Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(8): 1990-2019, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33811810

ABSTRACT

The population is aging at a rate never seen before in human history. As the number of elderly adults grows, it is imperative we expand our understanding of the underpinnings of aging biology. Human lungs are composed of a unique panoply of cell types that face ongoing chemical, mechanical, biological, immunological, and xenobiotic stress over a lifetime. Yet, we do not fully appreciate the mechanistic drivers of lung aging and why age increases the risk of parenchymal lung disease, fatal respiratory infection, and primary lung cancer. Here, we review the molecular and cellular aspects of lung aging, local stress response pathways, and how the aging process predisposes to the pathogenesis of pulmonary disease. We place these insights into context of the COVID-19 pandemic and discuss how innate and adaptive immunity within the lung is altered with age.


Subject(s)
Aging , Cellular Senescence , Lung Diseases , Lung , Adaptive Immunity , Aged , Aging/immunology , Aging/pathology , COVID-19/immunology , COVID-19/pathology , Humans , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/pathology , Oxidative Stress
2.
Cell ; 180(1): 20-22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31951518

ABSTRACT

Idiopathic pulmonary fibrosis is a fatal disease involving destruction of the lung alveolar structure. In this issue of Cell, Wu et al. (2020) show that impaired alveolar (AT2) stem cells produce mechanical tension that leads to spatially regulated fibrosis, initiating a new chapter in understanding what underlies the periphery to center progression of this lung disease.


Subject(s)
Alveolar Epithelial Cells , Pulmonary Fibrosis , Humans , Pulmonary Alveoli , Stem Cells , Stress, Mechanical
3.
Cell ; 169(4): 563-565, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475887

ABSTRACT

Tumors have long been suspected of hijacking stem cell mechanisms used for tissue maintenance and repair. Ge et al. now show that skin tumors exhibit merged chromatin profiles from distinct stem cell lineages. This "lineage infidelity" recreates a state akin to transient wound repair that persists to maintain uncontrolled growth.


Subject(s)
Cell Lineage , Stem Cells , Chromatin , Humans , Skin Neoplasms , Wound Healing
4.
Cell ; 170(6): 1149-1163.e12, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886383

ABSTRACT

The diversity of mesenchymal cell types in the lung that influence epithelial homeostasis and regeneration is poorly defined. We used genetic lineage tracing, single-cell RNA sequencing, and organoid culture approaches to show that Lgr5 and Lgr6, well-known markers of stem cells in epithelial tissues, are markers of mesenchymal cells in the adult lung. Lgr6+ cells comprise a subpopulation of smooth muscle cells surrounding airway epithelia and promote airway differentiation of epithelial progenitors via Wnt-Fgf10 cooperation. Genetic ablation of Lgr6+ cells impairs airway injury repair in vivo. Distinct Lgr5+ cells are located in alveolar compartments and are sufficient to promote alveolar differentiation of epithelial progenitors through Wnt activation. Modulating Wnt activity altered differentiation outcomes specified by mesenchymal cells. This identification of region- and lineage-specific crosstalk between epithelium and their neighboring mesenchymal partners provides new understanding of how different cell types are maintained in the adult lung.


Subject(s)
Lung/cytology , Mesoderm/cytology , Animals , Homeostasis , Lung/physiology , Mice , Organoids/cytology , Pulmonary Alveoli/cytology , Receptors, G-Protein-Coupled/analysis , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic
5.
Cell ; 156(3): 440-55, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485453

ABSTRACT

Lung stem cells are instructed to produce lineage-specific progeny through unknown factors in their microenvironment. We used clonal 3D cocultures of endothelial cells and distal lung stem cells, bronchioalveolar stem cells (BASCs), to probe the instructive mechanisms. Single BASCs had bronchiolar and alveolar differentiation potential in lung endothelial cell cocultures. Gain- and loss-of-function experiments showed that BMP4-Bmpr1a signaling triggers calcineurin/NFATc1-dependent expression of thrombospondin-1 (Tsp1) in lung endothelial cells to drive alveolar lineage-specific BASC differentiation. Tsp1 null mice exhibited defective alveolar injury repair, confirming a crucial role for the BMP4-NFATc1-TSP1 axis in lung epithelial differentiation and regeneration in vivo. Discovery of this pathway points to methods to direct the derivation of specific lung epithelial lineages from multipotent cells. These findings elucidate a pathway that may be a critical target in lung diseases and provide tools to understand the mechanisms of respiratory diseases at the single-cell level.


Subject(s)
Bronchioles/cytology , Cell Differentiation , Endothelial Cells/metabolism , Pulmonary Alveoli/cytology , Signal Transduction , Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bronchioles/metabolism , Cells, Cultured , Coculture Techniques , Mice , NFATC Transcription Factors/metabolism , Pulmonary Alveoli/metabolism , Stem Cells/cytology , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
6.
EMBO J ; 43(14): 2843-2861, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38755258

ABSTRACT

Glycine-12 mutations in the GTPase KRAS (KRASG12) are an initiating event for development of lung adenocarcinoma (LUAD). KRASG12 mutations promote cell-intrinsic rewiring of alveolar type-II progenitor (AT2) cells, but to what extent such changes interplay with lung homeostasis and cell fate pathways is unclear. Here, we generated single-cell RNA-seq (scRNA-seq) profiles from AT2-mesenchyme organoid co-cultures, mice, and stage-IA LUAD patients, identifying conserved regulators of AT2 transcriptional dynamics and defining the impact of KRASG12D mutation with temporal resolution. In AT2WT organoids, we found a transient injury/plasticity state preceding AT2 self-renewal and AT1 differentiation. Early-stage AT2KRAS cells exhibited perturbed gene expression dynamics, most notably retention of the injury/plasticity state. The injury state in AT2KRAS cells of patients, mice, and organoids was distinguishable from AT2WT states via altered receptor expression, including co-expression of ITGA3 and SRC. The combination of clinically relevant KRASG12D and SRC inhibitors impaired AT2KRAS organoid growth. Together, our data show that an injury/plasticity state essential for lung repair is co-opted during AT2 self-renewal and LUAD initiation, suggesting that early-stage LUAD may be susceptible to interventions that target specifically the oncogenic nature of this cell state.


Subject(s)
Lung Neoplasms , Organoids , Proto-Oncogene Proteins p21(ras) , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Cell Differentiation , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Mutation , Organoids/metabolism , Organoids/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , src-Family Kinases/metabolism , src-Family Kinases/genetics
7.
Article in English | MEDLINE | ID: mdl-39115548

ABSTRACT

RATIONALE: Despite significant advances in precision treatments and immunotherapy, lung cancer is the most common cause of cancer death worldwide. To reduce incidence and improve survival rates, a deeper understanding of lung premalignancy and the multistep process of tumorigenesis is essential, allowing for timely and effective intervention before cancer development. OBJECTIVES: To summarize existing information, identify knowledge gaps, formulate research questions, prioritize potential research topics, and propose strategies for future investigations into the premalignant progression in the lung. METHODS: An international multidisciplinary team of basic, translational, and clinical scientists reviewed available data to develop and refine research questions pertaining to the transformation of premalignant lung lesions to advanced lung cancer. RESULTS: This research statement identifies significant gaps in knowledge and proposes potential research questions aimed at expanding our understanding of the mechanisms underlying the progression of premalignant lung lesions to lung cancer in an effort to explore potential innovative modalities to intercept lung cancer at its nascent stages. CONCLUSIONS: The identified gaps in knowledge about the biological mechanisms of premalignant progression in the lung, along with ongoing challenges in screening, detection, and early intervention, highlight the critical need to prioritize research in this domain. Such focused investigations are essential to devise effective preventive strategies that may ultimately decrease lung cancer incidence and improve patient outcomes.

8.
Nature ; 563(7732): E27, 2018 11.
Article in English | MEDLINE | ID: mdl-30250255

ABSTRACT

We wish to correct two mutations in Supplementary Table 4 of this Letter. The NCI-H460 cell line was annotated as being mutant for TP53. NCI-H460 has been verified to be TP53 wild type by several sources1. The NCI-H2009 cell line was annotated as being mutant for PIK3CA. As annotated by COSMIC (ref. 24 of the original Letter) and CCLE (ref. 25 of the original Letter), the NCI-H2009 cell line has a mutation in PIK3C3, rather than PIK3CA. The cell line is wild type for PIK3CA. The Supplementary Information of this Amendment contains the corrected Supplementary Table 4. These errors do not affect our conclusions. The original Letter has not been corrected.

9.
Am J Respir Cell Mol Biol ; 65(1): 22-29, 2021 07.
Article in English | MEDLINE | ID: mdl-33625958

ABSTRACT

The National Heart, Lung, and Blood Institute of the National Institutes of Health, together with the Longfonds BREATH consortium, convened a working group to review the field of lung regeneration and suggest avenues for future research. The meeting took place on May 22, 2019, at the American Thoracic Society 2019 conference in Dallas, Texas, United States, and brought together investigators studying lung development, adult stem-cell biology, induced pluripotent stem cells, biomaterials, and respiratory disease. The purpose of the working group was 1) to examine the present status of basic science approaches to tackling lung disease and promoting lung regeneration in patients and 2) to determine priorities for future research in the field.


Subject(s)
Induced Pluripotent Stem Cells , Lung Diseases , Lung/physiology , Regeneration , Respiratory Mucosa/physiology , Animals , Cell- and Tissue-Based Therapy , Congresses as Topic , Education , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Lung Diseases/metabolism , Lung Diseases/therapy , National Heart, Lung, and Blood Institute (U.S.) , United States
10.
Nature ; 520(7546): 239-42, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25629630

ABSTRACT

Non-small-cell lung cancer is the leading cause of cancer-related death worldwide. Chemotherapies such as the topoisomerase II (TopoII) inhibitor etoposide effectively reduce disease in a minority of patients with this cancer; therefore, alternative drug targets, including epigenetic enzymes, are under consideration for therapeutic intervention. A promising potential epigenetic target is the methyltransferase EZH2, which in the context of the polycomb repressive complex 2 (PRC2) is well known to tri-methylate histone H3 at lysine 27 (H3K27me3) and elicit gene silencing. Here we demonstrate that EZH2 inhibition has differential effects on the TopoII inhibitor response of non-small-cell lung cancers in vitro and in vivo. EGFR and BRG1 mutations are genetic biomarkers that predict enhanced sensitivity to TopoII inhibitor in response to EZH2 inhibition. BRG1 loss-of-function mutant tumours respond to EZH2 inhibition with increased S phase, anaphase bridging, apoptosis and TopoII inhibitor sensitivity. Conversely, EGFR and BRG1 wild-type tumours upregulate BRG1 in response to EZH2 inhibition and ultimately become more resistant to TopoII inhibitor. EGFR gain-of-function mutant tumours are also sensitive to dual EZH2 inhibition and TopoII inhibitor, because of genetic antagonism between EGFR and BRG1. These findings suggest an opportunity for precision medicine in the genetically complex disease of non-small-cell lung cancer.


Subject(s)
DNA Helicases/genetics , Genes, erbB-1/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Polycomb Repressive Complex 2/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Transcription Factors/genetics , Anaphase/drug effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Etoposide/pharmacology , Etoposide/therapeutic use , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Molecular Targeted Therapy , Xenograft Model Antitumor Assays
11.
Mol Cell ; 45(3): 330-43, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22325351

ABSTRACT

Polycomb repressive complexes (PRCs) play key roles in developmental epigenetic regulation. Yet the mechanisms that target PRCs to specific loci in mammalian cells remain incompletely understood. In this study we show that Bmi1, a core component of Polycomb Repressive Complex 1 (PRC1), binds directly to the Runx1/CBFß transcription factor complex. Genome-wide studies in megakaryocytic cells demonstrate significant chromatin occupancy overlap between the PRC1 core component Ring1b and Runx1/CBFß and functional regulation of a considerable fraction of commonly bound genes. Bmi1/Ring1b and Runx1/CBFß deficiencies generate partial phenocopies of one another in vivo. We also show that Ring1b occupies key Runx1 binding sites in primary murine thymocytes and that this occurs via PRC2-independent mechanisms. Genetic depletion of Runx1 results in reduced Ring1b binding at these sites in vivo. These findings provide evidence for site-specific PRC1 chromatin recruitment by core binding transcription factors in mammalian cells.


Subject(s)
Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Chromatography, Affinity , Cluster Analysis , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/genetics , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Hematopoietic Stem Cells/physiology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Binding , Protein Multimerization , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , T-Lymphocytes/metabolism , Thymocytes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Zebrafish/embryology , Zebrafish/genetics
13.
Am J Respir Cell Mol Biol ; 59(2): 237-245, 2018 08.
Article in English | MEDLINE | ID: mdl-29447458

ABSTRACT

Metastatic disease is the primary cause of death of patients with lung cancer, but the mouse models of lung adenocarcinoma do not accurately recapitulate the tumor microenvironment or metastatic disease observed in patients. In this study, we conditionally deleted E-cadherin in an autochthonous lung adenocarcinoma mouse model driven by activated oncogenic Kras and p53 loss. Loss of E-cadherin significantly accelerated lung adenocarcinoma progression and decreased survival of the mice. Kras;p53;E-cadherin mice had a 41% lung tumor burden, invasive grade 4 tumors, and a desmoplastic stroma just 8 weeks after tumor initiation. One hundred percent of the mice developed local metastases to the lymph nodes or chest wall, and 38% developed distant metastases to the liver or kidney. Lung adenocarcinoma cancer cell lines derived from these tumors also had high migratory rates. These studies demonstrate that the Kras;p53;E-cadherin mouse model better emulates the tumor microenvironment and metastases observed in patients with lung adenocarcinoma than previous models and may therefore be useful for studying metastasis and testing new lung cancer treatments in vivo.


Subject(s)
Adenocarcinoma/pathology , Cadherins/metabolism , Lung Neoplasms/pathology , Neoplasm Metastasis , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma/genetics , Animals , Disease Models, Animal , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Mice, Inbred C57BL , Neoplasm Metastasis/genetics , Proto-Oncogene Proteins p21(ras)/genetics
14.
EMBO J ; 38(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30718273
15.
EMBO J ; 33(5): 468-81, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24497554

ABSTRACT

Metastasis is the leading cause of morbidity for lung cancer patients. Here we demonstrate that murine tumor propagating cells (TPCs) with the markers Sca1 and CD24 are enriched for metastatic potential in orthotopic transplantation assays. CD24 knockdown decreased the metastatic potential of lung cancer cell lines resembling TPCs. In lung cancer patient data sets, metastatic spread and patient survival could be stratified with a murine lung TPC gene signature. The TPC signature was enriched for genes in the Hippo signaling pathway. Knockdown of the Hippo mediators Yap1 or Taz decreased in vitro cellular migration and transplantation of metastatic disease. Furthermore, constitutively active Yap was sufficient to drive lung tumor progression in vivo. These results demonstrate functional roles for two different pathways, CD24-dependent and Yap/Taz-dependent pathways, in lung tumor propagation and metastasis. This study demonstrates the utility of TPCs for identifying molecules contributing to metastatic lung cancer, potentially enabling the therapeutic targeting of this devastating disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Lung Neoplasms/pathology , Neoplasm Metastasis/pathology , Phosphoproteins/metabolism , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Disease Models, Animal , Gene Knockdown Techniques , Humans , Lung/pathology , Mice , Phosphoproteins/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
17.
Am J Respir Cell Mol Biol ; 57(6): 711-720, 2017 12.
Article in English | MEDLINE | ID: mdl-28708422

ABSTRACT

Cystic fibrosis (CF) remains the most lethal genetic disease in the Caucasian population. However, there is great variability in clinical phenotypes and survival times, even among patients harboring the same genotype. We identified five patients with CF and a homozygous F508del mutation in the CFTR gene who were in their fifth or sixth decade of life and had shown minimal changes in lung function over a longitudinal period of more than 20 years. Because of the rarity of this long-term nonprogressive phenotype, we hypothesized these individuals may carry rare genetic variants in modifier genes that ameliorate disease severity. Individuals at the extremes of survival time and lung-function trajectory underwent whole-exome sequencing, and the sequencing data were filtered to include rare missense, stopgain, indel, and splicing variants present with a mean allele frequency of <0.2% in general population databases. Epithelial sodium channel (ENaC) mutants were generated via site-directed mutagenesis and expressed for Xenopus oocyte assays. Four of the five individuals carried extremely rare or never reported variants in the SCNN1D and SCNN1B genes of the ENaC. Separately, an independently enriched rare variant in SCNN1D was identified in the Exome Variant Server database associated with a milder pulmonary disease phenotype. Functional analysis using Xenopus oocytes revealed that two of the three variants in δ-ENaC encoded by SCNN1D exhibited hypomorphic channel activity. Our data suggest a potential role for δ-ENaC in controlling sodium reabsorption in the airways, and advance the plausibility of ENaC as a therapeutic target in CF.


Subject(s)
Amino Acid Sequence , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Epithelial Sodium Channels/metabolism , Sequence Deletion , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channels/genetics , Female , Humans , Male , Xenopus , Xenopus laevis
18.
Proc Natl Acad Sci U S A ; 111(28): 10299-304, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982195

ABSTRACT

Lung cancer is notorious for its ability to metastasize, but the pathways regulating lung cancer metastasis are largely unknown. An in vitro system designed to discover factors critical for lung cancer cell migration identified brain-derived neurotrophic factor, which stimulates cell migration through activation of tropomyosin-related kinase B (TrkB; also called NTRK2). Knockdown of TrkB in human lung cancer cell lines significantly decreased their migratory and metastatic ability in vitro and in vivo. In an autochthonous lung adenocarcinoma model driven by activated oncogenic Kras and p53 loss, TrkB deficiency significantly reduced metastasis. Hypoxia-inducible factor-1 directly regulated TrkB expression, and, in turn, TrkB activated Akt signaling in metastatic lung cancer cells. Finally, TrkB expression was correlated with metastasis in patient samples, and TrkB was detected more often in tumors that did not have Kras or epidermal growth factor receptor mutations. These studies demonstrate that TrkB is an important therapeutic target in metastatic lung adenocarcinoma.


Subject(s)
Adenocarcinoma/enzymology , Cell Movement , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lung Neoplasms/enzymology , Membrane Glycoproteins/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Receptor, trkB/biosynthesis , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Membrane Glycoproteins/genetics , Mice, Mutant Strains , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkB/genetics , Signal Transduction/genetics
19.
Stem Cells ; 31(12): 2759-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23681901

ABSTRACT

The view that adult stem cells are lineage restricted has been challenged by numerous reports of bone marrow (BM)-derived cells giving rise to epithelial cells. Previously, we demonstrated that nonhematopoietic BM cells are the primary source of BM-derived lung epithelial cells. Here, we tested the hypothesis that very small embryonic like cells (VSELs) are responsible for this engraftment. We directly compared the level of BM-derived epithelial cells after transplantation of VSELs, hematopoietic stem/progenitor cells, or other nonhematopoietic cells. VSELs clearly had the highest rate of forming epithelial cells in the lung. By transplanting VSELs from donor mice expressing H2B-GFP under a type 2 pneumocyte-specific promoter, we demonstrate that this engraftment occurs by differentiation and not fusion. This is the first report of VSELs differentiating into an endodermal lineage in vivo, thereby potentially crossing germ layer lineages. Our data suggest that Oct4+ VSELs in the adult BM exhibit broad differentiation potential.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Embryonic Stem Cells/cytology , Lung/cytology , Animals , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Guided Tissue Regeneration , Guinea Pigs , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Semin Cancer Biol ; 22(5-6): 462-70, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22548722

ABSTRACT

Tumors have been increasingly recognized as organs with a complexity that approaches, and may even exceed, that of healthy tissues. When viewed from this perspective, the biology of a tumor can be understood only by studying tumor cell heterogeneity and the microenvironment that is constructed during the course of tumorigenesis and malignant progression. Recent work has revealed the existence of cancer stem cells, the "bugs", with the capacity for self-renewal and tumor propagation. In addition, it is now recognized that the tumor microenvironment, the "bed", plays a critical role in supporting cancer stem cells and also may promote neoplasia and malignant progression. The interdependence of the cell-intrinsic features of cancer, including the cancer stem cell "bugs" and the tumor microenvironment "bed", is only beginning to be understood. In this review, we highlight the rapidly evolving concepts about the interactions between tumor stem cells and their microenvironment, the insights gained from studying their normal tissue counterparts, and the questions and controversies surrounding this area of research, with an emphasis on breast and lung cancer. Finally, we address evidence supporting the notion that eliminating the bed as well as the bugs should lead to more effective and personalized cancer treatments that improve patient outcome.


Subject(s)
Neoplastic Stem Cells/metabolism , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Stem Cell Niche/drug effects , Stem Cells/metabolism , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL