Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Cell Mol Life Sci ; 80(3): 79, 2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36867267

ABSTRACT

Pulmonary neuroendocrine (NE) cells represent a small population in the airway epithelium, but despite this, hyperplasia of NE cells is associated with several lung diseases, such as congenital diaphragmatic hernia and bronchopulmonary dysplasia. The molecular mechanisms causing the development of NE cell hyperplasia remains poorly understood. Previously, we showed that the SOX21 modulates the SOX2-initiated differentiation of epithelial cells in the airways. Here, we show that precursor NE cells start to develop in the SOX2 + SOX21 + airway region and that SOX21 suppresses the differentiation of airway progenitors to precursor NE cells. During development, clusters of NE cells start to form and NE cells mature by expressing neuropeptide proteins, such as CGRP. Deficiency in SOX2 resulted in decreased clustering, while deficiency in SOX21 increased both the numbers of NE ASCL1 + precursor cells early in development, and the number of mature cell clusters at E18.5. In addition, at the end of gestation (E18.5), a number of NE cells in Sox2 heterozygous mice, did not yet express CGRP suggesting a delay in maturation. In conclusion, SOX2 and SOX21 function in the initiation, migration and maturation of NE cells.


Subject(s)
Neuroendocrine Cells , SOXB1 Transcription Factors , SOXB2 Transcription Factors , Animals , Mice , Calcitonin Gene-Related Peptide , Cell Differentiation/genetics , Epithelium , Hyperplasia , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism
2.
Aging Cell ; 21(7): e13632, 2022 07.
Article in English | MEDLINE | ID: mdl-35653631

ABSTRACT

Cellular senescence is characterized by a stable proliferation arrest in response to stresses and the acquisition of a senescence-associated secretory phenotype, called SASP, composed of numerous factors including pro-inflammatory molecules, proteases, and growth factors. The SASP affects the environment of senescent cells, especially during aging, by inducing and modulating various phenotypes such as paracrine senescence, immune cell activity, and extracellular matrix deposition and organization, which critically impact various pathophysiological situations, including fibrosis and cancer. Here, we uncover a novel paracrine effect of the SASP: the neuroendocrine transdifferentiation (NED) of some epithelial cancer cells, evidenced both in the breast and prostate. Mechanistically, this effect is mediated by NF-κB-dependent SASP factors, and leads to an increase in intracellular Ca2+ levels. Consistently, buffering Ca2+ by overexpressing the CALB1 buffering protein partly reverts SASP-induced NED, suggesting that the SASP promotes NED through a SASP-induced Ca2+ signaling. Human breast cancer dataset analyses support that NED occurs mainly in p53 WT tumors and in older patients, in line with a role of senescent cells and its secretome, as they are increasing during aging. In conclusion, our work, uncovering SASP-induced NED in some cancer cells, paves the way for future studies aiming at better understanding the functional link between senescent cell accumulation during aging, NED and clinical patient outcome.


Subject(s)
Breast Neoplasms , Cell Transdifferentiation , NF-kappa B , Aged , Breast Neoplasms/metabolism , Cell Transdifferentiation/physiology , Cellular Senescence/genetics , Cellular Senescence/physiology , Humans , Male , NF-kappa B/metabolism , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Secretome
3.
Int J Mol Sci ; 22(24)2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34948181

ABSTRACT

Malignant tumours are traditionally classified according to their organ of origin and whether they are of epithelial (carcinomas) or mesenchymal (sarcomas) origin. By histological appearance the site of origin may often be confirmed. Using same treatment for tumours from the same organ is rational only when there is no principal heterogeneity between the tumours of that organ. Organ tumour heterogeneity is typical for the lungs with small cell and non-small cell tumours, for the kidneys where clear cell renal carcinoma (CCRCC) is the dominating type among other subgroups, and in the stomach with adenocarcinomas of intestinal and diffuse types. In addition, a separate type of neuroendocrine tumours (NETs) is found in most organs. Every cell type able to divide may develop into a tumour, and the different subtypes most often reflect different cell origin. In this article the focus is on the cells of origin in tumours arising in the stomach and kidneys and the close relationship between normal neuroendocrine cells and NETs. Furthermore, that the erythropoietin producing cell may be the cell of origin of CCRCC (a cancer with many similarities to NETs), and that gastric carcinomas of diffuse type may originate from the ECL cell, whereas the endodermal stem cell most probably gives rise to cancers of intestinal type.


Subject(s)
Kidney Neoplasms/classification , Stomach Neoplasms/classification , Adenocarcinoma/classification , Biomarkers, Tumor/metabolism , Carcinoma/classification , Humans , Kidney/metabolism , Kidney/pathology , Neoplasms/classification , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Stomach/metabolism , Stomach/pathology
4.
Int J Mol Sci ; 22(24)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34948351

ABSTRACT

The polybasic juxtamembrane region (5RK) of the plasma membrane neuronal SNARE, syntaxin1A (Syx), was previously shown by us to act as a fusion clamp in PC12 cells, as charge neutralization of 5RK promotes spontaneous and inhibits Ca2+-triggered release. Using a Syx-based FRET probe (CSYS), we demonstrated that 5RK is required for a depolarization-induced Ca+2-dependent opening (close-to-open transition; CDO) of Syx, which involves the vesicular SNARE synaptobrevin2 and occurs concomitantly with Ca2+-triggered release. Here, we investigated the mechanism underlying the CDO requirement for 5RK and identified phosphorylation of Syx at Ser-14 (S14) by casein kinase 2 (CK2) as a crucial molecular determinant. Thus, following biochemical verification that both endogenous Syx and CSYS are constitutively S14 phosphorylated in PC12 cells, dynamic FRET analysis of phospho-null and phospho-mimetic mutants of CSYS and the use of a CK2 inhibitor revealed that the S14 phosphorylation confers the CDO requirement for 5RK. In accord, amperometric analysis of catecholamine release revealed that the phospho-null mutant does not support Ca2+-triggered release. These results identify a functionally important CK2 phosphorylation of Syx that is required for the 5RK-regulation of CDO and for concomitant Ca2+-triggered release. Further, also spontaneous release, conferred by charge neutralization of 5RK, was abolished in the phospho-null mutant.


Subject(s)
Calcium/metabolism , Casein Kinase II/metabolism , Neuroendocrine Cells/metabolism , Syntaxin 1/metabolism , Animals , Cells, Cultured , Exocytosis , Neuroendocrine Cells/cytology , PC12 Cells , Phosphorylation , Rats , Syntaxin 1/chemistry , Xenopus
5.
STAR Protoc ; 2(3): 100787, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34485946

ABSTRACT

The hypothalamic magnocellular neuroendocrine cells (MNCs) project to the posterior pituitary (PPi), regulating reproduction and fluid homeostasis. It has been challenging to selectively label and manipulate MNCs, as they are intermingled with parvocellular neuroendocrine cells projecting to the median eminence. Here, we provide a step-by-step protocol for specifically targeting the MNCs by infusing retrograde viral tracers into the PPi. When combined with optogenetics, chemogenetics, and transgenic animals, this approach allows cell-type-specific manipulation of MNCs in multiple sites for functional dissection. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021) and Tang et al. (2020).


Subject(s)
Hypothalamus/cytology , Neuroendocrine Cells , Optogenetics/methods , Pituitary Gland, Posterior/cytology , Animals , Animals, Genetically Modified , Male , Median Eminence/cytology , Nerve Net/cytology , Nerve Net/physiology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/physiology , Rats , Rats, Sprague-Dawley
6.
Nat Commun ; 12(1): 3970, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172739

ABSTRACT

Rab-GTPases and their interacting partners are key regulators of secretory vesicle trafficking, docking, and fusion to the plasma membrane in neurons and neuroendocrine cells. Where and how these proteins are positioned and organized with respect to the vesicle and plasma membrane are unknown. Here, we use correlative super-resolution light and platinum replica electron microscopy to map Rab-GTPases (Rab27a and Rab3a) and their effectors (Granuphilin-a, Rabphilin3a, and Rim2) at the nanoscale in 2D. Next, we apply a targetable genetically-encoded electron microscopy labeling method that uses histidine based affinity-tags and metal-binding gold-nanoparticles to determine the 3D axial location of these exocytic proteins and two SNARE proteins (Syntaxin1A and SNAP25) using electron tomography. Rab proteins are distributed across the entire surface and t-SNARE proteins at the base of docked vesicles. We propose that the circumferential distribution of Rabs and Rab-effectors could aid in the efficient transport, capture, docking, and rapid fusion of calcium-triggered exocytic vesicles in excitable cells.


Subject(s)
Molecular Imaging/methods , Neuroendocrine Cells/cytology , Secretory Vesicles/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Carbocyanines/chemistry , Cells, Cultured , Exocytosis , Gold , HeLa Cells , Humans , Imaging, Three-Dimensional , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Metal Nanoparticles/chemistry , Microscopy/methods , Neuroendocrine Cells/metabolism , PC12 Cells , Rats , SNARE Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Red Fluorescent Protein
7.
Genes Dev ; 35(11-12): 870-887, 2021 06.
Article in English | MEDLINE | ID: mdl-34016692

ABSTRACT

Small cell lung carcinoma (SCLC) is among the most lethal of all solid tumor malignancies. In an effort to identify novel therapeutic approaches for this recalcitrant cancer type, we applied genome-scale CRISPR/Cas9 inactivation screens to cell lines that we derived from a murine model of SCLC. SCLC cells were particularly sensitive to the deletion of NEDD8 and other neddylation pathway genes. Genetic suppression or pharmacological inhibition of this pathway using MLN4924 caused cell death not only in mouse SCLC cell lines but also in patient-derived xenograft (PDX) models of pulmonary and extrapulmonary small cell carcinoma treated ex vivo or in vivo. A subset of PDX models were exceptionally sensitive to neddylation inhibition. Neddylation inhibition suppressed expression of major regulators of neuroendocrine cell state such as INSM1 and ASCL1, which a subset of SCLC rely upon for cell proliferation and survival. To identify potential mechanisms of resistance to neddylation inhibition, we performed a genome-scale CRISPR/Cas9 suppressor screen. Deletion of components of the COP9 signalosome strongly mitigated the effects of neddylation inhibition in small cell carcinoma, including the ability of MLN4924 to suppress neuroendocrine transcriptional program expression. This work identifies neddylation as a regulator of neuroendocrine cell state and potential therapeutic target for small cell carcinomas.


Subject(s)
Carcinoma, Small Cell/therapy , Cyclopentanes , Lung Neoplasms/therapy , NEDD8 Protein/metabolism , Pyrimidines , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , COP9 Signalosome Complex/genetics , Carcinoma, Small Cell/physiopathology , Cell Death/drug effects , Cell Line, Tumor , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lung Neoplasms/physiopathology , Mice , NEDD8 Protein/genetics , Neuroendocrine Cells/cytology , Neuroendocrine Cells/drug effects , Proteins/genetics , Proteins/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Repressor Proteins/genetics , Sequence Deletion
9.
Science ; 371(6524): 52-57, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33384370

ABSTRACT

Neuroendocrine (NE) cells are epithelial cells that possess many of the characteristics of neurons, including the presence of secretory vesicles and the ability to sense environmental stimuli. The normal physiologic functions of solitary airway NE cells remain a mystery. We show that mouse and human airway basal stem cells sense hypoxia. Hypoxia triggers the direct differentiation of these stem cells into solitary NE cells. Ablation of these solitary NE cells during hypoxia results in increased epithelial injury, whereas the administration of the NE cell peptide CGRP rescues this excess damage. Thus, we identify stem cells that directly sense hypoxia and respond by differentiating into solitary NE cells that secrete a protective peptide that mitigates hypoxic injury.


Subject(s)
Cell Differentiation , Hypoxia/pathology , Neuroendocrine Cells/physiology , Oxygen/physiology , Stem Cells/physiology , Trachea/cytology , Anaerobiosis , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Receptor-Like Protein/metabolism , Cell Count , Gene Deletion , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Mutant Strains , Neuroendocrine Cells/cytology , Prolyl Hydroxylases/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Trans-Activators/genetics
10.
STAR Protoc ; 1(2): 100068, 2020 09 18.
Article in English | MEDLINE | ID: mdl-33111106

ABSTRACT

Pulmonary neuroendocrine cells (PNECs) are sensory cells within the lung airway epithelia. Here, we provide a detailed protocol for generating induced PNECs (iPNECs) from human induced pluripotent stem cells (iPSCs). The cellular and molecular profile of iPNECs resembles primary human PNECs. Primary human PNECs are exceedingly rare, comprising only 1% of the adult lung. Therefore, a self-renewing source of patient-specific iPNECs facilitates the creation of reproducible human cellular models to study lung diseases characterized by PNEC dysfunction. For complete details on the use and execution of this protocol, please refer to Hor et al. (2020).


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Lung/cytology , Neuroendocrine Cells/cytology , Cells, Cultured , Humans
11.
PLoS One ; 15(10): e0239601, 2020.
Article in English | MEDLINE | ID: mdl-33112876

ABSTRACT

APC mutations drive human colorectal cancer (CRC) development. A major contributing factor is colonic stem cell (SC) overpopulation. But, the mechanism has not been fully identified. A possible mechanism is the dysregulation of neuroendocrine cell (NEC) maturation by APC mutations because SCs and NECs both reside together in the colonic crypt SC niche where SCs mature into NECs. So, we hypothesized that sequential inactivation of APC alleles in human colonic crypts leads to progressively delayed maturation of SCs into NECs and overpopulation of SCs. Accordingly, we used quantitative immunohistochemical mapping to measure indices and proportions of SCs and NECs in human colon tissues (normal, adenomatous, malignant), which have different APC-zygosity states. In normal crypts, many cells staining for the colonic SC marker ALDH1 co-stained for chromogranin-A (CGA) and other NEC markers. In contrast, in APC-mutant tissues from familial adenomatous polyposis (FAP) patients, the proportion of ALDH+ SCs progressively increased while NECs markedly decreased. To explain how these cell populations change in FAP tissues, we used mathematical modelling to identify kinetic mechanisms. Computational analyses indicated that APC mutations lead to: 1) decreased maturation of ALDH+ SCs into progenitor NECs (not progenitor NECs into mature NECs); 2) diminished feedback signaling by mature NECs. Biological experiments using human CRC cell lines to test model predictions showed that mature GLP-2R+ and SSTR1+ NECs produce, via their signaling peptides, opposing effects on rates of NEC maturation via feedback regulation of progenitor NECs. However, decrease in this feedback signaling wouldn't explain the delayed maturation because both progenitor and mature NECs are depleted in CRCs. So the mechanism for delayed maturation must explain how APC mutation causes the ALDH+ SCs to remain immature. Given that ALDH is a key component of the retinoic acid (RA) signaling pathway, that other components of the RA pathway are selectively expressed in ALDH+ SCs, and that exogenous RA ligands can induce ALDH+ cancer SCs to mature into NECs, RA signaling must be attenuated in ALDH+ SCs in CRC. Thus, attenuation of RA signaling explains why ALDH+ SCs remain immature in APC mutant tissues. Since APC mutation causes increased WNT signaling in FAP and we found that sequential inactivation of APC in FAP patient tissues leads to progressively delayed maturation of colonic ALDH+ SCs, the hypothesis is developed that human CRC evolves due to an imbalance between WNT and RA signaling.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colon/cytology , Colon/metabolism , Colorectal Neoplasms/genetics , Genes, APC , Glucagon-Like Peptide 2/metabolism , Mutation , Somatostatin/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromogranin A/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Feedback, Physiological , Glucagon-Like Peptide-2 Receptor/metabolism , HCT116 Cells , HT29 Cells , Humans , Mice , Models, Genetic , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction , Stem Cell Niche , Tretinoin/metabolism , Wnt Signaling Pathway
12.
Int J Mol Sci ; 21(14)2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32674311

ABSTRACT

Intestinal organoids have emerged as the new paradigm for modelling the healthy and diseased intestine with patient-relevant properties. In this study, we show directed differentiation of induced pluripotent stem cells towards intestinal-like phenotype within a microfluidic device. iPSCs are cultured against a gel in microfluidic chips of the OrganoPlate, in which they undergo stepwise differentiation. Cells form a tubular structure, lose their stem cell markers and start expressing mature intestinal markers, including markers for Paneth cells, enterocytes and neuroendocrine cells. Tubes develop barrier properties as confirmed by transepithelial electrical resistance (TEER). Lastly, we show that tubules respond to pro-inflammatory cytokine triggers. The whole procedure for differentiation lasts 14 days, making it an efficient process to make patient-specific organoid tubules. We anticipate the usage of the platform for disease modelling and drug candidate screening.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Biomarkers/metabolism , Caco-2 Cells , Cell Line , Cell Line, Tumor , Cytokines/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/metabolism , Lab-On-A-Chip Devices , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Organoids/cytology , Organoids/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism
13.
J Endocrinol Invest ; 43(9): 1259-1269, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32157664

ABSTRACT

PURPOSE: Nowadays, no human neuroendocrine cell models derived from the neural crest are available. In this study, we present non-transformed long-term primary Neural Crest Cells (NCCs) isolated from the trunk region of the neural crest at VIII-XII gestational weeks of human foetuses obtained from voluntary legal abortion. METHODS AND RESULTS: In NCC, quantitative real-time RT PCR demonstrated the expression of neural crest specifier genes, such as Snail1, Snail2/SLUG, Sox10, FoxD3, c-Myc, and p75NTR. Moreover, these cell populations expressed stemness markers (such as Nanog and nestin), as well as markers of motility and invasion (TAGLN, MMP9, CXCR4, and CXCR7), and of neuronal/glial differentiation (MAP2, GFAP, SYP, and TAU). Functional analysis demonstrated that these cells not only possessed high migration properties, but most importantly, they expressed markers of sympatho-adrenal lineage, such as ASCL1 and tyrosine hydroxylase (TH). Moreover, the expression of TH increased after the induction with two different protocols of differentiation towards neuronal and sympatho-adrenal phenotypes. Finally, exposure to conditioned culture media from NCC induced a mature phenotype in a neuronal cell model (namely SH-SY5Y), suggesting that NCC may also act like Schwann precursors. CONCLUSION: This unique human cell model provides a solid tool for future studies addressing the bases of human neural crest-derived neuroendocrine tumours.


Subject(s)
Cell Separation , Fetus/cytology , Neural Crest/cytology , Neuroendocrine Cells/cytology , Cell Differentiation , Cell Line , Cell Movement , Cell Separation/methods , Female , Humans , Neural Crest/embryology , Neural Crest/physiology , Neuroendocrine Cells/physiology , Phenotype , Pregnancy , Primary Cell Culture
14.
J Nucl Med ; 61(11): 1602-1606, 2020 11.
Article in English | MEDLINE | ID: mdl-32169910

ABSTRACT

Neuroendocrine differentiation is associated with treatment failure and poor outcome in metastatic castration-resistant prostate cancer. We investigated the effect of circulating neuroendocrine biomarkers on the efficacy of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT). Methods: Neuroendocrine biomarker profiles (progastrin-releasing peptide, neuron-specific enolase, and chromogranin-A) were analyzed in 50 patients commencing 177Lu-PSMA-617 RLT. The primary endpoint was a prostate-specific antigen response in relation to baseline neuroendocrine marker profiles. An additional endpoint was progression-free survival. Tumor uptake on posttherapeutic scans, a known predictive marker for response, was used as a control variable. Results: Neuroendocrine biomarker profiles were abnormal in most patients. Neuroendocrine biomarker levels did not predict treatment failure or early progression (P ≥ 0.13). By contrast, intense PSMA-ligand uptake in metastases predicted both treatment response (P = 0.0030) and reduced risk of early progression (P = 0.0111). Conclusion: Neuroendocrine marker profiles do not predict an adverse outcome from RLT. By contrast, high ligand uptake was confirmed to be crucial for achieving a tumor response.


Subject(s)
Antigens, Surface/metabolism , Chromogranin A/blood , Dipeptides/therapeutic use , Glutamate Carboxypeptidase II/metabolism , Heterocyclic Compounds, 1-Ring/therapeutic use , Peptide Fragments/blood , Phosphopyruvate Hydratase/blood , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Biomarkers , Cell Differentiation , Dipeptides/adverse effects , Dipeptides/pharmacokinetics , Heterocyclic Compounds, 1-Ring/adverse effects , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Lutetium , Male , Neuroendocrine Cells/chemistry , Neuroendocrine Cells/cytology , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Recombinant Proteins/blood
15.
Cell Signal ; 65: 109463, 2020 01.
Article in English | MEDLINE | ID: mdl-31693875

ABSTRACT

Lung cancer is the leading cause of cancer death for both men and women and accounts for almost 18.4% of all deaths due to cancer worldwide, with the global incidence increasing by approximately 0.5% per year. Lung cancer is regarded as a devastating type of cancer owing to its high prevalence, reduction in the health-related quality of life, frequently delayed diagnosis, low response rate, high toxicity, and resistance to available therapeutic options. The highly heterogeneous nature of this cancer with a proximal-to-distal distribution throughout the respiratory tract dramatically affects its diagnostic and therapeutic management. The diverse composition and plasticity of lung epithelial cells across the respiratory tract are regarded as significant factors underlying lung cancer heterogeneity. Therefore, definitions of the cells of origin for different types of lung cancer are urgently needed to understand lung cancer biology and to achieve early diagnosis and develop cell-targeted therapies. In the present review, we will discuss the current understanding of the cellular and molecular alterations in distinct lung epithelial cells that result in each type of lung cancer.


Subject(s)
Adenocarcinoma of Lung/metabolism , Carcinoma, Squamous Cell/metabolism , Epithelial Cells/cytology , Lung Neoplasms/metabolism , Neoplasms, Basal Cell/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Plasticity , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/pathology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism
16.
Sci Rep ; 9(1): 18471, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804600

ABSTRACT

FTY-720 (Fingolimod) was one of the first compounds authorized for the treatment of multiple sclerosis. Among its other activities, this sphingosine analogue enhances exocytosis in neuroendocrine chromaffin cells, altering the quantal release of catecholamines. Surprisingly, the size of chromaffin granules is reduced within few minutes of treatment, a process that is paralleled by the homotypic fusion of granules and their heterotypic fusion with mitochondria, as witnessed by dynamic confocal and TIRF microscopy. Electron microscopy studies support these observations, revealing the fusion of several vesicles with individual mitochondria to form large, round mixed organelles. This cross-fusion is SNARE-dependent, being partially prevented by the expression of an inactive form of SNAP-25. Fused mitochondria exhibit an altered redox potential, which dramatically enhances cell death. Therefore, the cross-fusion of intracellular organelles appears to be a new mechanism to be borne in mind when considering the effect of FTY-720 on the survival of neuroendocrine cells.


Subject(s)
Chromaffin Granules/drug effects , Fingolimod Hydrochloride/toxicity , Multiple Sclerosis/drug therapy , Neuroendocrine Cells/drug effects , Animals , Cattle , Cells, Cultured , Chromaffin Granules/metabolism , Chromaffin Granules/pathology , Humans , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Primary Cell Culture , Synaptosomal-Associated Protein 25/metabolism , Toxicity Tests
17.
Genes Dev ; 33(23-24): 1718-1738, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31727771

ABSTRACT

More than 90% of small cell lung cancers (SCLCs) harbor loss-of-function mutations in the tumor suppressor gene RB1 The canonical function of the RB1 gene product, pRB, is to repress the E2F transcription factor family, but pRB also functions to regulate cellular differentiation in part through its binding to the histone demethylase KDM5A (also known as RBP2 or JARID1A). We show that KDM5A promotes SCLC proliferation and SCLC's neuroendocrine differentiation phenotype in part by sustaining expression of the neuroendocrine transcription factor ASCL1. Mechanistically, we found that KDM5A sustains ASCL1 levels and neuroendocrine differentiation by repressing NOTCH2 and NOTCH target genes. To test the role of KDM5A in SCLC tumorigenesis in vivo, we developed a CRISPR/Cas9-based mouse model of SCLC by delivering an adenovirus (or an adeno-associated virus [AAV]) that expresses Cre recombinase and sgRNAs targeting Rb1, Tp53, and Rbl2 into the lungs of Lox-Stop-Lox Cas9 mice. Coinclusion of a KDM5A sgRNA decreased SCLC tumorigenesis and metastasis, and the SCLCs that formed despite the absence of KDM5A had higher NOTCH activity compared to KDM5A+/+ SCLCs. This work establishes a role for KDM5A in SCLC tumorigenesis and suggests that KDM5 inhibitors should be explored as treatments for SCLC.


Subject(s)
Cell Differentiation/genetics , Neuroendocrine Cells/cytology , Receptors, Notch/physiology , Retinoblastoma-Binding Protein 2/metabolism , Signal Transduction/genetics , Small Cell Lung Carcinoma/enzymology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Line , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Histone Demethylases/metabolism , Humans , In Vitro Techniques , Mice , Neuroendocrine Cells/pathology , Small Cell Lung Carcinoma/physiopathology
18.
Proc Natl Acad Sci U S A ; 116(44): 22189-22195, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611391

ABSTRACT

The Wnt signaling pathway is of paramount importance for development and disease. However, the tissue-specific regulation of Wnt pathway activity remains incompletely understood. Here we identify FOXB2, an uncharacterized forkhead box family transcription factor, as a potent activator of Wnt signaling in normal and cancer cells. Mechanistically, FOXB2 induces multiple Wnt ligands, including WNT7B, which increases TCF/LEF-dependent transcription without activating Wnt coreceptor LRP6 or ß-catenin. Proximity ligation and functional complementation assays identified several transcription regulators, including YY1, JUN, and DDX5, as cofactors required for FOXB2-dependent pathway activation. Although FOXB2 expression is limited in adults, it is induced in select cancers, particularly advanced prostate cancer. RNA-seq data analysis suggests that FOXB2/WNT7B expression in prostate cancer is associated with a transcriptional program that favors neuronal differentiation and decreases recurrence-free survival. Consistently, FOXB2 controls Wnt signaling and neuroendocrine differentiation of prostate cancer cell lines. Our results suggest that FOXB2 is a tissue-specific Wnt activator that promotes the malignant transformation of prostate cancer.


Subject(s)
Forkhead Transcription Factors/metabolism , Prostatic Neoplasms/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Cell Differentiation , DEAD-box RNA Helicases/metabolism , Forkhead Transcription Factors/genetics , HCT116 Cells , HEK293 Cells , Humans , Male , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , YY1 Transcription Factor/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 317(4): L486-L495, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31313615

ABSTRACT

This article traces the beginnings of the various areas of physiological research on airway epithelium. First mentioned in 1600, it was not until 1834 that it was found to be ciliated. Goblet and basal cells were described in 1852, to be followed by ~10 other epithelial cell types (the most recent in 2018). It also contains nerve endings and resident leukocytes. Mucociliary clearance was documented in 1835, but the first studies on the ciliary beat cycle did not appear until 1890, and a definitive description was not published until 1981. It was established in 1932 that goblet cells in the cat trachea were unresponsive to cholinergic agents; but only since 1980 or so has any significant progress been made on what does cause them to degranulate. Active transfer of salts across epithelia creates local osmotic gradients that drive transepithelial water flows. Vectorial salt transport was first described for airway epithelium in 1968, and the associated volume flows were measured in 1981. Evidence that airway epithelium releases signaling molecules first appeared in 1981. Since then, scores of molecules have been identified. The pace of research in most areas increased dramatically after the development of confluent, polarized cultures of airway epithelium in the early 1980s.


Subject(s)
Alveolar Epithelial Cells/physiology , Cilia/physiology , Goblet Cells/physiology , Leukocytes/physiology , Neuroendocrine Cells/physiology , Alveolar Epithelial Cells/cytology , Animals , Biological Transport , Cats , Cell Communication/physiology , Cilia/ultrastructure , Goblet Cells/cytology , History, 17th Century , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Leukocytes/cytology , Mucociliary Clearance/physiology , Neuroendocrine Cells/cytology , Respiratory System/anatomy & histology , Respiratory System/cytology , Respiratory System/metabolism , Sheep
20.
Curr Top Dev Biol ; 132: 67-89, 2019.
Article in English | MEDLINE | ID: mdl-30797518

ABSTRACT

While the lung is commonly known for its gas exchange function, it is exposed to signals in the inhaled air and responds to them by collaborating with other systems including immune cells and the neural circuit. This important aspect of lung physiology led us to consider the lung as a sensory organ. Among different cell types within the lung that mediate this role, several recent studies have renewed attention on pulmonary neuroendocrine cells (PNECs). PNECs are a rare, innervated airway epithelial cell type that accounts for <1% of the lung epithelium population. They are enriched at airway branch points. Classical in vitro studies have shown that PNECs can respond to an array of aerosol stimuli such as hypoxia, hypercapnia and nicotine. Recent in vivo evidence suggests an essential role of PNECs at neuroimmunomodulatory sites of action, releasing neuropeptides, neurotransmitters and facilitating asthmatic responses to allergen. In addition, evidence supports that PNECs can function both as progenitor cells and progenitor niches following airway epithelial injury. Increases in PNECs have been documented in a large array of chronic lung diseases. They are also the cells-of-origin for small cell lung cancer. A better understanding of the specificity of their responses to distinct insults, their impact on normal lung function and their roles in the pathogenesis of pulmonary ailments will be the next challenge toward designing therapeutics targeting the neuroendocrine system in lung.


Subject(s)
Epithelial Cells/metabolism , Lung/embryology , Neuroendocrine Cells/metabolism , Neurosecretory Systems/embryology , Animals , Cell Lineage/genetics , Epithelial Cells/cytology , Epithelium/embryology , Epithelium/metabolism , Gene Expression Regulation, Developmental , Humans , Lung/cytology , Lung/metabolism , Neuroendocrine Cells/cytology , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Stem Cells/cytology , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...