introduction and infant weight trajectory. Levels of TGF-ß1 and TGF-ß2 were measured one month after delivery. There was a significant relationship between levels of TGF-ß1 and LMS (X2 = 8.92, p = 0.003) on logistic regression analysis, while controlling for lactation stage (X2 = 1.28, p = 0.25), maternal pre-pregnancy body mass index (X2 = 0.038, p = 0.84), and previous breastfeeding experience (X2 = 7.43, p = 0.006). The model accounted for 16.8% of variance in the data (p = 0.005) and correctly predicted LMS for 84.6% of mothers (22/26; AUC = 0.72). Interactions between TGF-ß1 and miR-22-3p displayed significant effect on LMS status (Z = 2.67, p = 0.008). Further, incubation of HMECs with TGF-ß1 significantly reduced mammary cell number (t = -4.23, p = 0.003) and increased levels of miR-22-3p (t = 3.861, p = 0.008). Interactions between TGF-ß1 and miR-22-3p may impact mammary function and milk levels of TGF-ß1 could have clinical utility for identifying mothers with LMS. Such information could be used to provide early, targeted lactation support.


Subject(s)
Breast Feeding , MicroRNAs , Milk, Human , Transforming Growth Factor beta1 , Humans , Female , Milk, Human/metabolism , Transforming Growth Factor beta1/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Adult , Lactation , Transforming Growth Factor beta2/metabolism , Longitudinal Studies , Epithelial Cells/metabolism , Infant , Mothers , Infant, Newborn , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology
3.
Ann Endocrinol (Paris) ; 85(3): 248-251, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38871512

ABSTRACT

Adipose tissue is highly plastic, as illustrated mainly by the transdifferentiation of white adipocytes into beige adipocytes, depending on environmental conditions. However, during gestation and lactation in rodent, there is an amazing phenomenon of transformation of subcutaneous adipose tissue into mammary glandular tissue, known as pink adipose tissue, capable of synthesizing and secreting milk. Recent work using transgenic lineage-tracing experiments, mainly carried out in Saverio Cinti's team, has demonstrated very convincingly that this process does indeed correspond to a transdifferentiation of white adipocytes into mammary alveolar cells (pink adipocytes) during gestation and lactation. This phenomenon is reversible, since during the post-lactation phase, pink adipocytes revert to the white adipocyte phenotype. The molecular mechanisms underlying this reversible transdifferentiation remain poorly understood.


Subject(s)
Adipose Tissue , Lactation , Animals , Humans , Female , Adipose Tissue/physiology , Adipose Tissue/metabolism , Adipose Tissue/cytology , Lactation/physiology , Pregnancy , Cell Transdifferentiation/physiology , Mammary Glands, Animal/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Adipocytes, White/physiology , Adipocytes, White/metabolism , Adipocytes, White/cytology , Cell Plasticity/physiology , Mammary Glands, Human/physiology , Mammary Glands, Human/growth & development , Mammary Glands, Human/cytology , Adipocytes/physiology , Adipocytes/cytology
4.
Stem Cells Dev ; 33(17-18): 449-467, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38943275

ABSTRACT

The origin of breast cancer (BC) has traditionally been a focus of medical research. It is widely acknowledged that BC originates from immortal mammary stem cells and that these stem cells participate in two division modes: symmetric cell division (SCD) and asymmetrical cell division (ACD). Although both of these modes are key to the process of breast development and their imbalance is closely associated with the onset of BC, the molecular mechanisms underlying these phenomena deserve in-depth exploration. In this review, we first outline the molecular mechanisms governing ACD/SCD and analyze the role of ACD/SCD in various stages of breast development. We describe that the changes in telomerase activity, the role of polar proteins, and the stimulation of ovarian hormones subsequently lead to two distinct consequences: breast development or carcinogenesis. Finally, gene mutations, abnormalities in polar proteins, modulation of signal-transduction pathways, and alterations in the microenvironment disrupt the balance of BC stem cell division modes and cause BC. Important regulatory factors such as mammalian Inscuteable mInsc, Numb, Eya1, PKCα, PKCθ, p53, and IL-6 also play significant roles in regulating pathways of ACD/SCD and may constitute key targets for future research on stem cell division, breast development, and tumor therapy.


Subject(s)
Breast Neoplasms , Carcinogenesis , Mammary Glands, Human , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Animals , Mammary Glands, Human/growth & development , Mammary Glands, Human/pathology , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Carcinogenesis/pathology , Carcinogenesis/metabolism , Carcinogenesis/genetics , Stem Cells/metabolism , Stem Cells/cytology , Cell Division , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mammary Glands, Animal/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Signal Transduction
5.
J Cell Biochem ; 125(7): e30606, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38779980

ABSTRACT

The Hippo pathway, a signaling cascade involved in the regulation of organ size and several other processes, acts as a conduit between extracellular matrix (ECM) cues and cellular responses. We asked whether the basement membrane (BM), a specialized ECM component known to induce quiescence and differentiation in mammary epithelial cells, would regulate the localization, activity, and interactome of YAP, a Hippo pathway effector. To address this question, we used a broad range of experimental approaches, including 2D and 3D cultures of both mouse and human mammary epithelial cells, as well as the developing mouse mammary gland. In contrast to malignant cells, nontumoral cells cultured with a reconstituted BM (rBM) displayed higher concentrations of YAP in the cytoplasm. Incidentally, when in the nucleus of rBM-treated cells, YAP resided preferentially at the nuclear periphery. In agreement with our cell culture experiments, YAP exhibited cytoplasmic predominance in ductal cells of developing mammary epithelia, where a denser BM is found. Conversely, terminal end bud (TEB) cells with a thinner BM displayed higher nucleus-to-cytoplasm ratios of YAP. Bioinformatic analysis revealed that genes regulated by YAP were overrepresented in the transcriptomes of microdissected TEBs. Consistently, mouse epithelial cells exposed to the rBM expressed lower levels of YAP-regulated genes, although the protein level of YAP and Hippo components were slightly altered by the treatment. Mass spectrometry analysis identified a differential set of proteins interacting with YAP in cytoplasmic fractions of mouse epithelial cells in the absence or presence of rBM. In untreated cells, YAP interactants were enriched in processes related to ubiquitin-mediated proteolysis, whereas in cells exposed to rBM YAP interactants were mainly key proteins related to amino acid, amino sugar, and carbohydrate metabolism. Collectively, we unraveled that the BM induces YAP translocation or retention in the cytoplasm of nontumoral epithelial cells and that in the cytoplasm YAP seems to undertake novel functions in metabolic pathways.


Subject(s)
Adaptor Proteins, Signal Transducing , Basement Membrane , Cytoplasm , Epithelial Cells , Transcription Factors , YAP-Signaling Proteins , Animals , Humans , Mice , Epithelial Cells/metabolism , YAP-Signaling Proteins/metabolism , Female , Cytoplasm/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Basement Membrane/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology , Cell Nucleus/metabolism , Signal Transduction
6.
Cells ; 13(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38786098

ABSTRACT

Breast cancer develops upon sequential acquisition of driver mutations in mammary epithelial cells; however, how these mutations collaborate to transform normal cells remains unclear in most cases. We aimed to reconstitute this process in a particular case. To this end, we combined the activated form of the PI 3-kinase harboring the H1047R mutation with the inactivation of the histone lysine methyl-transferase KMT2D in the non-tumorigenic human mammary epithelial cell line MCF10A. We found that PI 3-kinase activation promoted cell-cycle progression, especially when growth signals were limiting, as well as cell migration, both in a collective monolayer and as single cells. Furthermore, we showed that KMT2D inactivation had relatively little influence on these processes, except for single-cell migration, which KMT2D inactivation promoted in synergy with PI 3-kinase activation. The combination of these two genetic alterations induced expression of the ARPC5L gene that encodes a subunit of the Arp2/3 complex. ARPC5L depletion fully abolished the enhanced migration persistence exhibited by double-mutant cells. Our reconstitution approach in MCF10A has thus revealed both the cell function and the single-cell migration, and the underlying Arp2/3-dependent mechanism, which are synergistically regulated when KMT2D inactivation is combined with the activation of the PI 3-kinase.


Subject(s)
Actin-Related Protein 2-3 Complex , Cell Movement , Epithelial Cells , Histone-Lysine N-Methyltransferase , Phosphatidylinositol 3-Kinases , Humans , Cell Movement/genetics , Epithelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , Female , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Mutation/genetics , Cell Line
7.
J Mammary Gland Biol Neoplasia ; 29(1): 11, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38761238

ABSTRACT

The transcription factor STAT3 is activated by multiple cytokines and other extrinsic factors. It plays a key role in immune and inflammatory responses and, when dysregulated, in tumourigenesis. STAT3 is also an indispensable mediator of the cell death process that occurs during post-lactational regression of the mammary gland, one of the most dramatic examples of physiological cell death in adult mammals. During this involution of the gland, STAT3 powerfully enhances the lysosomal system to efficiently remove superfluous milk-producing mammary epithelial cells via a lysosomal-mediated programmed cell death pathway. The lysosome is a membrane-enclosed  cytoplasmic organelle that digests and recycles cellular waste, with an important role as a signalling centre that monitors cellular metabolism. Here, we describe key strategies for investigating the role of STAT3 in regulating lysosomal function using a mammary epithelial cell culture model system. These include protocols for lysosome enrichment and enzyme activity assays, in addition to microscopic analyses of the vesicular compartment in cell lines. Collectively, these approaches provide the tools to investigate multiple aspects of lysosome biogenesis and function, and to define both direct and indirect roles for STAT3.


Subject(s)
Epithelial Cells , Lysosomes , Mammary Glands, Animal , STAT3 Transcription Factor , Lysosomes/metabolism , STAT3 Transcription Factor/metabolism , Female , Animals , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology , Mice , Signal Transduction
8.
J Vis Exp ; (207)2024 May 03.
Article in English | MEDLINE | ID: mdl-38767371

ABSTRACT

The mammary gland is a fundamental structure of the breast and plays an essential role in reproduction. Human mammary epithelial cells (HMECs), which are the origin cells of breast cancer and other breast-related inflammatory diseases, have garnered considerable attention. However, isolating and culturing primary HMECs in vitro for research purposes has been challenging due to their highly differentiated, keratinized nature and their short lifespan. Therefore, developing a simple and efficient method to isolate and culture HMECs is of great scientific value for the study of breast biology and breast-related diseases. In this study, we successfully isolated primary HMECs from small amounts of mammary tissue by digestion with a mixture of enzymes combined with an initial culture in 5% fetal bovine serum-DMEM containing the Rho-associated kinase (ROCK) inhibitor Y-27632, followed by culture expansion in serum-free keratinocyte medium. This approach selectively promotes the growth of epithelial cells, resulting in an optimized cell yield. The simplicity and convenience of this method make it suitable for both laboratory and clinical research, which should provide valuable insights into these important areas of study.


Subject(s)
Cell Culture Techniques , Epithelial Cells , Mammary Glands, Human , Humans , Epithelial Cells/cytology , Female , Mammary Glands, Human/cytology , Cell Culture Techniques/methods , Amides/pharmacology , Pyridines/pharmacology , Cytological Techniques/methods , rho-Associated Kinases/antagonists & inhibitors
9.
Biomater Adv ; 160: 213847, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657288

ABSTRACT

Three-dimensional (3D) organoid models have been instrumental in understanding molecular mechanisms responsible for many cellular processes and diseases. However, established organic biomaterial scaffolds used for 3D hydrogel cultures, such as Matrigel, are biochemically complex and display significant batch variability, limiting reproducibility in experiments. Recently, there has been significant progress in the development of synthetic hydrogels for in vitro cell culture that are reproducible, mechanically tuneable, and biocompatible. Self-assembling peptide hydrogels (SAPHs) are synthetic biomaterials that can be engineered to be compatible with 3D cell culture. Here we investigate the ability of PeptiGel® SAPHs to model the mammary epithelial cell (MEC) microenvironment in vitro. The positively charged PeptiGel®Alpha4 supported MEC viability, but did not promote formation of polarised acini. Modifying the stiffness of PeptiGel® Alpha4 stimulated changes in MEC viability and changes in protein expression associated with altered MEC function, but did not fully recapitulate the morphologies of MECs grown in Matrigel. To supply the appropriate biochemical signals for MEC organoids, we supplemented PeptiGels® with laminin. Laminin was found to require negatively charged PeptiGel® Alpha7 for functionality, but was then able to provide appropriate signals for correct MEC polarisation and expression of characteristic proteins. Thus, optimisation of SAPH composition and mechanics allows tuning to support tissue-specific organoids.


Subject(s)
Cell Culture Techniques, Three Dimensional , Collagen , Drug Combinations , Epithelial Cells , Hydrogels , Laminin , Peptides , Proteoglycans , Laminin/pharmacology , Laminin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Proteoglycans/pharmacology , Proteoglycans/chemistry , Collagen/chemistry , Collagen/pharmacology , Peptides/pharmacology , Peptides/chemistry , Epithelial Cells/drug effects , Epithelial Cells/cytology , Humans , Female , Cell Culture Techniques, Three Dimensional/methods , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Mammary Glands, Human/cytology , Organoids/drug effects , Organoids/cytology , Cell Culture Techniques/methods
10.
Cancer Sci ; 115(5): 1576-1586, 2024 May.
Article in English | MEDLINE | ID: mdl-38468443

ABSTRACT

While loss of function (LOF) of retinoblastoma 1 (RB1) tumor suppressor is known to drive initiation of small-cell lung cancer and retinoblastoma, RB1 mutation is rarely observed in breast cancers at their initiation. In this study, we investigated the impact on untransformed mammary epithelial cells given by RB1 LOF. Depletion of RB1 in anon-tumorigenic MCF10A cells induced reversible growth arrest (quiescence) featured by downregulation of multiple cyclins and MYC, upregulation of p27KIP1, and lack of expression of markers which indicate cellular senescence or epithelial-mesenchymal transition (EMT). We observed a similar phenomenon in human mammary epithelial cells (HMEC) as well. Additionally, we found that RB1 depletion attenuated the activity of RAS and the downstream MAPK pathway in an RBL2/p130-dependent manner. The expression of farnesyltransferase ß, which is essential for RAS maturation, was found to be downregulated following RB1 depletion also in an RBL2/p130-dependent manner. These findings unveiled an unexpected mechanism whereby normal mammary epithelial cells resist to tumor initiation upon RB1 LOF.


Subject(s)
Down-Regulation , Epithelial Cells , Retinoblastoma Binding Proteins , Signal Transduction , ras Proteins , Humans , Epithelial Cells/metabolism , Female , Retinoblastoma Binding Proteins/metabolism , Retinoblastoma Binding Proteins/genetics , ras Proteins/metabolism , ras Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mammary Glands, Human/cytology , Cell Line, Tumor , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics
11.
J Biol Chem ; 299(7): 104922, 2023 07.
Article in English | MEDLINE | ID: mdl-37321449

ABSTRACT

In normal tissue homeostasis, bidirectional communication between different cell types can shape numerous biological outcomes. Many studies have documented instances of reciprocal communication between fibroblasts and cancer cells that functionally change cancer cell behavior. However, less is known about how these heterotypic interactions shape epithelial cell function in the absence of oncogenic transformation. Furthermore, fibroblasts are prone to undergo senescence, which is typified by an irreversible cell cycle arrest. Senescent fibroblasts are also known to secrete various cytokines into the extracellular space; a phenomenon that is termed the senescence-associated secretory phenotype (SASP). While the role of fibroblast-derived SASP factors on cancer cells has been well studied, the impact of these factors on normal epithelial cells remains poorly understood. We discovered that treatment of normal mammary epithelial cells with conditioned media from senescent fibroblasts (SASP CM) results in a caspase-dependent cell death. This capacity of SASP CM to cause cell death is maintained across multiple senescence-inducing stimuli. However, the activation of oncogenic signaling in mammary epithelial cells mitigates the ability of SASP CM to induce cell death. Despite the reliance of this cell death on caspase activation, we discovered that SASP CM does not cause cell death by the extrinsic or intrinsic apoptotic pathway. Instead, these cells die by an NLRP3, caspase-1, and gasdermin D-dependent induction of pyroptosis. Taken together, our findings reveal that senescent fibroblasts can cause pyroptosis in neighboring mammary epithelial cells, which has implications for therapeutic strategies that perturb the behavior of senescent cells.


Subject(s)
Cellular Senescence , Epithelial Cells , Fibroblasts , Pyroptosis , Caspases/metabolism , Epithelial Cells/cytology , Fibroblasts/metabolism , Mammary Glands, Human/cytology , Humans , Culture Media, Conditioned , Cells, Cultured
12.
Iberoam. j. med ; 5(4): 160-169, 2023. tab, graf
Article in English | IBECS | ID: ibc-226859

ABSTRACT

Introduction: Breast cancer in women is an actual global medical and social problem. The etiology of this disease remains largely unclear. However, it is well known that the incidence of breast cancer increases with age. In the presented work, for the first time, the age dependence of Al, Ca, Cu, Fe, K, Mg, Na, P, S, Si, Sr, and Zn content in the mammary gland of women aged 16-60 years was investigated.Material and methods: For this purpose, a method of inductively coupled plasma atomic emission spectrometry (ICP-AES) was developed, which makes it possible to determine the content of these elements in microsamples (mass from 10 mg) of breast tissue. With the help of the developed technique, the material obtained during the autopsy of 38 practically healthy women aged 16-60 years who died suddenly was studied.Results: Using the parametric Student's t-test and the non-parametric Wilcoxon-Mann-Whitney U-test to compare two age groups (16-40 years and 41-60 years), as well as Pearson's correlation coefficients between age and chemical element content, it was found that the level of K, Mg, Na and S in normal breast tissue decrease with age.Conclusions: The phenomenon of the age-related decrease in the chemical element contents in the normal mammary gland, discovered for the first time, requires further detailed study. (AU)


Introducción: El cáncer de mama en la mujer es un problema médico y social actual a nivel mundial. La etiología de esta enfermedad sigue sin estar clara. Sin embargo, es bien sabido que la incidencia del cáncer de mama aumenta con la edad. En el trabajo presentado se analiza por primera vez la dependencia de la edad del contenido de Al, Ca, Cu, Fe, K, Mg, Na, P, S, Si, Sr y Zn en la glándula mamaria de mujeres de 16 a 60 años. fue investigado.Material y métodos: Para ello se desarrolló un método de espectrometría de emisión atómica con plasma acoplado inductivamente (ICP-AES), que permite determinar el contenido de estos elementos en micromuestras (masa a partir de 10 mg) de tejido mamario. Con la ayuda de la técnica desarrollada se estudió el material obtenido durante la autopsia de 38 mujeres prácticamente sanas de entre 16 y 60 años que murieron repentinamente.Resultados: Utilizando la prueba t de Student paramétrica y la prueba U no paramétrica de Wilcoxon-Mann-Whitney para comparar dos grupos de edad (16-40 años y 41-60 años), así como los coeficientes de correlación de Pearson entre edad y elemento químico. contenido, se encontró que el nivel de K, Mg, Na y S en el tejido mamario normal disminuye con la edad.Conclusiones: El fenómeno de la disminución del contenido de elementos químicos en la glándula mamaria normal relacionado con la edad, descubierto por primera vez, requiere un estudio más detallado. (AU)


Subject(s)
Humans , Female , Adolescent , Young Adult , Adult , Middle Aged , Aged , Mammary Glands, Human/cytology , Spectrophotometry, Atomic/methods , Age Distribution , Simple Random Sampling
13.
Methods Mol Biol ; 2471: 159-183, 2022.
Article in English | MEDLINE | ID: mdl-35175596

ABSTRACT

Lentiviral vectors are the workhorses of modern cell biology. They can infect a wide variety of cells including non-dividing cells and stem cells. They integrate into the genome of infected cells leading to stable expression. It is easy to transduce 100% of the cells in a culture and possible to infect cells simultaneously with multiple vectors, greatly facilitating studies on malignant transformation. We present simple protocols to produce and titrate lentiviral vectors, infect mammary epithelial cells, and check for contamination with replication competent viruses.


Subject(s)
Epithelial Cells , Genetic Vectors , Lentivirus , Animals , Cell Count , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Mammary Glands, Human/cytology , Transduction, Genetic
14.
Nat Cell Biol ; 24(2): 148-154, 2022 02.
Article in English | MEDLINE | ID: mdl-35165416

ABSTRACT

Metabolic characteristics of adult stem cells are distinct from their differentiated progeny, and cellular metabolism is emerging as a potential driver of cell fate conversions1-4. How these metabolic features are established remains unclear. Here we identified inherited metabolism imposed by functionally distinct mitochondrial age-classes as a fate determinant in asymmetric division of epithelial stem-like cells. While chronologically old mitochondria support oxidative respiration, the electron transport chain of new organelles is proteomically immature and they respire less. After cell division, selectively segregated mitochondrial age-classes elicit a metabolic bias in progeny cells, with oxidative energy metabolism promoting differentiation in cells that inherit old mitochondria. Cells that inherit newly synthesized mitochondria with low levels of Rieske iron-sulfur polypeptide 1 have a higher pentose phosphate pathway activity, which promotes de novo purine biosynthesis and redox balance, and is required to maintain stemness during early fate determination after division. Our results demonstrate that fate decisions are susceptible to intrinsic metabolic bias imposed by selectively inherited mitochondria.


Subject(s)
Adult Stem Cells/metabolism , Cell Differentiation , Cell Lineage , DNA, Mitochondrial/genetics , Energy Metabolism , Genes, Mitochondrial , Mammary Glands, Human/metabolism , Mitochondria/metabolism , Animals , Cell Line , Cell Proliferation , Cellular Senescence , Female , Humans , Mammary Glands, Human/cytology , Metabolome , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/genetics , Phenotype , Proteome
15.
J Biol Chem ; 298(3): 101649, 2022 03.
Article in English | MEDLINE | ID: mdl-35104504

ABSTRACT

RNA-binding proteins (RBPs) regulate the expression of large cohorts of RNA species to produce programmatic changes in cellular phenotypes. To describe the function of RBPs within a cell, it is key to identify their mRNA-binding partners. This is often done by crosslinking nucleic acids to RBPs, followed by chemical release of the nucleic acid fragments for analysis. However, this methodology is lengthy, which involves complex processing with attendant sample losses, thus large amounts of starting materials and prone to artifacts. To evaluate potential alternative technologies, we tested "exclusion-based" purification of immunoprecipitates (IFAST or SLIDE) and report here that these methods can efficiently, rapidly, and specifically isolate RBP-RNA complexes. The analysis requires less than 1% of the starting material required for techniques that include crosslinking. Depending on the antibody used, 50% to 100% starting protein can be retrieved, facilitating the assay of endogenous levels of RBPs; the isolated ribonucleoproteins are subsequently analyzed using standard techniques, to provide a comprehensive portrait of RBP complexes. Using exclusion-based techniques, we show that the mRNA-binding partners for RBP IGF2BP1 in cultured mammary epithelial cells are enriched in mRNAs important for detoxifying superoxides (specifically glutathione peroxidase [GPX]-1 and GPX-2) and mRNAs encoding mitochondrial proteins. We show that these interactions are functionally significant, as loss of function of IGF2BP1 leads to destabilization of GPX mRNAs and reduces mitochondrial membrane potential and oxygen consumption. We speculate that this underlies a consistent requirement for IGF2BP1 for the expression of clonogenic activity in vitro.


Subject(s)
Mammary Glands, Animal , Mammary Glands, Human , RNA-Binding Proteins , Animals , Epithelial Cells/metabolism , Female , Humans , Immunoprecipitation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , RNA/metabolism , RNA, Messenger , RNA-Binding Proteins/metabolism
16.
FEBS Lett ; 596(4): 417-426, 2022 02.
Article in English | MEDLINE | ID: mdl-34990021

ABSTRACT

PI3Kß is required for invadopodia-mediated matrix degradation by breast cancer cells. Invadopodia maturation requires GPCR activation of PI3Kß and its coupling to SHIP2 to produce PI(3,4)P2 . We now test whether selectivity for PI3Kß is preserved under conditions of mutational increases in PI3K activity. In breast cancer cells where PI3Kß is inhibited, short-chain diC8-PIP3 rescues gelatin degradation in a SHIP2-dependent manner; rescue by diC8-PI(3,4)P2 is SHIP2-independent. Surprisingly, the expression of either activated PI3Kß or PI3Kα mutants rescued the effects of PI3Kß inhibition. In both cases, gelatin degradation was SHIP2-dependent. These data confirm the requirement for PIP3 conversion to PI(3,4)P2 for invadopodia function and suggest that selectivity for distinct PI3K isotypes may be obviated by mutational activation of the PI3K pathway.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Extracellular Matrix/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Podosomes/metabolism , Cell Line, Tumor , Cell Movement , Class I Phosphatidylinositol 3-Kinases/metabolism , Diglycerides/chemistry , Extracellular Matrix/ultrastructure , Female , Gene Expression Regulation , HEK293 Cells , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mutation , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Podosomes/ultrastructure , Signal Transduction
17.
Nat Commun ; 13(1): 562, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35091553

ABSTRACT

Under normal conditions, the most significant expansion and differentiation of the adult mammary gland occurs in response to systemic reproductive hormones during pregnancy and lactation to enable milk synthesis and secretion to sustain the offspring. However, human mammary tissue remodelling that takes place during pregnancy and lactation remains poorly understood due to the challenge of acquiring samples. We report here single-cell transcriptomic analysis of 110,744 viable breast cells isolated from human milk or non-lactating breast tissue, isolated from nine and seven donors, respectively. We found that human milk largely contains epithelial cells belonging to the luminal lineage and a repertoire of immune cells. Further transcriptomic analysis of the milk cells identified two distinct secretory cell types that shared similarities with luminal progenitors, but no populations comparable to hormone-responsive cells. Taken together, our data offers a reference map and a window into the cellular dynamics that occur during human lactation and may provide further insights on the interplay between pregnancy, lactation and breast cancer.


Subject(s)
Breast/metabolism , Gene Expression Profiling/methods , Lactation/genetics , Mammary Glands, Human/metabolism , Milk, Human/metabolism , Single-Cell Analysis/methods , Breast/cytology , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Cluster Analysis , Female , Gene Regulatory Networks , Humans , Immune System/cytology , Immune System/metabolism , Mammary Glands, Human/cytology , Milk, Human/cytology , Pregnancy , Stromal Cells/cytology , Stromal Cells/metabolism
18.
Sci Rep ; 12(1): 317, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013350

ABSTRACT

This manuscript describes a new method for forming basal-in MCF10A organoids using commercial 384-well ultra-low attachment (ULA) microplates and the development of associated live-cell imaging and automated analysis protocols. The use of a commercial 384-well ULA platform makes this method more broadly accessible than previously reported hanging drop systems and enables in-incubator automated imaging. Therefore, time points can be captured on a more frequent basis to improve tracking of early organoid formation and growth. However, one major challenge of live-cell imaging in multi-well plates is the rapid accumulation of large numbers of images. In this paper, an automated MATLAB script to handle the increased image load is developed. This analysis protocol utilizes morphological image processing to identify cellular structures within each image and quantify their circularity and size. Using this script, time-lapse images of aggregating and non-aggregating culture conditions are analyzed to profile early changes in size and circularity. Moreover, this high-throughput platform is applied to widely screen concentration combinations of Matrigel and epidermal growth factor (EGF) or heparin-binding EGF-like growth factor (HB-EGF) for their impact on organoid formation. These results can serve as a practical resource, guiding future research with basal-in MCF10A organoids.


Subject(s)
Cell Culture Techniques, Three Dimensional/instrumentation , Cell Proliferation , High-Throughput Screening Assays , Image Processing, Computer-Assisted , Mammary Glands, Human/physiology , Microscopy, Fluorescence , Organoids , Time-Lapse Imaging , Algorithms , Cell Line , Cell Proliferation/drug effects , Collagen/pharmacology , Drug Combinations , Epidermal Growth Factor/pharmacology , Female , Heparin-binding EGF-like Growth Factor/pharmacology , Humans , Laminin/pharmacology , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Proteoglycans/pharmacology , Time Factors
19.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119159, 2022 01.
Article in English | MEDLINE | ID: mdl-34653580

ABSTRACT

Nearly all mammals rely on lactation to support their young and to ensure the continued survival of their species. Despite its importance, relatively little is known about how milk is produced and how it is ejected from the lumen of mammary alveoli and ducts. This review focuses on the latter. We discuss how a relatively small number of basal cells, wrapping around each alveolar unit, contract to forcibly expel milk from the alveolar lumen. We consider how individual basal cells coordinate their activity, the fate of these cells at the end of lactation and avenues for future deliberation and exploration.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/metabolism , Animals , Cell Plasticity , Epithelial Cells/cytology , Female , Humans , Lactation , Mammary Glands, Human/cytology , Mammary Glands, Human/physiology
20.
Cell Rep ; 37(13): 110181, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965432

ABSTRACT

The EGFR/Erk pathway is triggered by extracellular ligand stimulation, leading to stimulus-dependent dynamics of pathway activity. Although mechanical properties of the microenvironment also affect Erk activity, their effects on Erk signaling dynamics are poorly understood. Here, we characterize how the stiffness of the underlying substratum affects Erk signaling dynamics in mammary epithelial cells. We find that soft microenvironments attenuate Erk signaling, both at steady state and in response to epidermal growth factor (EGF) stimulation. Optogenetic manipulation at multiple signaling nodes reveals that intracellular signal transmission is largely unaffected by substratum stiffness. Instead, we find that soft microenvironments decrease EGF receptor (EGFR) expression and alter the amount and spatial distribution of EGF binding at cell membranes. Our data demonstrate that the mechanical microenvironment tunes Erk signaling dynamics via receptor-ligand interactions, underscoring how multiple microenvironmental signals are jointly processed through a highly conserved pathway that regulates tissue development, homeostasis, and disease progression.


Subject(s)
Cellular Microenvironment , Extracellular Matrix/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Mammary Glands, Human/metabolism , Cell Movement , Cells, Cultured , ErbB Receptors/metabolism , Female , Humans , Mammary Glands, Human/cytology , Phosphorylation , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL