Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(21)2022 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-36358627

RESUMO

The human breast gland is a unique organ as most of its development occurs postnatally between menarche and menopause, a period ranging from 30 to 40 years. During this period, the monthly menstruation cycle drives the mammary gland through phases of cell proliferation, differentiation, and apoptosis, facilitated via a closely choreographed interaction between the epithelial cells and the surrounding stroma preparing the gland for pregnancy. If pregnancy occurs, maximal differentiation is reached to prepare for lactation. After lactation, the mammary gland involutes to a pre-pregnant state. These cycles of proliferation, differentiation, and involution necessitate the presence of epithelial stem cells that give rise to progenitor cells which differentiate further into the luminal and myoepithelial lineages that constitute the epithelial compartment and are responsible for the branching structure of the gland. Maintaining homeostasis and the stem cell niche depends strongly on signaling between the stem and progenitor cells and the surrounding stroma. Breast cancer is a slowly progressing disease whose initiation can take decades to progress into an invasive form. Accumulating evidence indicates that stem cells and/or progenitor cells at different stages, rather than terminally differentiated cells are the main cells of origin for most breast cancer subgroups. Stem cells and cancer cells share several similarities such as increased survival and cellular plasticity which is reflected in their ability to switch fate by receiving intrinsic and extrinsic signals. In this review, we discuss the concept of cellular plasticity in normal breast morphogenesis and cancer, and how the stromal environment plays a vital role in cancer initiation and progression.

2.
Methods Mol Biol ; 2429: 391-403, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35507176

RESUMO

Capturing breast morphogenesis and cancer progression in 3D culture using cell lines with stem cell properties can greatly increase understanding of the underlying mechanisms involved in these processes, highlighting the importance of the culture method. D492 is a breast epithelial progenitor cell line that provides a model for branching morphogenesis when cultured in 3D reconstituted basement membrane matrix (rBM). Along with its derivate cell lines D492M and D492HER2, D492 also serves as a robust model for epithelial to mesenchymal transition (EMT) and tumorigenicity, respectively. Here, we describe the routine maintenance and application of the D492 cell lines in 3D culture for the study of branching morphogenesis, EMT and epithelial-endothelial interaction.


Assuntos
Células Epiteliais , Transição Epitelial-Mesenquimal , Comunicação Celular , Morfogênese , Células-Tronco/metabolismo
3.
Methods Mol Biol ; 2471: 283-299, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35175604

RESUMO

Epithelial-stromal interactions play an essential role in regulation of mammary gland development, homeostasis, and tumorigenesis. Fibroblasts constitute a substantial proportion of mammary gland stromal cells in human breast and have been recognized for their paracrine signaling and extracellular matrix production and remodeling roles during normal breast development as well as in breast cancer. However, our current knowledge on human breast fibroblast functions is incomplete. Here we provide a detailed protocol for an organotypic human breast assay to facilitate research in the roles of human breast fibroblasts in mammary epithelial morphogenesis and early tumorigenesis.


Assuntos
Neoplasias da Mama , Glândulas Mamárias Humanas , Animais , Mama , Células Epiteliais , Feminino , Fibroblastos , Humanos , Glândulas Mamárias Animais , Comunicação Parácrina , Células Estromais
4.
J Mammary Gland Biol Neoplasia ; 26(4): 321-338, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34964086

RESUMO

The human breast is composed of terminal duct lobular units (TDLUs) that are surrounded by stroma. In the TDLUs, basement membrane separates the stroma from the epithelial compartment, which is divided into an inner layer of luminal epithelial cells and an outer layer of myoepithelial cells. Stem cells and progenitor cells also reside within the epithelium and drive a continuous cycle of gland remodelling that occurs throughout the reproductive period. D492 is an epithelial cell line originally isolated from the stem cell population of the breast and generates both luminal and myoepithelial cells in culture. When D492 cells are embedded into 3D reconstituted basement membrane matrix (3D-rBM) they form branching colonies mimicking the TDLUs of the breast, thereby providing a well-suited in vitro model for studies on branching morphogenesis and breast development. Peroxidasin (PXDN) is a heme-containing peroxidase that crosslinks collagen IV with the formation of sulfilimine bonds. Previous studies indicate that PXDN plays an integral role in basement membrane stabilisation by crosslinking collagen IV and as such contributes to epithelial integrity. Although PXDN has been linked to fibrosis and cancer in some organs there is limited information on its role in development, including in the breast. In this study, we demonstrate expression of PXDN in breast epithelium and stroma and apply the D492 cell line to investigate the role of PXDN in cell differentiation and branching morphogenesis in the human breast. Overexpression of PXDN induced basal phenotype in D492 cells, loss of plasticity and inhibition of epithelial-to-mesenchymal transition as is displayed by complete inhibition of branching morphogenesis in 3D culture. This is supported by results from RNA-sequencing which show significant enrichment in genes involved in epithelial differentiation along with significant negative enrichment of EMT factors. Taken together, we provide evidence for a novel role of PXDN in breast epithelial differentiation and mammary gland development.


Assuntos
Células Epiteliais , Células-Tronco , Colágeno/metabolismo , Células Epiteliais/metabolismo , Proteínas da Matriz Extracelular , Humanos , Morfogênese/fisiologia , Peroxidase , Fenótipo , Peroxidasina
5.
Org Biomol Chem ; 19(47): 10417-10423, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34817496

RESUMO

Herein we disclose the transformation of maleimides into water-soluble tris(2-carboxyethyl)phosphonium ylides and their subsequent application in the bioconjugation of protein- and peptide-linked aldehydes. The new entry into Wittig bioconjugate chemistry proceeds under mild conditions and relies on highly water soluble reagents, which are likely already part of most biochemists' inventory.


Assuntos
Maleimidas
6.
J Mammary Gland Biol Neoplasia ; 26(3): 221-226, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34448098

RESUMO

The twelfth annual workshop of the European Network for Breast Development and Cancer focused on methods in mammary gland biology and breast cancer, was scheduled to take place on March 26-28, 2020, in Weggis, Switzerland. Due to the COVID-19 pandemic, the meeting was rescheduled twice and eventually happened as a virtual meeting on April 22 and 23, 2021. The main topics of the meeting were branching and development of the mammary gland, tumor microenvironment, circulating tumor cells, tumor dormancy and breast cancer metastasis. Novel and unpublished findings related to these topics were presented, with a particular focus on the methods used to obtain them. Virtual poster sessions were a success, with many constructive and fruitful interactions between researchers and covered many areas of mammary gland biology and breast cancer.


Assuntos
Pesquisa Biomédica/métodos , Neoplasias da Mama/patologia , Glândulas Mamárias Humanas/patologia , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Terapia Combinada , Europa (Continente) , Feminino , Humanos , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Metástase Neoplásica , Estadiamento de Neoplasias , Células Neoplásicas Circulantes , Prognóstico , Microambiente Tumoral
7.
Cancers (Basel) ; 13(7)2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810334

RESUMO

Multiple myeloma (MM) is characterized by extensive immunoglobulin production leading to an excessive load on protein homeostasis in tumor cells. Aminopeptidases contribute to proteolysis by catalyzing the hydrolysis of amino acids from proteins or peptides and function downstream of the ubiquitin-proteasome pathway. Notably, aminopeptidases can be utilized in the delivery of antibody and peptide-conjugated drugs, such as melflufen, currently in clinical trials. We analyzed the expression of 39 aminopeptidase genes in MM samples from 122 patients treated at Finnish cancer centers and 892 patients from the CoMMpass database. Based on ranked abundance, LAP3, ERAP2, METAP2, TTP2, and DPP7 were highly expressed in MM. ERAP2, XPNPEP1, DPP3, RNPEP, and CTSV were differentially expressed between relapsed/refractory and newly diagnosed MM samples (p < 0.05). Sensitivity to melflufen was detected ex vivo in 11/15 MM patient samples, and high sensitivity was observed, especially in relapsed/refractory samples. Survival analysis revealed that high expression of XPNPEP1, RNPEP, DPP3, and BLMH (p < 0.05) was associated with shorter overall survival. Hydrolysis analysis demonstrated that melflufen is a substrate for aminopeptidases LAP3, LTA4H, RNPEP, and ANPEP. The sensitivity of MM cell lines to melflufen was reduced by aminopeptidase inhibitors. These results indicate critical roles of aminopeptidases in disease progression and the activity of melflufen in MM.

9.
J Mammary Gland Biol Neoplasia ; 25(4): 273-288, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33210256

RESUMO

3D cell culture methods have been an integral part of and an essential tool for mammary gland and breast cancer research for half a century. In fact, mammary gland researchers, who discovered and deciphered the instructive role of extracellular matrix (ECM) in mammary epithelial cell functional differentiation and morphogenesis, were the pioneers of the 3D cell culture techniques, including organoid cultures. The last decade has brought a tremendous increase in the 3D cell culture techniques, including modifications and innovations of the existing techniques, novel biomaterials and matrices, new technological approaches, and increase in 3D culture complexity, accompanied by several redefinitions of the terms "3D cell culture" and "organoid". In this review, we provide an overview of the 3D cell culture and organoid techniques used in mammary gland biology and breast cancer research. We discuss their advantages, shortcomings and current challenges, highlight the recent progress in reconstructing the complex mammary gland microenvironment in vitro and ex vivo, and identify the missing 3D cell cultures, urgently needed to aid our understanding of mammary gland development, function, physiology, and disease, including breast cancer.


Assuntos
Neoplasias da Mama/patologia , Técnicas de Cultura de Células/instrumentação , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/patologia , Esferoides Celulares/patologia , Animais , Diferenciação Celular , Técnicas de Cocultura/métodos , Células Epiteliais/patologia , Matriz Extracelular/patologia , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Humanas/citologia , Camundongos , Modelos Animais , Organoides
10.
Front Cell Dev Biol ; 8: 461, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32612992

RESUMO

Epithelial-to-mesenchymal transition (EMT) and its reversed process mesenchymal-to-epithelial transition (MET) play a critical role in epithelial plasticity during development and cancer progression. Among important regulators of these cellular processes are non-coding RNAs (ncRNAs). The imprinted DLK1-DIO3 locus, containing numerous maternally expressed ncRNAs including the lncRNA maternally expressed gene 3 (MEG3) and a cluster of over 50 miRNAs, has been shown to be a modulator of stemness in embryonic stem cells and in cancer progression, potentially through the tumor suppressor role of MEG3. In this study we analyzed the expression pattern and functional role of ncRNAs from the DLK1-DIO3 locus in epithelial plasticity of the breast. We studied their expression in various cell types of breast tissue and revisit the role of the locus in EMT/MET using a breast epithelial progenitor cell line (D492) and its isogenic mesenchymal derivative (D492M). Marked upregulation of ncRNAs from the DLK1-DIO3 locus was seen after EMT induction in two cell line models of EMT. In addition, the expression of MEG3 and the maternally expressed ncRNAs was higher in stromal cells compared to epithelial cell types in primary breast tissue. We also show that expression of MEG3 is concomitant with the expression of the ncRNAs from the DLK1-DIO3 locus and its expression is therefore likely indicative of activation of all ncRNAs at the locus. MEG3 expression is correlated with stromal markers in normal tissue and breast cancer tissue and negatively correlated with the survival of breast cancer patients in two different cohorts. Overexpression of MEG3 using CRISPR activation in a breast epithelial cell line induced partial EMT and enriched for a basal-like phenotype. Conversely, knock down of MEG3 using CRISPR inhibition in a mesenchymal cell line reduced the mesenchymal and basal-like phenotype of the cell line. In summary our study shows that maternally expressed ncRNAs are markers of EMT and suggests that MEG3 is a novel regulator of EMT/MET in breast tissue. Nevertheless, further studies are needed to fully dissect the molecular pathways influenced by non-coding RNAs at the DLK1-DIO3 locus in breast tissue.

11.
Cancer Med ; 9(18): 6726-6738, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32717133

RESUMO

Melphalan flufenamide (hereinafter referred to as "melflufen") is a peptide-conjugated drug currently in phase 3 trials for the treatment of relapsed or refractory multiple myeloma. Due to its lipophilic nature, it readily enters cells, where it is converted to the known alkylator melphalan leading to enrichment of hydrophilic alkylator payloads. Here, we have analysed in vitro and in vivo the efficacy of melflufen on normal and cancerous breast epithelial lines. D492 is a normal-derived nontumorigenic epithelial progenitor cell line whereas D492HER2 is a tumorigenic version of D492, overexpressing the HER2 oncogene. In addition we used triple negative breast cancer cell line MDA-MB231. The tumorigenic D492HER2 and MDA-MB231 cells were more sensitive than normal-derived D492 cells when treated with melflufen. Compared to the commonly used anti-cancer drug doxorubicin, melflufen was significantly more effective in reducing cell viability in vitro while it showed comparable effects in vivo. However, melflufen was more efficient in inhibiting metastasis of MDA-MB231 cells. Melflufen induced DNA damage was confirmed by the expression of the DNA damage proteins Æ´H2Ax and 53BP1. The effect of melflufen on D492HER2 was attenuated if cells were pretreated with the aminopeptidase inhibitor bestatin, which is consistent with previous reports demonstrating the importance of aminopeptidase CD13 in facilitating melflufen cleavage. Moreover, analysis of CD13high and CD13low subpopulations of D492HER2 cells and knockdown of CD13 showed that melflufen efficacy is mediated at least in part by CD13. Knockdown of LAP3 and DPP7 aminopeptidases led to similar efficacy reduction, suggesting that also other aminopeptidases may facilitate melflufen conversion. In summary, we have shown that melflufen is a highly efficient anti-neoplastic agent in breast cancer cell lines and its efficacy is facilitated by aminopeptidases.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias da Mama/tratamento farmacológico , Melfalan/análogos & derivados , Fenilalanina/análogos & derivados , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígenos CD13/genética , Antígenos CD13/metabolismo , Linhagem Celular Tumoral , Embrião de Galinha , Dano ao DNA , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Humanos , Leucil Aminopeptidase/genética , Leucil Aminopeptidase/metabolismo , Melfalan/farmacologia , Fenilalanina/farmacologia , Transdução de Sinais , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
12.
Oncogene ; 39(28): 5138-5151, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32533097

RESUMO

Waldenström's macroglobulinemia (WM) is a non-Hodgkin lymphoma, resulting in antibody-secreting lymphoplasmacytic cells in the bone marrow and pathologies resulting from high levels of monoclonal immunoglobulin M (IgM) in the blood. Despite the key role for BLIMP1 in plasma cell maturation and antibody secretion, its potential effect on WM cell biology has not yet been explored. Here we provide evidence of a crucial role for BLIMP1 in the survival of cells from WM cell line models and further demonstrate that BLIMP1 is necessary for the expression of the histone methyltransferase EZH2 in both WM and multiple myeloma cell lines. The effect of BLIMP1 on EZH2 levels is post-translational, at least partially through the regulation of proteasomal targeting of EZH2. Chromatin immunoprecipitation analysis and transcriptome profiling suggest that the two factors co-operate in regulating genes involved in cancer cell immune evasion. Co-cultures of natural killer cells and cells from a WM cell line further suggest that both factors participate in immune evasion by promoting escape from natural killer cell-mediated cytotoxicity. Together, the interplay of BLIMP1 and EZH2 plays a vital role in promoting the survival of WM cell lines, suggesting a role for the two factors in Waldenström's macroglobulinaemia.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Linfoma não Hodgkin/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Células HEK293 , Humanos , Linfoma não Hodgkin/metabolismo , Linfoma não Hodgkin/patologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Ligação Proteica , Macroglobulinemia de Waldenstrom/genética , Macroglobulinemia de Waldenstrom/metabolismo , Macroglobulinemia de Waldenstrom/patologia
13.
Lab Invest ; 100(7): 928-944, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32203150

RESUMO

The tumor microenvironment is increasingly recognized as key player in cancer progression. Investigating heterotypic interactions between cancer cells and their microenvironment is important for understanding how specific cell types support cancer. Forming the vasculature, endothelial cells (ECs) are a prominent cell type in the microenvironment of both normal and neoplastic breast gland. Here, we sought out to analyze epithelial-endothelial cross talk in the breast using isogenic non-tumorigenic vs. tumorigenic breast epithelial cell lines and primary ECs. The cellular model used here consists of D492, a breast epithelial cell line with stem cell properties, and two isogenic D492-derived EMT cell lines, D492M and D492HER2. D492M was generated by endothelial-induced EMT and is non-tumorigenic while D492HER2 is tumorigenic, expressing the ErbB2/HER2 oncogene. To investigate cellular cross talk, we used both conditioned medium (CM) and 2D/3D co-culture systems. Secretome analysis of D492 cell lines was performed using mass spectrometry and candidate knockdown (KD), and overexpression (OE) was done using siRNA and CRISPRi/CRISPRa technology. D492HER2 directly enhances endothelial network formation and activates a molecular axis in ECs promoting D492HER2 migration and invasion, suggesting an endothelial feedback response. Secretome analysis identified extracellular matrix protein 1 (ECM1) as potential angiogenic inducer in D492HER2. Confirming its involvement, KD of ECM1 reduced the ability of D492HER2-CM to increase endothelial network formation and induce the endothelial feedback, while recombinant ECM1 (rECM1) increased both. Interestingly, NOTCH1 and NOTCH3 expression was upregulated in ECs upon treatment with D492HER2-CM or rECM1 but not by CM from D492HER2 with ECM1 KD. Blocking endothelial NOTCH signaling inhibited the increase in network formation and the ability of ECs to promote D492HER2 migration and invasion. In summary, our data demonstrate that cancer-secreted ECM1 induces a NOTCH-mediated endothelial feedback promoting cancer progression by enhancing migration and invasion. Targeting this interaction may provide a novel possibility to improve cancer treatment.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Invasividade Neoplásica/genética , Receptor ErbB-2/metabolismo , Microambiente Tumoral/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proteínas da Matriz Extracelular/genética , Feminino , Humanos , Receptor ErbB-2/genética
14.
J Mammary Gland Biol Neoplasia ; 25(4): 233-236, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33479879

RESUMO

The field of mammary gland biology and breast cancer research encompasses a wide range of topics and scientific questions, which span domains of molecular, cell and developmental biology, cancer research, and veterinary and human medicine, with interdisciplinary overlaps to non-biological domains. Accordingly, mammary gland and breast cancer researchers employ a wide range of molecular biology methods, in vitro techniques, in vivo approaches as well as in silico analyses. The list of techniques is ever-expanding; together with the refinement of established, staple techniques in the field, new technologies keep emerging thanks to technological advances and scientific creativity. This issue of the Journal of Mammary Gland Biology and Neoplasia represents a compilation of original articles and reviews focused on methods used in mammary gland biology and breast cancer research.


Assuntos
Pesquisa Biomédica/métodos , Neoplasias da Mama/patologia , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/patologia , Neoplasias Mamárias Animais/patologia , Animais , Feminino , Humanos , Lactação/fisiologia , Glândulas Mamárias Animais/fisiologia , Glândulas Mamárias Humanas/fisiologia , Gravidez
15.
In Vitro Cell Dev Biol Anim ; 55(10): 838-853, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31482369

RESUMO

Epithelial to mesenchymal transition (EMT) is a developmental event that is hijacked in some diseases such as fibrosis and cancer. In cancer, EMT has been linked to increased invasion and metastasis and is generally associated with a poor prognosis. In this study, we have compared phenotypic and functional differences between two isogenic cell lines with an EMT profile: D492M and D492HER2 that are both derived from D492, a breast epithelial cell line with stem cell properties. D492M is non-tumorigenic while D492HER2 is tumorigenic. Thus, the aim of this study was to analyze the expression profile of these cell lines, identify potential oncogenes, and evaluate their effects on cellular phenotype. We performed transcriptome and secretome analyses of D492M and D492HER2 and verified expression of selected genes at the RNA and protein level. One candidate, YKL-40 (also known as CHI3L1), was selected for further studies due to its differential expression between D492M and D492HER2, being considerably higher in D492HER2. YKL-40 has been linked to chronic inflammation diseases and cancer, yet its function is not fully understood. Knock-down experiments of YKL-40 in D492HER2 resulted in reduced migration and invasion as well as reduced ability to induce angiogenesis in an in vitro assay, plus changes in the EMT-phenotype. In summary, our data suggest that YKL-40 may provide D492HER2 with increased aggressiveness, supporting cancer progression and facilitating angiogenesis.


Assuntos
Proteína 1 Semelhante à Quitinase-3/metabolismo , Glândulas Mamárias Humanas/citologia , Neovascularização Patológica/metabolismo , Técnicas de Cultura de Células , Movimento Celular , Proliferação de Células , Proteína 1 Semelhante à Quitinase-3/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Receptor ErbB-2/metabolismo , Células-Tronco/metabolismo
16.
Cytokine Growth Factor Rev ; 46: 17-27, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30930082

RESUMO

Delta like non-canonical Notch ligand 1 (Dlk1) is an imprinted gene, mainly known for its involvement in adipogenesis, although it has been associated with many other stem cells/progenitors and is known to be widely expressed during organism development and tissue regeneration. In a systematic manner, we have outlined the overall expression pattern of Dlk1 in both man and mouse, and found Dlk1 to be expressed in tissues from all three germ layers. Yet, Dlk1 expression decreases along with increased differentiation as gestation proceeds and in most tissues Dlk1 is absent around birth. Thus, in adults, expression of Dlk1 is restricted to a few tissues and progenitor cells, but is re-expressed during disease and regeneration. Although diffferences exist, we found an overall conservation of Dlk1 expression between mouse and man, and conclude in that sense that the mouse is an appropiate model to study Dlk1. In agreement with the observed Dlk1 expression pattern, we found that the majority of published Dlk1 studies, report Dlk1 to have an inhibitory effect on both cell proliferation and differentiation, but the levels of the different DLK1 isoforms may be critical and have an impact on the overall outcome. This may also be an issue during tissue regeneration where several studies have reported Dlk1's impact during skeletal muscle and liver regeneration without establishing the exact role. Likewise, the underlying mechanism of Dlk1 action is unknown, and seems to depend on both Notch dependent and independent pathways. However, from our data it is intriguing to speculate that the actual role of DLK1 may be to function as a checkpoint to slow down proliferation while forcing cells into the process of differentiation, and thus switch the cell/organ to a state of growth and hypertrophy. This may fit well with its reported impact on growth restiction and body size. Thus, our study which for the first time summarizes reported knowledge on Dlk1 in tissue development and regeneration as well as on the Dlk1 mechanism may provide novel insight to the general role of this remarkable imprinted gene in controlling cell growth, from which new hypotheses can be made in the field of stem cell biology and regenerative medicine.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Mamíferos/crescimento & desenvolvimento , Proteínas de Membrana/genética , Receptores Notch/metabolismo , Regeneração , Transdução de Sinais , Animais , Diferenciação Celular , Proliferação de Células , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Transporte Proteico , Receptores Notch/genética
17.
J Mammary Gland Biol Neoplasia ; 24(2): 139-147, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30684066

RESUMO

The human female breast gland is composed of branching epithelial ducts that extend from the nipple towards the terminal duct lobular units (TDLUs), which are the functional, milk-producing units of the gland and the site of origin of most breast cancers. The epithelium of ducts and TDLUs is composed of an inner layer of polarized luminal epithelial cells and an outer layer of contractile myoepithelial cells, separated from the vascular-rich stroma by a basement membrane. The luminal- and myoepithelial cells share an origin and in recent years, there has been increasing understanding of how these cell types interact and how they contribute to breast cancer. Accumulating evidence links stem/or progenitor cells in the mammary/breast gland to breast cancer. In that regard, much knowledge has been gained from studies in mice due to specific strains that have allowed for gene knock out/in studies and lineage tracing of cellular fates. However, there is a large histologic difference between the human female breast gland and the mouse mammary gland that necessitates that research needs to be done on human material where primary cultures are important due to their close relation to the tissue of origin. However, due to difficulties of long-term cultures and lack of access to material, human cell lines are of great importance to bridge the gap between studies on mouse mammary gland and human primary breast cells. In this review, we describe D492, a breast epithelial progenitor cell line that can generate both luminal- and myoepithelial cells in culture, and in 3D culture it forms branching ducts similar to TDLUs. We have applied D492 and its daughter cell lines to explore cellular and molecular mechanisms of branching morphogenesis and cellular plasticity including EMT and MET. In addition to discussing the application of D492 in studying normal morphogenesis, we will also discuss how this cell line has been used to study breast cancer progression.


Assuntos
Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Células Epiteliais/fisiologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Células-Tronco/fisiologia , Técnicas de Cultura de Células/métodos , Linhagem Celular , Plasticidade Celular , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/patologia , MicroRNAs/metabolismo , Morfogênese/fisiologia , Receptor ErbB-2/metabolismo
18.
Mech Dev ; 155: 34-47, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30508578

RESUMO

MicroRNAs regulate developmental events such as branching morphogenesis, epithelial to mesenchymal transition (EMT) and its reverse process mesenchymal to epithelial transition (MET). In this study, we performed small RNA sequencing of a breast epithelial progenitor cell line (D492), and its mesenchymal derivative (D492M) cultured in three-dimensional microenvironment. Among the most downregulated miRNAs in D492M was miR-203a, a miRNA that plays an important role in epithelial differentiation. Increased expression of miR-203a was seen in D492, concomitant with increased complexity of branching. When miR-203a was overexpressed in D492M, a partial reversion towards epithelial phenotype was seen. Gene expression analysis of D492M and D492MmiR-203a revealed peroxidasin, a collagen IV cross-linker, as the most significantly downregulated gene in D492MmiR-203a. Collectively, we demonstrate that miR-203a expression temporally correlates with branching morphogenesis and is suppressed in D492M. Overexpression of miR-203a in D492M induces a partial MET and reduces the expression of peroxidasin. Furthermore, we demonstrate that miR-203a is a novel repressor of peroxidasin. MiR-203-peroxidasin axis may be an important regulator in branching morphogenesis, EMT/MET and basement membrane remodeling.


Assuntos
Mama/fisiologia , Transição Epitelial-Mesenquimal/genética , Proteínas da Matriz Extracelular/genética , MicroRNAs/genética , Peroxidase/genética , Células-Tronco/fisiologia , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Regulação para Baixo/genética , Células Epiteliais/fisiologia , Feminino , Expressão Gênica/fisiologia , Células HEK293 , Humanos , Morfogênese/fisiologia , Transdução de Sinais/genética , Peroxidasina
19.
Int J Biol Macromol ; 97: 460-467, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28099888

RESUMO

Delta-like 1 homolog (DLK1) is an imprinted gene, which is widely expressed during mammalian development and plays a pivotal role in differentiation of various tissue types. Most recently, we have shown that DLK1 interacts with NOTCH1, yet several Notch independent mechanisms have previously been suggested as well, but only poorly confirmed in a mammalian context. In the present study, we employed the mammalian two-hybrid (MTH) system, a genetic in vivo protein-protein interaction system, to show robust DLK1-DLK1, DLK1-FnI (Fibronectin) and DLK1-CFR (cysteine-rich FGF receptor) interactions, whereas the proposed DLK1-IGFBP1 interaction was not supported by MTH. Very little has previously been described on the DLK1 self-interaction. Herein, we showed by immunoprecipitation as well as Sulfo-SBED label transfer that the DLK1-DLK1 interaction likely is part of Dlk1's function in preadipocytes. Furthermore our data suggest that DLK1 interacts with itself through EGF domain 4 and 5, which is distinct from the recently described NOTCH1-DLK1 interaction, which occurs between EGF domain 5 and 6. This opens up the possibility that Notch independent mechanisms like the DLK1-DLK1 interaction may modulate the non-canonical NOTCH1-DLK1 interaction further complexing this system.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Agregados Proteicos , Receptor Notch1/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Fibronectinas/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Ligantes , Proteínas de Membrana/química , Camundongos , Células NIH 3T3 , Ligação Proteica , Domínios Proteicos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
20.
Int J Cardiol ; 222: 448-456, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27505332

RESUMO

BACKGROUND/OBJECTIVES: Epicardium-derived progenitor cells (EPDCs) differentiate into all heart cell types in the embryonic heart, yet their differentiation into cardiomyocytes in the adult heart is limited and poorly described. This may be due to EPDCs lacking myogenic potential or the inert adult heart missing regenerative signals essential for directed differentiation of EPDCs. Herein, we aimed to evaluate the myogenic potential of neonatal EPDCs in adult and neonatal mouse myocardium, as well as in skeletal muscle. The two latter tissues have an intrinsic capability to develop and regenerate, in contrast to the adult heart. METHODS: Highly purified mouse EPDCs were transplanted into damaged neonatal and adult myocardium as well as regenerating skeletal muscle. Co-cultures with skeletal myoblasts were used to distinguish fusion independent myogenic conversion. RESULTS: No donor EPDC-derived cardiomyocytes were observed in hearts. In contrast, a remarkable contribution of EPDCs to skeletal muscle myofiber formation was evident in vivo. Furthermore, co-cultures of EPDCs with myoblasts showed that EPDCs became part of multinucleated fibers and appeared to acquire myogenic traits independent of a fusion event. Fluorescence activated cell sorting of EPDCs co-cultured with and without myoblasts and subsequent qRT-PCR of 64 transcripts established that the myogenic phenotype conversion was accomplished through induction of a transcriptional myogenic program. CONCLUSION: These results suggest that EPDCs may be more myogenic than previously anticipated. But, the heart may lack factors for induction of myogenesis of EPDCs, a scenario that should be taken into consideration when aiming for repair of damaged myocardium by stem cell transplantation.


Assuntos
Músculo Esquelético/citologia , Miocárdio/citologia , Miócitos Cardíacos , Pericárdio/citologia , Células-Tronco , Animais , Animais Recém-Nascidos , Células Cultivadas , Técnicas de Cocultura/métodos , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/fisiologia , Miócitos Cardíacos/fisiologia , Pericárdio/fisiologia , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA