Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 584(7822): 608-613, 2020 08.
Article in English | MEDLINE | ID: mdl-32848220

ABSTRACT

Glandular epithelia, including the mammary and prostate glands, are composed of basal cells (BCs) and luminal cells (LCs)1,2. Many glandular epithelia develop from multipotent basal stem cells (BSCs) that are replaced in adult life by distinct pools of unipotent stem cells1,3-8. However, adult unipotent BSCs can reactivate multipotency under regenerative conditions and upon oncogene expression3,9-13. This suggests that an active mechanism restricts BSC multipotency under normal physiological conditions, although the nature of this mechanism is unknown. Here we show that the ablation of LCs reactivates the multipotency of BSCs from multiple epithelia both in vivo in mice and in vitro in organoids. Bulk and single-cell RNA sequencing revealed that, after LC ablation, BSCs activate a hybrid basal and luminal cell differentiation program before giving rise to LCs-reminiscent of the genetic program that regulates multipotency during embryonic development7. By predicting ligand-receptor pairs from single-cell data14, we find that TNF-which is secreted by LCs-restricts BC multipotency under normal physiological conditions. By contrast, the Notch, Wnt and EGFR pathways were activated in BSCs and their progeny after LC ablation; blocking these pathways, or stimulating the TNF pathway, inhibited regeneration-induced BC multipotency. Our study demonstrates that heterotypic communication between LCs and BCs is essential to maintain lineage fidelity in glandular epithelial stem cells.


Subject(s)
Cell Communication , Epithelial Cells/cytology , Multipotent Stem Cells/cytology , Animals , Cell Lineage , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Female , Homeostasis , Humans , Male , Mammary Glands, Animal/cytology , Mice , Multipotent Stem Cells/metabolism , Organoids/cytology , Prostate/cytology , RNA, Messenger/genetics , RNA-Seq , Receptors, Notch/metabolism , Salivary Glands/cytology , Single-Cell Analysis , Skin/cytology , Tumor Necrosis Factor-alpha/metabolism , Wnt Proteins/metabolism
2.
Nature ; 556(7702): 463-468, 2018 04.
Article in English | MEDLINE | ID: mdl-29670281

ABSTRACT

In cancer, the epithelial-to-mesenchymal transition (EMT) is associated with tumour stemness, metastasis and resistance to therapy. It has recently been proposed that, rather than being a binary process, EMT occurs through distinct intermediate states. However, there is no direct in vivo evidence for this idea. Here we screen a large panel of cell surface markers in skin and mammary primary tumours, and identify the existence of multiple tumour subpopulations associated with different EMT stages: from epithelial to completely mesenchymal states, passing through intermediate hybrid states. Although all EMT subpopulations presented similar tumour-propagating cell capacity, they displayed differences in cellular plasticity, invasiveness and metastatic potential. Their transcriptional and epigenetic landscapes identify the underlying gene regulatory networks, transcription factors and signalling pathways that control these different EMT transition states. Finally, these tumour subpopulations are localized in different niches that differentially regulate EMT transition states.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms/pathology , Animals , Chromatin/genetics , Epigenesis, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Male , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mesoderm/metabolism , Mesoderm/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasms/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription, Genetic
3.
4.
Breast Cancer Res ; 17: 121, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26328589

ABSTRACT

Bisphosphonates (BPs) are approved as standard therapy in breast cancer for the treatment of bone metastases, since they were demonstrated to reduce the prevalence of skeletal-related events including fractures and hypercalcemia. In the adjuvant setting, BPs can be given to prevent and treat tumor therapy-induced bone loss in premenopausal and postmenopausal women and, owing to their beneficial effect on bone turnover, have also been evaluated for prevention of bone metastases occurrence. In this article we will review the mechanisms through which BPs have been demonstrated to prevent premetastatic niche formation and cell proliferation in bone lesions. Moreover, preclinical evidence of antitumoral effects of BPs will be presented and results from the most important clinical trials will be described critically. BPs may clearly play a clinically important role in early breast cancer in a postmenopausal adjuvant setting.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Breast Neoplasms/drug therapy , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Animals , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans
5.
Expert Opin Emerg Drugs ; 20(4): 637-51, 2015.
Article in English | MEDLINE | ID: mdl-26113304

ABSTRACT

INTRODUCTION: Bone metastases are virtually incurable resulting in significant disease morbidity, reduced quality of life and mortality. Bone provides a unique microenvironment whose local interactions with tumor cells offer novel targets for therapeutic interventions. Increased understanding of the pathogenesis of bone disease has led to the discovery and clinical utility of bone-targeted agents other than bisphosphonates and denosumab, currently, the standard of care in this setting. AREAS COVERED: In this review, we present the recent advances in molecular targeted therapies focusing on therapies that inhibit bone resorption and/or stimulate bone formation and novel anti-tumoral agents that exerts significant effects on skeletal metastases, nowadays available in clinical practice or in phase of development. EXPERT OPINION: New emergent bone target therapies radium-223, mTOR inhibitors, anti-androgens have demonstrated the ability to increase overall survival in bone metastatic patients, other compounds, such as ET-1 and SRC inhibitors, up to now failed to clearly confirm in clinical trials their promising preclinical data.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Denosumab/pharmacology , Denosumab/therapeutic use , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Drug Design , Humans , Molecular Targeted Therapy , Survival Rate , Tumor Microenvironment
6.
Molecules ; 19(7): 10115-28, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-25019555

ABSTRACT

The development of bone metastases requires multistep and multicellular machinery consisting not only of processes shared with any type of metastases (formation of a pre-metastatic niche, chemotaxis of tumor cells into the host tissue, tumor cells escape from the microvasculature), but also biological interactions that are strictly related to the particular bone microenvironment (bone marrow colonization by cancer cells, osteomimicry, deregulation of bone homeostasis). MiRNAs are highly conserved, small RNAs molecules that regulate gene expression. The functional consequence of miRNA deregulation lies in the mRNA targets whose expression is altered. MiRNA networks acting as upstream regulators of these genes interfere with the initial steps of tumor local invasion and cancer cell intravasation, mainly by regulating the epithelial-mesenchymal transition, the motility, invasiveness and survival abilities of these cells. The miRNA-mediated regulation on the steps of bone tropism, anchorage, homing and finally bone colonization is more tissue specific, being dependent on the expression pattern of target miRNAs in bone marrow sinusoids, bone cells and microenvironment. In that, miRNA specific expression signatures that can distinguish between primary tumors from their corresponding bone metastases might be determinants of clinical aggressiveness. In this review, we focus on the current advances on functions and molecular mechanisms by which miRNAs exert their biological roles in regulating bone metastases development.


Subject(s)
Bone Neoplasms , RNA, Neoplasm , Tumor Microenvironment , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Metastasis , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
7.
Heliyon ; 9(7): e17842, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37456014

ABSTRACT

The mammary gland (MG) is composed of three main epithelial lineages, the basal cells (BC), the estrogen receptor (ER) positive luminal cells (ER+ LC), and the ER negative LC (ER- LC). Defining the cell identity of each lineage and how it is modulated throughout the different stages of life is important to understand how these cells function and communicate throughout life. Here, we used transgenic mice specifically labelling ER+ LC combined to cell surface markers to isolate with high purity the 3 distinct cell lineages of the mammary gland and defined their expression profiles and chromatin landscapes by performing bulk RNAseq and ATACseq of these isolated populations in puberty, adulthood and mid-pregnancy. Our analysis identified conserved genes, ligands and transcription factor (TF) associated with a specific lineage throughout life as well as genes, ligands and TFs specific for a particular stage of the MG. In summary, our study identified genes and TF network associated with the identity, function and cell-cell communication of the different epithelial lineages of the MG at different stages of life.

8.
Curr Neuropharmacol ; 17(1): 59-83, 2019.
Article in English | MEDLINE | ID: mdl-28676012

ABSTRACT

BACKGROUND: Statins represent a class of medications widely prescribed to efficiently treat dyslipidemia. These drugs inhibit 3-ßhydroxy 3ß-methylglutaryl Coenzyme A reductase (HMGR), the rate-limiting enzyme of mevalonate (MVA) pathway. Besides cholesterol, MVA pathway leads to the production of several other compounds, which are essential in the regulation of a plethora of biological activities, including in the central nervous system. For these reasons, statins are able to induce pleiotropic actions, and acquire increased interest as potential and novel modulators in brain processes, especially during pathological conditions. OBJECTIVE: The purpose of this review is to summarize and examine the current knowledge about pharmacokinetic and pharmacodynamic properties of statins in the brain. In addition, effects of statin on brain diseases are discussed providing the most up-to-date information. METHODS: Relevant scientific information was identified from PubMed database using the following keywords: statins and brain, central nervous system, neurological diseases, neurodegeneration, brain tumors, mood, stroke. RESULTS: 315 scientific articles were selected and analyzed for the writing of this review article. Several papers highlighted that statin treatment is effective in preventing or ameliorating the symptomatology of a number of brain pathologies. However, other studies failed to demonstrate a neuroprotective effect. CONCLUSION: Even though considerable research studies suggest pivotal functional outcomes induced by statin therapy, additional investigation is required to better determine the pharmacological effectiveness of statins in the brain, and support their clinical use in the management of different neuropathologies.


Subject(s)
Brain Diseases/drug therapy , Brain/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Animals , Brain/metabolism , Brain Diseases/metabolism , Brain Diseases/prevention & control , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Randomized Controlled Trials as Topic
9.
Nat Cell Biol ; 20(6): 666-676, 2018 06.
Article in English | MEDLINE | ID: mdl-29784918

ABSTRACT

The mammary gland is composed of basal cells and luminal cells. It is generally believed that the mammary gland arises from embryonic multipotent progenitors, but it remains unclear when lineage restriction occurs and what mechanisms are responsible for the switch from multipotency to unipotency during its morphogenesis. Here, we perform multicolour lineage tracing and assess the fate of single progenitors, and demonstrate the existence of a developmental switch from multipotency to unipotency during embryonic mammary gland development. Molecular profiling and single cell RNA-seq revealed that embryonic multipotent progenitors express a unique hybrid basal and luminal signature and the factors associated with the different lineages. Sustained p63 expression in embryonic multipotent progenitors promotes unipotent basal cell fate and was sufficient to reprogram adult luminal cells into basal cells by promoting an intermediate hybrid multipotent-like state. Altogether, this study identifies the timing and the mechanisms mediating early lineage segregation of multipotent progenitors during mammary gland development.


Subject(s)
Cell Lineage , Epithelial Cells/physiology , Mammary Glands, Animal/physiology , Mouse Embryonic Stem Cells/physiology , Multipotent Stem Cells/physiology , Animals , Epithelial Cells/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gestational Age , Mammary Glands, Animal/embryology , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Morphogenesis , Mouse Embryonic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Sequence Analysis, RNA/methods , Signal Transduction , Single-Cell Analysis/methods , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome
10.
Nat Cell Biol ; 20(9): 1099, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30018320

ABSTRACT

In the version of this Article originally published, ref. 52 was incorrectly only attributed to its corresponding author, Fre, S., and an older title was used. The correct citation should have been: Lilja, A. M. et al. Clonal analysis of Notch1-expressing cells reveals the existence of unipotent stem cells that retain long-term plasticity in the embryonic mammary gland. Nat. Cell Biol. https://doi.org/10.1038/s41556-018-0108-1 (2018)'. This has now been amended in all online versions of the Article.

11.
Cell Rep ; 20(7): 1525-1532, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813665

ABSTRACT

The mammary gland (MG) is composed of different cell lineages, including the basal and the luminal cells (LCs) that are maintained by distinct stem cell (SC) populations. LCs can be subdivided into estrogen receptor (ER)+ and ER- cells. LCs act as the cancer cell of origin in different types of mammary tumors. It remains unclear whether the heterogeneity found in luminal-derived mammary tumors arises from a pre-existing heterogeneity within LCs. To investigate LC heterogeneity, we used lineage tracing to assess whether the ER+ lineage is maintained by multipotent SCs or by lineage-restricted SCs. To this end, we generated doxycycline-inducible ER-rtTA mice that allowed us to perform genetic lineage tracing of ER+ LCs and study their fate and long-term maintenance. Our results show that ER+ cells are maintained by lineage-restricted SCs that exclusively contribute to the expansion of the ER+ lineage during puberty and their maintenance during adult life.


Subject(s)
Cell Tracking/methods , Epithelial Cells/cytology , Homeostasis/genetics , Mammary Glands, Animal/cytology , Receptors, Estrogen/genetics , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Doxycycline/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Female , Founder Effect , Gene Expression/drug effects , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Receptors, Estrogen/metabolism , Regeneration/genetics , Stem Cell Transplantation , Stem Cells/metabolism
12.
Mater Sci Eng C Mater Biol Appl ; 77: 927-934, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28532113

ABSTRACT

Regenerative medicine is taking great advantage from the use of biomaterials in the treatments of a wide range of diseases and injuries. Among other biomaterials, self-assembling peptides are appealing systems due to their ability to spontaneously form nanostructured hydrogels that can be directly injected into lesions. Indeed, self-assembling peptide scaffolds are expected to behave as biomimetic matrices able to surround cells, to promote specific interactions, and to control and modify cell behavior by mimicking the native environment as well. We selected three pentadecapeptides inspired by Human Tropoelastin, a natural protein of the extracellular matrix, expected to show high biocompatibility. Moreover, the here proposed self-assembling peptides (SAPs) are able to spontaneously aggregate in nanofibers in biological environment, as revealed by AFM (Atomic Force Microscopy). Peptides were characterized by XPS (X-ray Photoelectron Spectroscopy) and IRRAS (Infrared Reflection Absorption Spectroscopy) both as lyophilized (not aggregated) and as aggregated (nanofibers) samples in order to investigate some potential differences in their chemical composition and intermolecular interactions, and to analyze the surface and interface of nanofibers. Finally, an accurate investigation of the biological properties of the SAPs and of their interaction with cells was performed by culturing for the first time human Mesenchymal Stem Cells (hMSCs) in presence of SAPs. The final aim of this work was to assess if Human Tropoelastin-inspired nanostructured fibers could exert a cytotoxic effect and to evaluate their biocompatibility, cellular adhesion and proliferation.


Subject(s)
Nanofibers , Cell Proliferation , Elastin , Humans , Peptides , Tropoelastin
13.
Sci Rep ; 7(1): 8965, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827726

ABSTRACT

The possibility of detecting and classifying living cells in a label-free and non-invasive manner holds significant theranostic potential. In this work, Hyperspectral Imaging (HSI) has been successfully applied to the analysis of macrophagic polarization, given its central role in several pathological settings, including the regulation of tumour microenvironment. Human monocyte derived macrophages have been investigated using hyperspectral reflectance confocal microscopy, and hyperspectral datasets have been analysed in terms of M1 vs. M2 polarization by Principal Components Analysis (PCA). Following PCA, Linear Discriminant Analysis has been implemented for semi-automatic classification of macrophagic polarization from HSI data. Our results confirm the possibility to perform single-cell-level in vitro classification of M1 vs. M2 macrophages in a non-invasive and label-free manner with a high accuracy (above 98% for cells deriving from the same donor), supporting the idea of applying the technique to the study of complex interacting cellular systems, such in the case of tumour-immunity in vitro models.


Subject(s)
Macrophages/classification , Macrophages/cytology , Microscopy, Confocal/methods , Optical Imaging/methods , Humans , Multivariate Analysis , Principal Component Analysis
14.
Oncotarget ; 8(12): 20113-20121, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28223547

ABSTRACT

Cabozantinib, a c-MET and vascular endothelial growth factor receptor 2 inhibitor, demonstrated to prolong progression free survival and improve skeletal disease-related endpoints in castration-resistant prostate cancer and in metastatic renal carcinoma. Our purpose is to investigate the direct effect of cabozantinib on bone microenvironment using a total human model of primary osteoclasts and osteoblasts.Osteoclasts were differentiated from monocytes isolated from healthy donors; osteoblasts were derived from human mesenchymal stem cells obtained from bone fragments of orthopedic surgery patients. Osteoclast activity was evaluated by tartrate resistant acid phosphatase (TRAP) staining and bone resorption assays and osteoblast differentiation was detected by alkaline phosphatase and alizarin red staining.Our results show that non-cytotoxic doses of cabozantinib significantly inhibit osteoclast differentiation (p=0.0145) and bone resorption activity (p=0.0252). Moreover, cabozantinib down-modulates the expression of osteoclast marker genes, TRAP (p=0.006), CATHEPSIN K (p=0.004) and Receptor Activator of Nuclear Factor k B (RANK) (p=0.001). Cabozantinib treatment has no effect on osteoblast viability or differentiation, but increases osteoprotegerin mRNA (p=0.015) and protein levels (p=0.004) and down-modulates Receptor Activator of Nuclear Factor k B Ligand (RANKL) at both mRNA (p<0.001) and protein levels (p=0.043). Direct cell-to-cell contact between cabozantinib pre-treated osteoblasts and untreated osteoclasts confirmed the indirect anti-resorptive effect of cabozantinib.We demonstrate that cabozantinib inhibits osteoclast functions "directly" and "indirectly" reducing the RANKL/osteoprotegerin ratio in osteoblasts.


Subject(s)
Anilides/pharmacology , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Osteoblasts/physiology , Osteoclasts/physiology , Osteogenesis/physiology , Pyridines/pharmacology , Apoptosis , Blotting, Western , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Proliferation , Cells, Cultured , Humans , Immunoenzyme Techniques , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Oncotarget ; 6(14): 12520-8, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25904051

ABSTRACT

Abiraterone acetate (ABI) is associated not only with a significant survival advantage in both chemotherapy-naive and -treated patients with metastatic castration-resistant prostate cancer (mCRPC), but also with a delay in time to development of Skeletal Related Events and in radiological skeletal progression. These bone benefits may be related to a direct effect on prostate cancer cells in bone or to a specific mechanism directed to bone microenvironment. To test this hypothesis we designed an in vitro study aimed to evaluate a potential direct effect of ABI on human primary osteoclasts/osteoblasts (OCLs/OBLs). We also assessed changes in bone turnover markers, serum carboxy-terminal collagen crosslinks (CTX) and alkaline phosphatase (ALP), in 49 mCRPC patients treated with ABI.Our results showed that non-cytotoxic doses of ABI have a statistically significant inhibitory effect on OCL differentiation and activity inducing a down-modulation of OCL marker genes TRAP, cathepsin K and metalloproteinase-9. Furthermore ABI promoted OBL differentiation and bone matrix deposition up-regulating OBL specific genes, ALP and osteocalcin. Finally, we observed a significant decrease of serum CTX values and an increase of ALP in ABI-treated patients.These findings suggest a novel biological mechanism of action of ABI consisting in a direct bone anabolic and anti-resorptive activity.


Subject(s)
Androstenes/pharmacology , Antineoplastic Agents/pharmacology , Bone Remodeling/drug effects , Bone and Bones/drug effects , Osteoblasts/drug effects , Osteoclasts/drug effects , Alkaline Phosphatase/blood , Blotting, Western , Cell Differentiation/drug effects , Collagen Type I/blood , Enzyme-Linked Immunosorbent Assay , Humans , Male , Peptides/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/drug therapy , Real-Time Polymerase Chain Reaction , Transcriptome
16.
Oncotarget ; 6(12): 10586-91, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25888631

ABSTRACT

Liposarcoma (LPS) is the most common soft tissue sarcoma. It has been demonstrated that mir-155 was the most overexpressed miRNA in well-differentiated LPS(WDLPS)/dedifferentiated LPS (DDLPS). The aim of this study is to evaluate the involvement of Dicer, Drosha and mir-155 in development of LPS and their possible role in stratification of different histological subtypes. Dicer, Drosha and mir-155 mRNA levels were analyzed in formalin-fixed paraffin-embedded specimens from patients diagnosed with 62 LPS and compared with samples of adipose tissues of healthy donors. The experimental data were obtained using qRT-PCR comparing Dicer, Drosha and mir-155 expression levels in tumor samples versus normal fat. The tumor samples from LPS patients showed a significantly lower Dicer expression versus normal adipose tissue, while Drosha levels did not differ. Concerning mir155 expression levels, our results demonstrated a significant mir-155 up-regulation in all LPS subtypes versus normal adipose tissue except for WDLS. These findings demonstrate for the first time that Dicer is deregulated in LPS and show that mir-155 is differentially expressed in LPS subgroups and it could be a promising tool to improve LPS disease stratification and differential diagnosis.


Subject(s)
DEAD-box RNA Helicases/biosynthesis , Liposarcoma/metabolism , MicroRNAs/biosynthesis , Ribonuclease III/biosynthesis , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Humans , Liposarcoma/genetics , Liposarcoma/pathology , Male , MicroRNAs/genetics , Microarray Analysis , Retrospective Studies , Ribonuclease III/genetics , Ribonuclease III/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL