Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 388
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(15): e23867, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39101950

ABSTRACT

There is a significant difference in prognosis and response to chemotherapy between basal and classical subtypes of pancreatic ductal adenocarcinoma (PDAC). Further biomarkers are required to identify subtypes of PDAC. We selected candidate biomarkers via review articles. Correlations between these candidate markers and the PDAC molecular subtype gene sets were analyzed using bioinformatics, confirming the biomarkers for identifying classical and basal subtypes. Subsequently, 298 PDAC patients were included, and their tumor tissues were immunohistochemically stratified using these biomarkers. Survival data underwent analysis, including Cox proportional hazards modeling. Our results indicate that the pairwise and triple combinations of KRT5/KRT17/S100A2 exhibit a higher correlation coefficient with the basal-like subtype gene set, whereas the corresponding combinations of GATA6/HNF4A/TFF1 show a higher correlation with the classical subtype gene set. Whether analyzing unmatched or propensity-matched data, the overall survival time was significantly shorter for the basal subtype compared with the classical subtype (p < .001), with basal subtype patients also facing a higher risk of mortality (HR = 4.017, 95% CI 2.675-6.032, p < .001). In conclusion, the combined expression of KRT5, KRT17, and S100A2, in both pairwise and triple combinations, independently predicts shorter overall survival in PDAC patients and likely identifies the basal subtype. Similarly, the combined expression of GATA6, HNF4A, and TFF1, in the same manner, may indicate the classical subtype. In our study, the combined application of established biomarkers offers valuable insights for the prognostic evaluation of PDAC patients.


Subject(s)
Biomarkers, Tumor , Carcinoma, Pancreatic Ductal , Keratin-17 , Keratin-5 , Pancreatic Neoplasms , S100 Proteins , Humans , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Male , Female , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Middle Aged , S100 Proteins/genetics , S100 Proteins/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Aged , Keratin-17/genetics , Keratin-17/metabolism , Prognosis , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Adult , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Chemotactic Factors
2.
Genesis ; 62(2): e23596, 2024 04.
Article in English | MEDLINE | ID: mdl-38665067

ABSTRACT

The vomeronasal organ (VNO) is a part of the accessory olfactory system, which detects pheromones and chemical factors that trigger a spectrum of sexual and social behaviors. The vomeronasal epithelium (VNE) shares several features with the epithelium of the main olfactory epithelium (MOE). However, it is a distinct neuroepithelium populated by chemosensory neurons that differ from the olfactory sensory neurons in cellular structure, receptor expression, and connectivity. The vomeronasal organ of rodents comprises a sensory epithelium (SE) and a thin non-sensory epithelium (NSE) that morphologically resembles the respiratory epithelium. Sox2-positive cells have been previously identified as the stem cell population that gives rise to neuronal progenitors in MOE and VNE. In addition, the MOE also comprises p63 positive horizontal basal cells, a second pool of quiescent stem cells that become active in response to injury. Immunolabeling against the transcription factor p63, Keratin-5 (Krt5), Krt14, NrCAM, and Krt5Cre tracing experiments highlighted the existence of horizontal basal cells distributed along the basal lamina of SE of the VNO. Single cell sequencing and genetic lineage tracing suggest that the vomeronasal horizontal basal cells arise from basal progenitors at the boundary between the SE and NSE proximal to the marginal zones. Moreover, our experiments revealed that the NSE of rodents is, like the respiratory epithelium, a stratified epithelium where the p63/Krt5+ basal progenitor cells self-replicate and give rise to the apical columnar cells facing the lumen of the VNO.


Subject(s)
Vomeronasal Organ , Vomeronasal Organ/metabolism , Vomeronasal Organ/cytology , Animals , Mice , Olfactory Mucosa/metabolism , Olfactory Mucosa/cytology , Keratin-15/metabolism , Keratin-15/genetics , Keratin-5/metabolism , Keratin-5/genetics , Keratin-14/metabolism , Keratin-14/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
3.
Am J Physiol Renal Physiol ; 326(6): F1078-F1090, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38634130

ABSTRACT

Urothelium forms a distensible yet impermeable barrier, senses and transduces stimuli, and defends the urinary tract from mechanical, chemical, and bacterial injuries. Biochemical and genetic labeling studies support the existence of one or more progenitor populations with the capacity to rapidly regenerate the urothelium following injury, but slow turnover, a low mitotic index, and inconsistent methodologies obscure progenitor identity. The progenitor properties of basal keratin 5 urothelial cells (K5-UCs) have been previously investigated, but those studies focused on embryonic or adult bladder urothelium. Urothelium undergoes desquamation and apoptosis after birth, which requires postnatal proliferation and restoration. Therefore, we mapped the fate of bladder K5-UCs across postnatal development/maturation and following administration of cyclophosphamide to measure homeostatic and reparative progenitor capacities, respectively. In vivo studies demonstrate that basal K5-UCs are age-restricted progenitors in neonates and juveniles, but not in adult mice. Neonatal K5-UCs retain a superior progenitor capacity in vitro, forming larger and more differentiated urothelial organoids than adult K5-UCs. Accordingly, K5-UC transcriptomes are temporally distinct, with enrichment of transcripts associated with cell proliferation and differentiation in neonates. Induction of urothelial proliferation is sufficient to restore adult K5-UC progenitor capacity. Our findings advance the understanding of urothelial progenitors and support a linear model of urothelial formation and regeneration, which may have significant impact on therapeutic development or tissue engineering strategies.NEW & NOTEWORTHY Fate mapping reveals an important linear relationship, whereby bladder basal urothelial cells give rise to intermediate and superficial cells in an age-restricted manner and contribute to tissue repair. Neonatal basal cells reprise their role as superior progenitors in vitro and display distinct transcriptional signatures, which suggest progenitor function is at least partially cell intrinsic. However, the urothelium progenitor niche cannot be overlooked, since FGF7 rescues adult basal cell progenitor function.


Subject(s)
Cell Proliferation , Keratin-5 , Regeneration , Stem Cells , Urinary Bladder , Urothelium , Animals , Mice , Age Factors , Animals, Newborn , Cell Differentiation , Cells, Cultured , Cyclophosphamide , Fibroblast Growth Factor 7/metabolism , Fibroblast Growth Factor 7/genetics , Gene Expression Regulation, Developmental , Keratin-5/metabolism , Keratin-5/genetics , Mice, Inbred C57BL , Stem Cells/metabolism , Transcriptome , Urinary Bladder/metabolism , Urothelium/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 326(2): L135-L148, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38084407

ABSTRACT

Bronchiolitis obliterans (BO) is a fibrotic lung disease characterized by progressive luminal narrowing and obliteration of the small airways. In the nontransplant population, inhalation exposure to certain chemicals is associated with BO; however, the mechanisms contributing to disease induction remain poorly understood. This study's objective was to use single-cell RNA sequencing for the identification of transcriptomic signatures common to primary human airway epithelial cells after chemical exposure to BO-associated chemicals-diacetyl or nitrogen mustard-to help explain BO induction. Primary airway epithelial cells were cultured at air-liquid interface and exposed to diacetyl, nitrogen mustard, or control vapors. Cultures were dissociated and sequenced for single-cell RNA. Differential gene expression and functional pathway analyses were compared across exposures. In total, 75,663 single cells were captured and sequenced from all exposure conditions. Unbiased clustering identified 11 discrete phenotypes, including 5 basal, 2 ciliated, and 2 secretory cell clusters. With chemical exposure, the proportion of cells assigned to keratin 5+ basal cells decreased, whereas the proportion of cells aligned to secretory cell clusters increased compared with control exposures. Functional pathway analysis identified interferon signaling and antigen processing/presentation as pathways commonly upregulated after diacetyl or nitrogen mustard exposure in a ciliated cell cluster. Conversely, the response of airway basal cells differed significantly with upregulation of the unfolded protein response in diacetyl-exposed basal cells, not seen in nitrogen mustard-exposed cultures. These new insights provide early identification of airway epithelial signatures common to BO-associated chemical exposures.NEW & NOTEWORTHY Bronchiolitis obliterans (BO) is a devastating fibrotic lung disease of the small airways, or bronchioles. This original manuscript uses single-cell RNA sequencing for identifying common signatures of chemically exposed airway epithelial cells in BO induction. Chemical exposure reduced the proportion of keratin 5+ basal cells while increasing the proportion of keratin 4+ suprabasal cells. Functional pathways contributory to these shifts differed significantly across exposures. These new results highlight similarities and differences in BO induction across exposures.


Subject(s)
Bronchiolitis Obliterans , Diacetyl , Humans , Keratin-5/metabolism , Diacetyl/metabolism , Mechlorethamine/metabolism , Respiratory Mucosa/metabolism , Bronchiolitis Obliterans/chemically induced , Bronchiolitis Obliterans/metabolism , Epithelial Cells/metabolism
5.
Exp Dermatol ; 33(8): e15143, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39073059

ABSTRACT

Immortalized keratinocytes can offer a low-cost experimental platform for human skin research, with increased cell yield compared to primary cultures. However, the usefulness of these surrogate cell models is highly dependent on their ability to retain the phenotypic attributes of the parent cells. Keratins K14 and K5 are the hallmarks of undifferentiated, mitotically active basal keratinocytes. We observed occasional progressive loss of K14 expression in growing keratinocyte cell lines, with persistent retention of K5 and an epithelial phenotype, and investigated possible reasons for this. We show that K14 repression occurs by DNA promoter methylation of KRT14 gene and is compounded by histone deacetylation and by the presence of EGF. In vivo, keratinocytes shut down K14 synthesis as they commit to terminal differentiation and move from the basal to spinous layer, but by laser-capture microdissection of human epidermis we could detect no evidence of increased selective KRT14 methylation in this normal process. Loss of K14 expression suggests that epidermal identity of cultured keratinocytes can be compromised in certain tissue culture situations, possibly due to the immortalization method and persistent EGF supplementation.


Subject(s)
DNA Methylation , Keratin-14 , Keratinocytes , Promoter Regions, Genetic , Keratinocytes/metabolism , Humans , Keratin-14/genetics , Keratin-14/metabolism , Cell Differentiation , Keratin-5/genetics , Keratin-5/metabolism , Epidermal Growth Factor/metabolism , Epidermis/metabolism , Histones/metabolism
6.
Histopathology ; 83(3): 426-434, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37195579

ABSTRACT

INTRODUCTION: Adamantinoma-like Ewing sarcoma (ALES) is a rare aggressive malignancy occasionally diagnosed in the thyroid gland. ALES shows basaloid cytomorphology, expresses keratins, p63, p40, frequently CD99, and harbours the t(11;22) EWSR1::FLI1 translocation. There is debate on whether ALES resembles more sarcoma or carcinoma. METHODS: We performed RNA sequencing from two ALES cases and compared findings with skeletal Ewing's sarcomas and nonneoplastic thyroid tissue. ALES was investigated by in situ hybridization (ISH) for high-risk human papillomavirus (HPV) DNA and immunohistochemistry for the following antigens: keratin 7, keratin 20, keratin 5, keratins (AE1/AE3 and CAM5.2), CD45, CD20, CD5, CD99, chromogranin, synaptophysin, calcitonin, thyroglobulin, PAX8, TTF1, S100, p40, p63, p16, NUT, desmin, ER, FLI1, INI1, and myogenin. RESULTS: An uncommon EWSR1::FLI transcript with retained EWSR1 exon 8 was detected in both ALES cases. Regulators of EWSR1::FLI1 splicing (HNRNPH1, SUPT6H, SF3B1) necessary for production of a functional fusion oncoprotein, as well as 53 genes (including TNNT1, NKX2.2) activated downstream to the EWSR1::FLI1 cascade, were overexpressed. Eighty-six genes were uniquely overexpressed in ALES, most of which were related to squamous differentiation. Immunohistochemically, ALES strongly expressed keratins 5, AE1/AE3 and CAM5.2, p63, p40, p16, and focally CD99. INI1 was retained. The remaining immunostains and HPV DNA ISH were negative. CONCLUSION: Comparative transcriptomic profiling reveals overlapping features of ALES with skeletal Ewing's sarcoma and an epithelial carcinoma, as evidenced by immunohistochemical expression of keratin 5, p63, p40, CD99, the transcriptome profile, and detection of EWSR1::FLI1 fusion transcript by RNA sequencing.


Subject(s)
Adamantinoma , Carcinoma , Papillomavirus Infections , Sarcoma, Ewing , Humans , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/genetics , Adamantinoma/diagnosis , Adamantinoma/genetics , Adamantinoma/chemistry , Thyroid Gland/pathology , Transcriptome , Keratin-5/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Transcription Factors/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
7.
Nature ; 550(7677): 529-533, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29019984

ABSTRACT

In several organ systems, the transitional zone between different types of epithelium is a hotspot for pre-neoplastic metaplasia and malignancy, but the cells of origin for these metaplastic epithelia and subsequent malignancies remain unknown. In the case of Barrett's oesophagus, intestinal metaplasia occurs at the gastro-oesophageal junction, where stratified squamous epithelium transitions into simple columnar cells. On the basis of a number of experimental models, several alternative cell types have been proposed as the source of this metaplasia but in all cases the evidence is inconclusive: no model completely mimics Barrett's oesophagus in terms of the presence of intestinal goblet cells. Here we describe a transitional columnar epithelium with distinct basal progenitor cells (p63+KRT5+KRT7+) at the squamous-columnar junction of the upper gastrointestinal tract in a mouse model. We use multiple models and lineage tracing strategies to show that this squamous-columnar junction basal cell population serves as a source of progenitors for the transitional epithelium. On ectopic expression of CDX2, these transitional basal progenitors differentiate into intestinal-like epithelium (including goblet cells) and thereby reproduce Barrett's metaplasia. A similar transitional columnar epithelium is present at the transitional zones of other mouse tissues (including the anorectal junction) as well as in the gastro-oesophageal junction in the human gut. Acid reflux-induced oesophagitis and the multilayered epithelium (believed to be a precursor of Barrett's oesophagus) are both characterized by the expansion of the transitional basal progenitor cells. Our findings reveal a previously unidentified transitional zone in the epithelium of the upper gastrointestinal tract and provide evidence that the p63+KRT5+KRT7+ basal cells in this zone are the cells of origin for multi-layered epithelium and Barrett's oesophagus.


Subject(s)
Barrett Esophagus/pathology , Cell Lineage , Epithelial Cells/pathology , Epithelium/pathology , Esophagogastric Junction/pathology , Stem Cells/pathology , Animals , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cell Tracking , Esophagitis/metabolism , Esophagitis/pathology , Esophagogastric Junction/metabolism , Gastroesophageal Reflux , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Keratin-5/metabolism , Keratin-7/metabolism , Metaplasia/metabolism , Metaplasia/pathology , Mice , Phosphoproteins/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism
8.
Cell Mol Life Sci ; 79(11): 556, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36261680

ABSTRACT

Epithelial cells orchestrate a series of intercellular signaling events in response to tissue damage. While the epididymis is composed of a pseudostratified epithelium that controls the acquisition of male fertility, the maintenance of its integrity in the context of tissue damage or inflammation remains largely unknown. Basal cells of the epididymis contain a primary cilium, an organelle that controls cellular differentiation in response to Hedgehog signaling cues. Hypothesizing its contribution to epithelial homeostasis, we knocked out the ciliary component ARL13B in keratin 5-positive basal cells. In this model, the reduced size of basal cell primary cilia was associated with impaired Hedgehog signaling and the loss of KRT5, KRT14, and P63 basal cell markers. When subjected to tissue injury, the epididymal epithelium from knock-out mice displayed imbalanced rates of cell proliferation/apoptosis and failed to properly regenerate in vivo. This response was associated with changes in the transcriptomic landscape related to immune response, cell differentiation, cell adhesion, and triggered severe hypoplasia of the epithelium. Together our results indicate that the ciliary GTPase, ARL13B, participates in the transduction of the Hedgehog signaling pathway to maintain basal cell stemness needed for tissue regeneration. These findings provide new insights into the role of basal cell primary cilia as safeguards of pseudostratified epithelia.


Subject(s)
ADP-Ribosylation Factors , Epididymis , Hedgehog Proteins , Animals , Male , Mice , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Epididymis/cytology , Epididymis/metabolism , GTP Phosphohydrolases/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Keratin-5/metabolism , Mice, Knockout
9.
Biochem Biophys Res Commun ; 586: 55-62, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34826701

ABSTRACT

Salivary gland hypofunction due to radiation therapy for head and neck cancer or Sjögren syndrome may cause various oral diseases, which can lead to a decline in the quality of life. Cell therapy using salivary gland stem cells is a promising method for restoring hypofunction. Herein, we show that salivary gland-like cells can be induced from epithelial tissues that were transdifferentiated from mouse embryonic fibroblasts (MEFs). We introduced four genes, Dnp63a, Tfap2a, Grhl2, and Myc (PTMG) that are known to transdifferentiate fibroblasts into oral mucosa-like epithelium in vivo into MEFs. MEFs overexpressing these genes showed epithelial cell characteristics, such as cobblestone appearance and E-cadherin positivity, and formed oral epithelial-like tissue under air-liquid interface culture conditions. The epithelial sheet detached from the culture dish was infected with adenoviruses encoding Sox9 and Foxc1, which we previously identified as essential factors to induce salivary gland formation. The cells detached from the cell sheet formed spheres 10 days after infection and showed a branching morphology. The spheres expressed genes encoding basal/myoepithelial markers, cytokeratin 5, cytokeratin 14, acinar cell marker, aquaporin 5, and the myoepithelial marker α-smooth muscle actin. The dissociated cells of these primary spheres had the ability to form secondary spheres. Taken together, our results provide a new strategy for cell therapy of salivary glands and hold implications in treating patients with dry mouth.


Subject(s)
Acinar Cells/metabolism , Fibroblasts/metabolism , Forkhead Transcription Factors/genetics , SOX9 Transcription Factor/genetics , Salivary Glands/metabolism , Spheroids, Cellular/metabolism , Acinar Cells/cytology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Aquaporin 5/genetics , Aquaporin 5/metabolism , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Transdifferentiation/genetics , Cell- and Tissue-Based Therapy/methods , Embryo, Mammalian , Fibroblasts/cytology , Forkhead Transcription Factors/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Keratin-14/genetics , Keratin-14/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/metabolism , Salivary Glands/cytology , Spheroids, Cellular/cytology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Cytokine ; 160: 156022, 2022 12.
Article in English | MEDLINE | ID: mdl-36099756

ABSTRACT

Although conventional knockout and transgenic mouse models have significantly advanced our understanding of Receptor Activator of NF-κB Ligand (RANKL) signaling in intra-thymic crosstalk that establishes self-tolerance and later stages of lymphopoiesis, the unique advantages of conditional mouse transgenesis have yet to be explored. A main advantage of conditional transgenesis is the ability to express a transgene in a spatiotemporal restricted manner, enabling the induction (or de-induction) of transgene expression during predetermined stages of embryogenesis or during defined postnatal developmental or physiological states, such as puberty, adulthood, and pregnancy. Here, we describe the K5: RANKL bigenic mouse, in which transgene derived RANKL expression is induced by doxycycline and targeted to cytokeratin 5 positive medullary thymic epithelial cells (mTECs). Short-term doxycycline induction reveals that RANKL transgene expression is significantly induced in the thymic medulla and only in response to doxycycline. Prolonged doxycycline induction in the K5: RANKL bigenic results in a significantly enlarged thymus in which mTECs are hyperproliferative. Flow cytometry showed that there is a marked enrichment of CD4+ and CD8+ single positive thymocytes with a concomitant depletion of CD4+ CD8+ double positives. Furthermore, there is an increase in the number of FOXP3+ T regulatory (Treg) cells and Ulex Europaeus Agglutinin 1+ (UEA1+) mTECs. Transcriptomics revealed that a remarkable array of signals-cytokines, chemokines, growth factors, transcription factors, and morphogens-are governed by RANKL and drive in part the K5: RANKL thymic phenotype. Extended doxycycline administration to 6-weeks results in a K5: RANKL thymus that begins to display distinct histopathological features, such as medullary epithelial hyperplasia, extensive immune cell infiltration, and central tissue necrosis. As there are intense efforts to develop clinical approaches to restore thymic medullary function in the adult to treat immunopathological conditions in which immune cell function is compromised following cancer therapy or toxin exposure, an improved molecular understanding of RANKL's involvement in thymic medulla enlargement will be required. We believe the versatility of the conditional K5: RANKL mouse represents a tractable model system to assist in addressing this requirement as well as many other questions related to RANKL's role in thymic normal physiology and disease processes.


Subject(s)
Doxycycline , RANK Ligand/metabolism , Transcriptome , Agglutinins/metabolism , Animals , Cytokines/metabolism , Doxycycline/pharmacology , Epithelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Ligands , Mice , Mice, Transgenic , NF-kappa B/metabolism , Phenotype , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Thymus Gland/metabolism
11.
Histopathology ; 80(3): 589-597, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34725848

ABSTRACT

AIMS: Basal-like breast cancer is an aggressive molecular subtype associated with younger age and early relapse. Most cases lack expression of oestrogen receptor (ER), progesterone receptor, and human epidermal growth factor receptor 2, limiting targeted therapeutic options. Basal-like breast cancer is defined by the expression of genes in the outer/basally located epithelial layer of mammary glands, including those encoding cytokeratin (CK) 5 and CK14, and epidermal growth factor receptor (EGFR). SRY-box transcription factor 10 (SOX10), for which there is a readily available immunohistochemical stain, is expressed in a subset of breast cancers, particularly triple-negative carcinomas. In this study, we sought to: (i) assess the association between SOX10 expression and intrinsic molecular subtypes as defined by Prediction Analysis of Microarray 50 (PAM50) gene expression; and (ii) compare the performance of SOX10 with that of other surrogate markers of the basal-like subtype, including CK5, EGFR, nestin, and inositol polyphosphate 4-phosphatase type II (INPP4B). METHODS AND RESULTS: SOX10 immunostaining was performed on tissue microarrays constructed from a contemporary series enriched for ER-negative and weakly ER-positive cancers that had also undergone PAM50 gene profiling. A total of 211 cases were informative for both SOX10 immunohistochemistry and PAM50 subtype, including 103 basal-like cancers. Staining for SOX10 was positive in 73 of 103 basal-like cancers and in only two of 108 cancers of other subtypes (P < 0.001), resulting in a sensitivity of 70.9% and a specificity of 98.1%. SOX10 was more specific than the other tested basal markers, and the results were independent of ER status. CONCLUSIONS: SOX10 is a moderately sensitive, but highly specific, immunohistochemical biomarker for the basal-like intrinsic subtype of breast cancer, which, unlike other commonly used immunohistochemical biomarkers, is independent of hormone receptor status.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Carcinoma, Basal Cell/pathology , SOXE Transcription Factors/metabolism , Carcinoma, Basal Cell/genetics , ErbB Receptors , Female , Humans , Immunohistochemistry , Keratin-5/metabolism , Middle Aged
12.
BMC Cancer ; 22(1): 1014, 2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36153494

ABSTRACT

BACKGROUND: Trophoblast cell-surface antigen 2 (TROP2) is related to tumor proliferation enhancement and poor prognosis. An antibody targeting TROP2 was developed to treat metastatic triple-negative breast cancer (TNBC) which has a limited treatment modality. To characterize the TROP2 expressing tumors in TNBC, we analyzed TROP2 expression in three cohorts; (1) primary tumor without neoadjuvant chemotherapy, (2) primary tumor with neoadjuvant chemotherapy, and (3) metastatic tumor. METHODS: A total of 807 TNBC cases were evaluated for TROP2 immunohistochemical expression. We evaluated the TROP2 H-score distribution in the three cohorts. Tumors were divided into two groups based on TROP2 expression (high vs. low). We analyzed the relationship between clinicopathologic features and markers, including epidermal growth factor receptor, cytokeratin 5/6, p53, and Ki-67, and prognostic significance at high vs. low TROP2 expression. RESULTS: There was no difference in TROP2 H-score distribution between the three cohorts. Moderate-to-strong membranous expression of TROP2 in at least 10% of tumor cells was present in 662 cases (82.0%) in Cohort 1, 59 cases (89.4%) in Cohort 2, and 23 cases (88.5%) in Cohort 3. There was no significant difference in clinicopathologic features between high vs. low TROP2 in all cohorts. TROP2 H-score was an independent poor prognostic factor for overall survival in Cohort 3. CONCLUSIONS: TNBC showed similar TROP2 expression regardless of neoadjuvant treatment or primary tumor/metastasis. Although the prognostic significance of TROP2 expression in metastatic TNBC has been revealed, further evaluation of the predictive value of TROP2 expression for targeted therapy is needed.


Subject(s)
Antigens, Neoplasm , Cell Adhesion Molecules , Triple Negative Breast Neoplasms , Antigens, Neoplasm/metabolism , Antigens, Surface , Biomarkers, Tumor , Cell Adhesion Molecules/metabolism , ErbB Receptors/metabolism , Humans , Keratin-5/metabolism , Ki-67 Antigen/metabolism , Prognosis , Triple Negative Breast Neoplasms/metabolism , Trophoblasts/pathology , Tumor Suppressor Protein p53/metabolism
13.
Ann Diagn Pathol ; 60: 152011, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35905533

ABSTRACT

BACKGROUND: Oncocytic myoepithelial carcinoma ex pleomorphic adenoma neoplastic is a rare neoplastic event and may not display overt malignant radiological features. METHODS: Using routine histopathology and immunohistochemistry, we characterize a case of low-grade oncocytic carcinoma ex pleomorphic adenoma. RESULTS: The tumor arose in the left parotid gland in a 59 year old female. Computed tomography (CT) imaging demonstrated a well-defined, lobulated, enhancing lesion with relative central stellate hypoenhancement. Histologically, the tumor displayed a multi-nodular, non-destructive, invasive pattern, low mitotic activity (one mitotic figure per 10 high power fields) and a small remnant focus of pleomorphic adenoma. The neoplastic cells showed significant expression of cytokeratin 5/6, S-100 protein, smooth muscle actin and p63. CONCLUSION: Low-grade oncocytic carcinoma ex pleomorphic adenoma is a challenging histopathological diagnosis which can be established with use of immunohistochemistry, generous tumor sampling and recognition of the multi-nodular, non-destructive, pattern of invasion. In the absence of clear-cut tumor encroachment into external structures, its malignant nature may not be easily identified on pre-operative imaging.


Subject(s)
Adenocarcinoma , Adenoma, Pleomorphic , Parotid Neoplasms , Salivary Gland Neoplasms , Actins/metabolism , Adenocarcinoma/pathology , Adenoma, Pleomorphic/pathology , Female , Humans , Keratin-5/metabolism , Middle Aged , Parotid Neoplasms/diagnosis , S100 Proteins , Salivary Gland Neoplasms/diagnosis , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/surgery
14.
J Cell Mol Med ; 25(16): 7890-7900, 2021 08.
Article in English | MEDLINE | ID: mdl-34184816

ABSTRACT

Cytokeratin 5 is a marker of basal molecular subtypes of muscle-invasive bladder cancer (MIBC), which correlates with worse overall survival compared to luminal subtypes. Our observations have not confirmed CK5 as a marker of high-grade (HG) disease in Ta non-muscle-invasive bladder cancer (NMIBC). Therefore, to understand the basal-luminal immunohistochemistry profile in Ta NMIBC, we performed immunohistochemistry for CK5, P40, P63 (basal), GATA3 and CK20 (luminal) and studied the correlation with HG and clinical outcome in 109 patients with Ta NMIBC. HG and low-grade (LG) diseases were scored in each patient. Four different CK5 patterns were evaluated: absent (median 41.3%), normal (72.5%), rising (84.4%) and full thickness (23.9%). The median percentage of GATA3 was 100%. HG disease and CK5 expression and rising CK5 pattern had a significant inverse correlation, whereas HG disease and CK20 expression had a significant positive correlation. We also found a significant inverse correlation between CK5 expression and CK20 expression. Quantitative PCR confirmed that the presence of CK5 correlated with up-regulation of CK5 RNA. None of the markers could differentiate patients with regard to clinical outcome. Our results suggest a role for CK5 and CK20 in differentiating between LG and HG disease in Ta NMIBC.


Subject(s)
Biomarkers, Tumor/metabolism , Keratin-5/metabolism , Urinary Bladder Neoplasms/pathology , Aged , Female , Humans , Immunohistochemistry/methods , Keratin-20/metabolism , Male , Middle Aged , Neoplasm Grading , Prognosis , Retrospective Studies , Urinary Bladder Neoplasms/metabolism
15.
Am J Pathol ; 190(8): 1752-1762, 2020 08.
Article in English | MEDLINE | ID: mdl-32339497

ABSTRACT

The biological processes of urothelial carcinogenesis are not fully understood, particularly regarding the relationship between specific genetic events, cell of origin, and molecular subtypes of subsequent tumors. N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-induced mouse bladder cancer is widely accepted as a useful model that recapitulates the pathway of human bladder tumorigenesis from dysplasia to invasive cancer via carcinoma in situ. However, the long and variable time of tumorigenesis often hinders efficient preclinical or translational research. We hypothesized that Trp53 mutation in specific types of urothelial cells facilitates efficient development of clinically relevant bladder cancer. Using lineage tracing, we showed that Trp53 mutation in Krt5-expressing cells resulted in more efficient tumorigenesis of mouse muscle-invasive bladder cancer (MIBC) with squamous differentiation compared with Trp53 mutation in Upk2-expressing cells, or wild-type or hemizygous Trp53 in the entire urothelium. Mouse MIBC that developed at 24 weeks of BBN treatment showed morphologic and genetic similarities to the basal squamous subtypes of human MIBC, irrespective of pre-induction of Trp53 mutation or whether the cell of origin was Krt5- or Upk2-expressing cells. Our findings suggest that intermediate cells as well as basal cells also can give rise to basal-like MIBC, with pre-induction of Trp53 mutation accelerating MIBC. Thus, in BBN chemical carcinogenesis, pre-induction of Trp53 mutation in basal cells facilitates efficient modeling of the basal squamous subtype of human MIBC.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Transitional Cell/genetics , Keratin-5/genetics , Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder/pathology , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Keratin-5/metabolism , Mice , Mutation , Urinary Bladder/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
16.
Exp Dermatol ; 30(3): 337-346, 2021 03.
Article in English | MEDLINE | ID: mdl-33135264

ABSTRACT

Hand-foot skin reaction (HFSR) is a common side effect caused by several tyrosine kinase inhibitors, including sunitinib. However, the nature of the cornifying factors related to the molecular biological mechanisms underlying HFSR remains poorly understood. We used human keratinocyte models to investigate the key cornifying factors for dermatological and biological abnormalities induced by sunitinib. On the basis of the results of microarray analysis using the three-dimensional (3D) human epidermal model, keratin (KRT)6A, serine protease inhibitor (SERPIN)B1, KRT5, and SERPIN Kazal-type 6 were selected as candidate genes related to HFSR. Sunitinib treatment significantly decreased the expression of SERPINB1 and KRT6A in the immunohistochemical staining of the 3D epidermal model. In PSVK1 cells, but not in normal human epidermal keratinocyte cells, both of which are human normal keratinocyte cell lines, sunitinib decreased the expression of KRT6A with a concomitant decrease in levels of phosphorylated extracellular signal-regulated kinases (ERK)1/2 and phosphorylated p38 mitogen-activated protein kinase (MAPK). Inhibitors of the ERK and p38 MAPK signal pathways also significantly decreased KRT6A expression. Sunitinib-induced decrease in KRT6A expression was suppressed by the inhibition of glycogen synthase kinase-3ß by enhancing ERK1/2 and p38 MAPK phosphorylation. Thus, sunitinib reduces the expression of KRT6A and SERPINB1 by inhibiting the ERK1/2 and p38 MAPK signalling pathways in the skin model. These changes in expression contribute to the pathology of HFSR.


Subject(s)
Antineoplastic Agents/pharmacology , Epidermis/metabolism , Keratin-6/metabolism , Serpins/metabolism , Sunitinib/pharmacology , Cell Line , Gene Expression/drug effects , Humans , Indoles/pharmacology , Keratin-5/metabolism , Keratin-6/genetics , MAP Kinase Signaling System/drug effects , Maleimides/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Serine Peptidase Inhibitors, Kazal Type/metabolism , Serpins/genetics
17.
J Cutan Pathol ; 48(7): 902-907, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33258154

ABSTRACT

Malignant mesothelioma is a locally aggressive malignancy most commonly arising from the pleural and/or peritoneal cavity. Distant cutaneous metastasis is extremely rare. Here, we describe two cases of mesothelioma metastatic to the head and neck skin. Case 1: A 64-year-old man diagnosed previously with extensive thoracic and abdominal mesothelioma, developed a rapidly growing right upper lip lesion, for which a wedge resection was performed. Case 2: A 77-year-old woman with a history of pleural mesothelioma developed a firm, mobile subcutaneous nodule on the right lateral forehead, clinically thought to represent either an epidermal inclusion cyst or a lipoma. A punch biopsy was performed. In both cases, histopathologic evaluation revealed dermal proliferation of epithelioid cells with moderate cytologic atypia and three mitotic figures per mm2 and two mitotic figures per mm2 for Cases 1 and 2, respectively. Immunohistochemical studies revealed the lesional cells to be positive for WT1, mesothelin, D2-40, CK5/6, while being negative for melanocytic and other keratinocytic markers, supporting a diagnosis of metastatic mesothelioma. Awareness of rare instances of cutaneous metastases from malignant mesothelioma is necessary to avoid possible misdiagnosis and ensure appropriate management.


Subject(s)
Mesothelioma, Malignant/diagnosis , Mesothelioma, Malignant/therapy , Pleural Neoplasms/pathology , Skin Neoplasms/secondary , Aged , Antibodies, Monoclonal, Murine-Derived/metabolism , Biopsy, Needle/methods , Chemoradiotherapy/methods , Fatal Outcome , Female , GPI-Linked Proteins/metabolism , Humans , Immunohistochemistry/methods , Keratin-5/metabolism , Keratin-6/metabolism , Male , Mesothelin , Mesothelioma, Malignant/secondary , Middle Aged , Neoplasm Metastasis/pathology , Skin Neoplasms/surgery , WT1 Proteins/metabolism
18.
Nature ; 517(7536): 616-20, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25383540

ABSTRACT

Lung diseases such as chronic obstructive pulmonary disease and pulmonary fibrosis involve the progressive and inexorable destruction of oxygen exchange surfaces and airways, and have emerged as a leading cause of death worldwide. Mitigating therapies, aside from impractical organ transplantation, remain limited and the possibility of regenerative medicine has lacked empirical support. However, it is clinically known that patients who survive sudden, massive loss of lung tissue from necrotizing pneumonia or acute respiratory distress syndrome often recover full pulmonary function within six months. Correspondingly, we recently demonstrated lung regeneration in mice following H1N1 influenza virus infection, and linked distal airway stem cells expressing Trp63 (p63) and keratin 5, called DASC(p63/Krt5), to this process. Here we show that pre-existing, intrinsically committed DASC(p63/Krt5) undergo a proliferative expansion in response to influenza-induced lung damage, and assemble into nascent alveoli at sites of interstitial lung inflammation. We also show that the selective ablation of DASC(p63/Krt5) in vivo prevents this regeneration, leading to pre-fibrotic lesions and deficient oxygen exchange. Finally, we demonstrate that single DASC(p63/Krt5)-derived pedigrees differentiate to type I and type II pneumocytes as well as bronchiolar secretory cells following transplantation to infected lung and also minimize the structural consequences of endogenous stem cell loss on this process. The ability to propagate these cells in culture while maintaining their intrinsic lineage commitment suggests their potential in stem cell-based therapies for acute and chronic lung diseases.


Subject(s)
Keratin-5/metabolism , Lung/cytology , Lung/physiology , Phosphoproteins/metabolism , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Bronchioles/cytology , Bronchioles/virology , Cell Differentiation , Cell Lineage , Cell Proliferation , Dogs , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Oxygen/metabolism , Pedigree , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/virology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Re-Epithelialization , Stem Cell Transplantation
19.
Nature ; 517(7536): 621-5, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25533958

ABSTRACT

Broadly, tissue regeneration is achieved in two ways: by proliferation of common differentiated cells and/or by deployment of specialized stem/progenitor cells. Which of these pathways applies is both organ- and injury-specific. Current models in the lung posit that epithelial repair can be attributed to cells expressing mature lineage markers. By contrast, here we define the regenerative role of previously uncharacterized, rare lineage-negative epithelial stem/progenitor (LNEP) cells present within normal distal lung. Quiescent LNEPs activate a ΔNp63 (a p63 splice variant) and cytokeratin 5 remodelling program after influenza or bleomycin injury in mice. Activated cells proliferate and migrate widely to occupy heavily injured areas depleted of mature lineages, at which point they differentiate towards mature epithelium. Lineage tracing revealed scant contribution of pre-existing mature epithelial cells in such repair, whereas orthotopic transplantation of LNEPs, isolated by a definitive surface profile identified through single-cell sequencing, directly demonstrated the proliferative capacity and multipotency of this population. LNEPs require Notch signalling to activate the ΔNp63 and cytokeratin 5 program, and subsequent Notch blockade promotes an alveolar cell fate. Persistent Notch signalling after injury led to parenchymal 'micro-honeycombing' (alveolar cysts), indicative of failed regeneration. Lungs from patients with fibrosis show analogous honeycomb cysts with evidence of hyperactive Notch signalling. Our findings indicate that distinct stem/progenitor cell pools repopulate injured tissue depending on the extent of the injury, and the outcomes of regeneration or fibrosis may depend in part on the dynamics of LNEP Notch signalling.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/pathology , Lung Injury/pathology , Lung/cytology , Lung/pathology , Re-Epithelialization , Stem Cells/cytology , Animals , Bleomycin , Cell Lineage , Cell Proliferation , Cell Separation , Cysts/metabolism , Cysts/pathology , Epithelial Cells/metabolism , Female , Humans , Keratin-5/metabolism , Lung/physiology , Lung Injury/chemically induced , Lung Injury/virology , Male , Mice , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Stem Cell Transplantation , Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
20.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830328

ABSTRACT

Epidermolysis bullosa simplex (EBS) is a group of inherited keratinopathies that, in most cases, arise due to mutations in keratins and lead to intraepidermal ruptures. The cellular pathology of most EBS subtypes is associated with the fragility of the intermediate filament network, cytolysis of the basal layer of the epidermis, or attenuation of hemidesmosomal/desmosomal components. Mutations in keratins 5/14 or in other genes that encode associated proteins induce structural disarrangements of different strengths depending on their locations in the genes. Keratin aggregates display impaired dynamics of assembly and diminished solubility and appear to be the trigger for endoplasmic reticulum (ER) stress upon being phosphorylated by MAPKs. Global changes in cellular signaling mainly occur in cases of severe dominant EBS mutations. The spectrum of changes initiated by phosphorylation includes the inhibition of proteasome degradation, TNF-α signaling activation, deregulated proliferation, abnormal cell migration, and impaired adherence of keratinocytes. ER stress also leads to the release of proinflammatory danger-associated molecular pattern (DAMP) molecules, which enhance avalanche-like inflammation. Many instances of positive feedback in the course of cellular stress and the development of sterile inflammation led to systemic chronic inflammation in EBS. This highlights the role of keratin in the maintenance of epidermal and immune homeostasis.


Subject(s)
Alarmins/genetics , Epidermis/metabolism , Epidermolysis Bullosa Simplex/genetics , Keratin-14/genetics , Keratin-5/genetics , Keratinocytes/metabolism , Alarmins/metabolism , Endoplasmic Reticulum Stress/genetics , Epidermis/pathology , Epidermolysis Bullosa Simplex/metabolism , Epidermolysis Bullosa Simplex/pathology , Gene Expression Regulation , Humans , Inflammation , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Intermediate Filaments/ultrastructure , Keratin-14/metabolism , Keratin-5/metabolism , Keratinocytes/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Aggregates , Proteolysis , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL