1
|
Stewart S, Stankunas K. Section Immunostaining for Protein Expression and Cell Proliferation Studies of Regenerating Fins. Methods Mol Biol 2024; 2707:235-254. [PMID: 37668917 DOI: 10.1007/978-1-0716-3401-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Adult zebrafish fins fully regenerate after resection, providing a highly accessible and remarkable vertebrate model of organ regeneration. Fin injury triggers wound epidermis formation and the dedifferentiation of injury-adjacent mature cells to establish an organized blastema of progenitor cells. Balanced cell proliferation and redifferentiation along with cell movements then progressively reestablish patterned tissues and restore the fin to its original size and shape. A mechanistic understanding of these coordinated cell behaviors and transitions requires direct knowledge of proteins in their physiological context, including expression, subcellular localization, and activity. Antibody-based staining of sectioned fins facilitates such high-resolution analyses of specific, native proteins. Therefore, such methods are mainstays of comprehensive, hypothesis-driven fin regeneration studies. However, section immunostaining requires labor-intensive, empirical optimization. Here, we present detailed, multistep procedures for antibody staining and co-detecting proliferating cells using paraffin and frozen fin sections. We include suggestions to avoid common pitfalls and to streamline the development of optimized, validated protocols for new and challenging antibodies.
Collapse
Affiliation(s)
- Scott Stewart
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA.
| | - Kryn Stankunas
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA.
- Department of Biology, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
2
|
Peralta S, Marcinczyk MM, Katt WP, Duhamel GE. Confirmation of canine acanthomatous ameloblastoma using RAS Q61R immunohistochemical staining of formalin-fixed paraffin-embedded tissues. Front Vet Sci 2023; 10:1281022. [PMID: 37901104 PMCID: PMC10611472 DOI: 10.3389/fvets.2023.1281022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/03/2023] [Indexed: 10/31/2023] Open
Abstract
Differentiating canine acanthomatous ameloblastoma (CAA) from oral squamous cell carcinoma (OSCC) based on routine histopathology can be challenging. We have previously shown that more than 95% of CAAs harbor an HRAS p.Q61R somatic mutation, while OSCCs carry either wild-type alleles or other MAPK pathway activating mutations (e.g., HRAS p.Q61L, BRAF p.V595E). Given that HRAS p.Q61R mutations are highly prevalent in CAA, we hypothesized that a RAS Q61R-specific rabbit monoclonal antibody may be a useful tool for confirmation of CAA by immunohistochemical (IHC) staining. In the present study, we assessed IHC staining of archived formalin-fixed and paraffin-embedded biopsy samples with a diagnosis of CAA (n = 23), using a RAS Q61R-specific rabbit monoclonal antibody (SP174) and an automated IHC stainer. Negative control samples consisted of HRAS p.Q61R mutation-negative OSCC tumors with either a known HRAS p.Q61L mutation (n = 1), BRAF p.V595E mutation (n = 4), or wild-type corresponding alleles (n = 3). We found that all 23 CAAs showed diffuse and strong membranous RAS Q61R immunoreactivity (100% sensitivity), while none of the 8 OSCCs showed immunoreactivity (100% specificity). The data supports the use of RAS Q61R-specific rabbit monoclonal antibody for diagnostic IHC confirmation of CAA and ruling out OSCC in dogs.
Collapse
Affiliation(s)
- Santiago Peralta
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Magdalena M. Marcinczyk
- Department of Biomedical Sciences and New York Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - William P. Katt
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Gerald E. Duhamel
- Department of Biomedical Sciences and New York Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
3
|
Strupp C, Corvaro M, Cohen SM, Corton JC, Ogawa K, Richert L, Jacobs MN. Increased Cell Proliferation as a Key Event in Chemical Carcinogenesis: Application in an Integrated Approach for the Testing and Assessment of Non-Genotoxic Carcinogenesis. Int J Mol Sci 2023; 24:13246. [PMID: 37686053 PMCID: PMC10488128 DOI: 10.3390/ijms241713246] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
In contrast to genotoxic carcinogens, there are currently no internationally agreed upon regulatory tools for identifying non-genotoxic carcinogens of human relevance. The rodent cancer bioassay is only used in certain regulatory sectors and is criticized for its limited predictive power for human cancer risk. Cancer is due to genetic errors occurring in single cells. The risk of cancer is higher when there is an increase in the number of errors per replication (genotoxic agents) or in the number of replications (cell proliferation-inducing agents). The default regulatory approach for genotoxic agents whereby no threshold is set is reasonably conservative. However, non-genotoxic carcinogens cannot be regulated in the same way since increased cell proliferation has a clear threshold. An integrated approach for the testing and assessment (IATA) of non-genotoxic carcinogens is under development at the OECD, considering learnings from the regulatory assessment of data-rich substances such as agrochemicals. The aim is to achieve an endorsed IATA that predicts human cancer better than the rodent cancer bioassay, using methodologies that equally or better protect human health and are superior from the view of animal welfare/efficiency. This paper describes the technical opportunities available to assess cell proliferation as the central gateway of an IATA for non-genotoxic carcinogenicity.
Collapse
Affiliation(s)
| | | | - Samuel M. Cohen
- Department of Pathology and Microbiology and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - J. Christopher Corton
- Center for Computational Toxicology and Exposure, United States Environmental Protection Agency (US EPA), Research Triangle Park, NC 27711, USA;
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki 210-9501, Japan
| | | | - Miriam N. Jacobs
- United Kingdom Health Security Agency (UK HSA), Radiation, Chemicals and Environmental Hazards, Harwell Innovation Campus, Dicot OX11 0RQ, UK
| |
Collapse
|
4
|
Camacho-Gomez D, Sorzabal-Bellido I, Ortiz-de-Solorzano C, Garcia-Aznar JM, Gomez-Benito MJ. A hybrid physics-based and data-driven framework for cellular biological systems: Application to the morphogenesis of organoids. iScience 2023; 26:107164. [PMID: 37485358 PMCID: PMC10359941 DOI: 10.1016/j.isci.2023.107164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/30/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
How cells orchestrate their cellular functions remains a crucial question to unravel how they organize in different patterns. We present a framework based on artificial intelligence to advance the understanding of how cell functions are coordinated spatially and temporally in biological systems. It consists of a hybrid physics-based model that integrates both mechanical interactions and cell functions with a data-driven model that regulates the cellular decision-making process through a deep learning algorithm trained on image data metrics. To illustrate our approach, we used data from 3D cultures of murine pancreatic ductal adenocarcinoma cells (PDAC) grown in Matrigel as tumor organoids. Our approach allowed us to find the underlying principles through which cells activate different cell processes to self-organize in different patterns according to the specific microenvironmental conditions. The framework proposed here expands the tools for simulating biological systems at the cellular level, providing a novel perspective to unravel morphogenetic patterns.
Collapse
Affiliation(s)
- Daniel Camacho-Gomez
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Ioritz Sorzabal-Bellido
- Solid Tumors and Biomarkers Program, IDISNA, and CIBERONC, Center for Applied Medical Research, University of Navarra, Zaragoza, Spain
| | - Carlos Ortiz-de-Solorzano
- Solid Tumors and Biomarkers Program, IDISNA, and CIBERONC, Center for Applied Medical Research, University of Navarra, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
5
|
Li D, Liu Y, Zhan Q, Zeng Y, Peng Z, He Q, Tan Q, Cao W, Wang S, Wang J. Astragaloside IV Blunts Epithelial-Mesenchymal Transition and G2/M Arrest to Alleviate Renal Fibrosis via Regulating ALDH2-Mediated Autophagy. Cells 2023; 12:1777. [PMID: 37443810 PMCID: PMC10340704 DOI: 10.3390/cells12131777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Previous studies show that astragaloside IV (ASIV) has anti-renal fibrosis effects. However, its mechanism remains elusive. In this study, we investigated the anti-fibrosis mechanisms of ASIV on chronic kidney disease (CKD) in vivo and in vitro. A CKD model was induced in rats with adenine (200 mg/kg/d, i.g.), and an in vitro renal fibrosis model was induced in human kidney-2 (HK-2) cells treated with TGF-β1. We revealed that ASIV significantly alleviated renal fibrosis by suppressing the expressions of epithelial-mesenchymal transition (EMT)-related proteins, including fibronectin, vimentin, and alpha-smooth muscle actin (α-SMA), and G2/M arrest-related proteins, including phosphorylated p53 (p-p53), p21, phosphorylated histone H3 (p-H3), and Ki67 in both of the in vivo and in vitro models. Transcriptomic analysis and subsequent validation showed that ASIV rescued ALDH2 expression and inhibited AKT/mTOR-mediated autophagy. Furthermore, in ALDH2-knockdown HK-2 cells, ASIV failed to inhibit AKT/mTOR-mediated autophagy and could not blunt EMT and G2/M arrest. In addition, we further demonstrated that rapamycin, an autophagy inducer, reversed the treatment of ASIV by promoting autophagy in TGF-β1-treated HK-2 cells. A dual-luciferase report assay indicated that ASIV enhanced the transcriptional activity of the ALDH2 promoter. In addition, a further molecular docking analysis showed the potential interaction of ALDH2 and ASIV. Collectively, our data indicate that ALDH2-mediated autophagy may be a novel target in treating renal fibrosis in CKD models, and ASIV may be an effective targeted drug for ALDH2, which illuminate a new insight into the treatment of renal fibrosis and provide new evidence of pharmacology to elucidate the anti-fibrosis mechanism of ASIV in treating renal fibrosis.
Collapse
Affiliation(s)
- Dong Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuzhe Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Quancao Zhan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yan Zeng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Ze Peng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qifeng He
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Qi Tan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Wenfu Cao
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
6
|
Cho YS, Gwak SJ. Novel Sensing Technique for Stem Cells Differentiation Using Dielectric Spectroscopy of Their Proteins. SENSORS (BASEL, SWITZERLAND) 2023; 23:2397. [PMID: 36904601 PMCID: PMC10007102 DOI: 10.3390/s23052397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Dielectric spectroscopy (DS) is the primary technique to observe the dielectric properties of biomaterials. DS extracts complex permittivity spectra from measured frequency responses such as the scattering parameters or impedances of materials over the frequency band of interest. In this study, an open-ended coaxial probe and vector network analyzer were used to characterize the complex permittivity spectra of protein suspensions of human mesenchymal stem cells (hMSCs) and human osteogenic sarcoma (Saos-2) cells in distilled water at frequencies ranging from 10 MHz to 43.5 GHz. The complex permittivity spectra of the protein suspensions of hMSCs and Saos-2 cells revealed two major dielectric dispersions, β and γ, offering three distinctive features for detecting the differentiation of stem cells: the distinctive values in the real and imaginary parts of the complex permittivity spectra as well as the relaxation frequency in the β-dispersion. The protein suspensions were analyzed using a single-shell model, and a dielectrophoresis (DEP) study was performed to determine the relationship between DS and DEP. In immunohistochemistry, antigen-antibody reactions and staining are required to identify the cell type; in contrast, DS eliminates the use of biological processes, while also providing numerical values of the dielectric permittivity of the material-under-test to detect differences. This study suggests that the application of DS can be expanded to detect stem cell differentiation.
Collapse
Affiliation(s)
- Young Seek Cho
- Department of Electronic Engineering, Wonkwang University, Iksan 54538, Jeollabuk-do, Republic of Korea
| | - So-Jung Gwak
- Department of Chemical Engineering, Wonkwang University, Iksan 54538, Jeollabuk-do, Republic of Korea
| |
Collapse
|
7
|
Gasperini C, Tuntevski K, Beatini S, Pelizzoli R, Lo Van A, Mangoni D, Cossu RM, Pascarella G, Bianchini P, Bielefeld P, Scarpato M, Pons‐Espinal M, Sanges R, Diaspro A, Fitzsimons CP, Carninci P, Gustincich S, De Pietri Tonelli D. Piwil2 (Mili) sustains neurogenesis and prevents cellular senescence in the postnatal hippocampus. EMBO Rep 2023; 24:e53801. [PMID: 36472244 PMCID: PMC9900342 DOI: 10.15252/embr.202153801] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Adult neural progenitor cells (aNPCs) ensure lifelong neurogenesis in the mammalian hippocampus. Proper regulation of aNPC fate has thus important implications for brain plasticity and healthy aging. Piwi proteins and the small noncoding RNAs interacting with them (piRNAs) have been proposed to control memory and anxiety, but the mechanism remains elusive. Here, we show that Piwil2 (Mili) is essential for proper neurogenesis in the postnatal mouse hippocampus. RNA sequencing of aNPCs and their differentiated progeny reveal that Mili and piRNAs are dynamically expressed in neurogenesis. Depletion of Mili and piRNAs in the adult hippocampus impairs aNPC differentiation toward a neural fate, induces senescence, and generates reactive glia. Transcripts modulated upon Mili depletion bear sequences complementary or homologous to piRNAs and include repetitive elements and mRNAs encoding essential proteins for proper neurogenesis. Our results provide evidence of a critical role for Mili in maintaining fitness and proper fate of aNPCs, underpinning a possible involvement of the piRNA pathway in brain plasticity and successful aging.
Collapse
Affiliation(s)
- Caterina Gasperini
- Neurobiology of miRNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
| | - Kiril Tuntevski
- Neurobiology of miRNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT)GenoaItaly
| | - Silvia Beatini
- Neurobiology of miRNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
| | - Roberta Pelizzoli
- Neurobiology of miRNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
| | - Amanda Lo Van
- Neurobiology of miRNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
| | - Damiano Mangoni
- Central RNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
| | - Rosa M Cossu
- Central RNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
| | - Giovanni Pascarella
- Division of Genomic TechnologiesRIKEN Center for Life Science TechnologiesYokohamaJapan
| | - Paolo Bianchini
- Nanoscopy, CHT ErzelliIstituto Italiano di TecnologiaGenoaItaly
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of ScienceUniversity of AmsterdamAmsterdamThe Netherlands
| | | | | | - Remo Sanges
- Central RNA LaboratoryIstituto Italiano di TecnologiaGenoaItaly
- Area of NeuroscienceSISSATriesteItaly
| | - Alberto Diaspro
- Nanoscopy, CHT ErzelliIstituto Italiano di TecnologiaGenoaItaly
| | - Carlos P Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of ScienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Piero Carninci
- Division of Genomic TechnologiesRIKEN Center for Life Science TechnologiesYokohamaJapan
- Human TechnopoleMilanItaly
| | | | | |
Collapse
|
8
|
Immunohistochemical Evaluation of Periodontal Regeneration Using a Porous Collagen Scaffold. Int J Mol Sci 2021; 22:ijms222010915. [PMID: 34681574 PMCID: PMC8535773 DOI: 10.3390/ijms222010915] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/03/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
(1) Aim: To immunohistochemically evaluate the effect of a volume-stable collagen scaffold (VCMX) on periodontal regeneration. (2) Methods: In eight beagle dogs, acute two-wall intrabony defects were treated with open flap debridement either with VCMX (test) or without (control). After 12 weeks, eight defects out of four animals were processed for paraffin histology and immunohistochemistry. (3) Results: All defects (four test + four control) revealed periodontal regeneration with cementum and bone formation. VCMX remnants were integrated in bone, periodontal ligament (PDL), and cementum. No differences in immunohistochemical labeling patterns were observed between test and control sites. New bone and cementum were labeled for bone sialoprotein, while the regenerated PDL was labeled for periostin and collagen type 1. Cytokeratin-positive epithelial cell rests of Malassez were detected in 50% of the defects. The regenerated PDL demonstrated a larger blood vessel area at the test (14.48% ± 3.52%) than at control sites (8.04% ± 1.85%, p = 0.0007). The number of blood vessels was higher in the regenerated PDL (test + control) compared to the pristine one (p = 0.012). The cell proliferative index was not statistically significantly different in pristine and regenerated PDL. (4) Conclusions: The data suggest a positive effect of VCMX on angiogenesis and an equally high cell turnover in the regenerated and pristine PDL. This VCMX supported periodontal regeneration in intrabony defects.
Collapse
|
9
|
Gopal A, Herr AE. Segmentation-based analysis of single-cell immunoblots. Electrophoresis 2021; 42:2070-2080. [PMID: 34357587 PMCID: PMC8526408 DOI: 10.1002/elps.202100144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/06/2021] [Accepted: 07/27/2021] [Indexed: 11/11/2022]
Abstract
From genomics to transcriptomics to proteomics, microfluidic tools underpin recent advances in single-cell biology. Detection of specific proteoforms-with single-cell resolution-presents challenges in detection specificity and sensitivity. Miniaturization of protein immunoblots to single-cell resolution mitigates these challenges. For example, in microfluidic western blotting, protein targets are separated by electrophoresis and subsequently detected using fluorescently labeled antibody probes. To quantify the expression level of each protein target, the fluorescent protein bands are fit to Gaussians; yet, this method is difficult to use with noisy, low-abundance, or low-SNR protein bands, and with significant band skew or dispersion. In this study, we investigate segmentation-based approaches to robustly quantify protein bands from single-cell protein immunoblots. As compared to a Gaussian fitting pipeline, the segmentation pipeline detects >1.5× more protein bands for downstream quantification as well as more of the low-abundance protein bands (i.e., with SNR ∼3). Utilizing deep learning-based segmentation approaches increases the recovery of low-SNR protein bands by an additional 50%. However, we find that segmentation-based approaches are less robust at quantifying poorly resolved protein bands (separation resolution, Rs < 0.6). With burgeoning needs for more single-cell protein analysis tools, we see microfluidic separations as benefitting substantially from segmentation-based analysis approaches.
Collapse
Affiliation(s)
- Anjali Gopal
- Department of Bioengineering, University of California, Berkeley, CA, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA, USA
| | - Amy E. Herr
- Department of Bioengineering, University of California, Berkeley, CA, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA, USA
- Chan Zuckerberg BioHub, San Francisco, CA, USA
| |
Collapse
|
10
|
de Greeff A, Schokker D, Roubos-van den Hil P, Ramaekers P, Vastenhouw SA, Harders F, Bossers A, Smits MA, Rebel JMJ. The effect of maternal antibiotic use in sows on intestinal development in offspring. J Anim Sci 2020; 98:5849914. [PMID: 32479635 PMCID: PMC7295330 DOI: 10.1093/jas/skaa181] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
The objective of this study is to investigate the effect of a maternal antibiotic administration during the last week of gestation on the early life intestinal development in neonatal piglets. Colonization of the gut with bacteria starts during birth and plays a major role in the intestinal and immunological development of the intestine. We demonstrate that maternal interventions induced changes in the sows (n = 6 to 8 per treatment) fecal microbiota diversity around birth (P < 0.001, day 1). Whole-genome microarray analysis in small intestinal samples of 1-d old piglets (n = 6 to 8 per treatment) showed significantly expressed genes (Padj < 0.05) which were involved in processes of tight junction formation and immunoglobulin production. Furthermore, when performing morphometry analysis, the number of goblet cells in jejunum was significantly (P < 0.001) lower in piglets from amoxicillin administered sows compared with the respective control piglets. Both significantly expressed genes (Padj < 0.05) and significant morphometry data (jejunum P < 0.05 and ileum P < 0.01) indicate that the crypts of piglets from amoxicillin administered sows deepen around weaning (day 26) as an effect of the amoxicillin administration in sows. The latter might imply that the intestinal development of piglets was delayed by maternal antibiotic administration. Taken together, these results show that maternally oral antibiotic administration changes in early life can affect intestinal development of the offspring piglets for a period of at least 5 wk after the maternal antibiotic administration was finished. These results show that modulation of the neonatal intestine is possible by maternal interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Frank Harders
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Alex Bossers
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Mari A Smits
- Wageningen Bioveterinary Research, Lelystad, The Netherlands.,Wageningen Livestock Research, Wageningen, The Netherlands
| | | |
Collapse
|
11
|
Jensen VFH, Brinck PR, Nowak J, Thorup I, Sjögren I, Mølck AM. Evaluation of Cell Proliferation in Rat Mammary Glands Is Not Predictive of the Carcinogenic Potential of Insulin In Vivo. Int J Toxicol 2020; 39:560-576. [PMID: 32723118 DOI: 10.1177/1091581820941776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For nonclinical safety-assessment of insulin analogues in vivo, mitogenic effects are compared to that of human insulin. Besides histopathologic evaluation, this usually includes assessment of cell proliferation (CP) in mammary glands. Insulin analogue X10 is recommended as positive control, due to its known carcinogenic effect in rat mammary glands. Here, we discuss the mitogenic effect of insulin in vivo and use of X10 as positive control. We present results from 4 nonclinical rat studies evaluating effects of repeated dosing with insulin detemir (≤26 weeks) or degludec (52 weeks) in mammary glands. Studies included human insulin-dosed groups as comparators, CP, and histopathologic evaluation. One study included an X10-dosed group (26 weeks), another ≤3 weeks of dosing with X10 or human insulin evaluating effects of these comparators. Neither human insulin, insulin detemir, degludec, nor X10 induced mammary tumors or increased CP in the studies. The CP marker proliferating cell nuclear antigen varied within/between studies and was not correlated with the remaining markers or CP fluctuations during estrous cycle, whereas the other CP markers, Ki-67 and 5-bromo-2'-deoxyuridine (BrdU), correlated with estrous cycle changes and each other. In conclusion, we propose that the mitogenic effect of insulin in rat mammary glands is weak in vivo. Cell proliferation evaluation in nonclinical safety assessment studies is not predictive of the carcinogenic potential of insulin, thus, the value of including this end point is debatable. Moreover, X10 is not recommended as positive control, due to lack of proliferative effects. Typical CP markers vary greatly in quality, BrdU seemingly most reliable.
Collapse
Affiliation(s)
| | - Peter R Brinck
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Jette Nowak
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Inger Thorup
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Ingrid Sjögren
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| | - Anne-Marie Mølck
- Department of Safety Sciences & Imaging, 1450Novo Nordisk A/S, Maaloev, Denmark
| |
Collapse
|
12
|
Liu L, Zhang P, Bai M, He L, Zhang L, Liu T, Yang Z, Duan M, Liu M, Liu B, Du R, Qian Q, Sun S. p53 upregulated by HIF-1α promotes hypoxia-induced G2/M arrest and renal fibrosis in vitro and in vivo. J Mol Cell Biol 2020; 11:371-382. [PMID: 30032308 PMCID: PMC7727266 DOI: 10.1093/jmcb/mjy042] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/21/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
Hypoxia plays an important role in the genesis and progression of renal fibrosis. The underlying mechanisms, however, have not been sufficiently elucidated. We examined the role of p53 in hypoxia-induced renal fibrosis in cell culture (human and rat renal tubular epithelial cells) and a mouse unilateral ureteral obstruction (UUO) model. Cell cycle of tubular cells was determined by flow cytometry, and the expression of profibrogenic factors was determined by RT-PCR, immunohistochemistry, and western blotting. Chromatin immunoprecipitation and luciferase reporter experiments were performed to explore the effect of HIF-1α on p53 expression. We showed that, in hypoxic tubular cells, p53 upregulation suppressed the expression of CDK1 and cyclins B1 and D1, leading to cell cycle (G2/M) arrest (or delay) and higher expression of TGF-β, CTGF, collagens, and fibronectin. p53 suppression by siRNA or by a specific p53 inhibitor (PIF-α) triggered opposite effects preventing the G2/M arrest and profibrotic changes. In vivo experiments in the UUO model revealed similar antifibrotic results following intraperitoneal administration of PIF-α (2.2 mg/kg). Using gain-of-function, loss-of-function, and luciferase assays, we further identified an HRE3 region on the p53 promoter as the HIF-1α-binding site. The HIF-1α–HRE3 binding resulted in a sharp transcriptional activation of p53. Collectively, we show the presence of a hypoxia-activated, p53-responsive profibrogenic pathway in the kidney. During hypoxia, p53 upregulation induced by HIF-1α suppresses cell cycle progression, leading to the accumulation of G2/M cells, and activates profibrotic TGF-β and CTGF-mediated signaling pathways, causing extracellular matrix production and renal fibrosis.
Collapse
Affiliation(s)
- Limin Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Peng Zhang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ming Bai
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lijie He
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Ting Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Zhen Yang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Menglu Duan
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Minna Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Baojian Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Rui Du
- Department of Radiation Oncology, Navy General Hospital, Beijing, China
| | - Qi Qian
- Department of Medicine, Mayo Clinic College of Medicine and Mayo Graduate School, Rochester, MN, USA
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
13
|
CD9 induces cellular senescence and aggravates atherosclerotic plaque formation. Cell Death Differ 2020; 27:2681-2696. [PMID: 32346137 DOI: 10.1038/s41418-020-0537-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/26/2022] Open
Abstract
CD9, a 24 kDa tetraspanin membrane protein, is known to regulate cell adhesion and migration, cancer progression and metastasis, immune and allergic responses, and viral infection. CD9 is upregulated in senescent endothelial cells, neointima hyperplasia, and atherosclerotic plaques. However, its role in cellular senescence and atherosclerosis remains undefined. We investigated the potential mechanism for CD9-mediated cellular senescence and its role in atherosclerotic plaque formation. CD9 knockdown in senescent human umbilical vein endothelial cells significantly rescued senescence phenotypes, while CD9 upregulation in young cells accelerated senescence. CD9 regulated cellular senescence through a phosphatidylinositide 3 kinase-AKT-mTOR-p53 signal pathway. CD9 expression increased in arterial tissues from humans and rats with age, and in atherosclerotic plaques in humans and mice. Anti-mouse CD9 antibody noticeably prevented the formation of atherosclerotic lesions in ApoE-/- mice and Ldlr-/- mice. Furthermore, CD9 ablation in ApoE-/- mice decreased atherosclerotic lesions in aorta and aortic sinus. These results suggest that CD9 plays critical roles in endothelial cell senescence and consequently the pathogenesis of atherosclerosis, implying that CD9 is a novel target for prevention and treatment of vascular aging and atherosclerosis.
Collapse
|
14
|
Phosphorylated STAT3 expression linked to SOCS3 methylation is associated with proliferative ability of gastric mucosa in patients with early gastric cancer. Oncol Lett 2020; 19:3542-3550. [PMID: 32269628 PMCID: PMC7115067 DOI: 10.3892/ol.2020.11462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Gastric cancers (GCs) may develop in the gastric mucosa after elimination of Helicobacter pylori (H. pylori) using eradication therapy. Cytokine signaling is a key mechanism underlying GC development and progression, and STAT3 signaling may serve a central role in gastritis-associated tumorigenesis. In the present study, suppressor of cytokine signaling 3 (SOCS3) methylation was examined, as an activator of phosphorylated (p-)STAT3 expression in the non-neoplastic gastric mucosa (non-NGM) of patients with early GC. The methylation status of the SOCS3 gene promoter was analyzed using methylation-specific PCR in the non-NGM of patients with or without early GC. Expression levels of p-STAT3 and Ki67 were investigated immunohistochemically in non-NGM with early GC before and after H. pylori eradication. In non-NGM, SOCS3 promoter methylation was detected in 17/51 patients (33.3%) with early GC. In those patients, the non-NGM labeling indices of both Ki67 and p-STAT3 were significantly higher compared with that in patients with early GC without SOCS3 methylation. A significant correlation between Ki67 and p-STAT3 expression levels was demonstrated in the non-NGM of patients with early GC. In patients with early GC without SOCS3 methylation, the labeling indices of both Ki67 and p-STAT3 in non-NGM were significantly reduced after H. pylori eradication, whereas no such change was observed in patients with early GC with SOCS3 methylation. SOCS3 methylation is associated with continuous p-STAT3 overexpression and enhanced epithelial cell proliferation in non-NGM of patients with early GC.
Collapse
|
15
|
Serfecz J, Bazick H, Al Salihi MO, Turner P, Fields C, Cruz P, Renne R, Notterpek L. Downregulation of the human peripheral myelin protein 22 gene by miR-29a in cellular models of Charcot-Marie-Tooth disease. Gene Ther 2019; 26:455-464. [PMID: 31455873 PMCID: PMC6920087 DOI: 10.1038/s41434-019-0098-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022]
Abstract
The majority of hereditary neuropathies are caused by duplication of the peripheral myelin protein 22 (PMP22) gene. Therefore, mechanisms to suppress the expression of the PMP22 gene have high therapeutic significance. Here we asked whether the human PMP22 gene is a target for regulation by microRNA 29a (miR-29a). Using bioinformatics, we determined that the human PMP22 gene contains the conserved seed sequence of the miR-29a binding site and this regulatory motif is included in the duplicated region in neuropathic patients. Using luciferase reporter assays in HEK293 cells, we demonstrated that transient transfection of a miR-29a mimic is associated with reduction in PMP22 3'UTR reporter activity. Transfecting normal and humanized transgenic neuropathic mouse Schwann cells with a miR-29a expression plasmid effectively lowered both the endogenous mouse and the transgenic human PMP22 transcripts compared with control vector. In dermal fibroblasts derived from neuropathic patients, ectopic expression of miR-29a led to ~50% reduction in PMP22 mRNA, which corresponded to ~20% decrease in PMP22 protein levels. Significantly, miR-29a-mediated reduction in PMP22 mitigated the reduced mitotic capacity of the neuropathic cells. Together, these results support further testing of miR-29a and/or PMP22-targeting siRNAs as therapeutic agents for correcting the aberrant expression of PMP22 in neuropathic patients.
Collapse
Affiliation(s)
- Jacquelyn Serfecz
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Hannah Bazick
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Mohammed Omar Al Salihi
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Peter Turner
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Christopher Fields
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Pedro Cruz
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Rolf Renne
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
- UF Health Cancer Center, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Lucia Notterpek
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
16
|
Gopal A, Herr AE. Multiplexed in-gel microfluidic immunoassays: characterizing protein target loss during reprobing of benzophenone-modified hydrogels. Sci Rep 2019; 9:15389. [PMID: 31659305 PMCID: PMC6817870 DOI: 10.1038/s41598-019-51849-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/27/2019] [Indexed: 12/28/2022] Open
Abstract
From whole tissues to single-cell lysate, heterogeneous immunoassays are widely utilized for analysis of protein targets in complex biospecimens. Recently, benzophenone-functionalized hydrogel scaffolds have been used to immobilize target protein for immunoassay detection with fluorescent antibody probes. In benzophenone-functionalized hydrogels, multiplex target detection occurs via serial rounds of chemical stripping (incubation with sodium-dodecyl-sulfate (SDS) and β-mercaptoethanol at 50-60 °C for ≥1 h), followed by reprobing (interrogation with additional antibody probes). Although benzophenone facilitates covalent immobilization of proteins to the hydrogel, we observe 50% immunoassay signal loss of immobilized protein targets during stripping rounds. Here, we identify and characterize signal loss mechanisms during stripping and reprobing. We posit that loss of immobilized target is responsible for ≥50% of immunoassay signal loss, and that target loss is attributable to disruption of protein immobilization by denaturing detergents (SDS) and incubation at elevated temperatures. Furthermore, our study suggests that protein losses under non-denaturing conditions are more sensitive to protein structure (i.e., hydrodynamic radius), than to molecular mass (size). We formulate design guidance for multiplexed in-gel immunoassays, including that low-abundance proteins be immunoprobed first, even when targets are covalently immobilized to the gel. We also recommend careful scrutiny of the order of proteins targets detected via multiple immunoprobing cycles, based on the protein immobilization buffer composition.
Collapse
Affiliation(s)
- Anjali Gopal
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, United States
- UC Berkeley/UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, California, 94720, United States
| | - Amy E Herr
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, United States.
- UC Berkeley/UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, California, 94720, United States.
- Chan Zuckerberg BioHub, San Francisco, California, 94158, United States.
| |
Collapse
|
17
|
O'Hara RE, Arsenault MG, Esparza Gonzalez BP, Patriquen A, Hartwig S. Three Optimized Methods for In Situ Quantification of Progenitor Cell Proliferation in Embryonic Kidneys Using BrdU, EdU, and PCNA. Can J Kidney Health Dis 2019; 6:2054358119871936. [PMID: 31523438 PMCID: PMC6734617 DOI: 10.1177/2054358119871936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/02/2019] [Indexed: 01/02/2023] Open
Abstract
Background: Nephron progenitor cells derived from the metanephric mesenchyme undergo a complex balance of self-renewal and differentiation throughout kidney development to give rise to the mature nephron. Cell proliferation is an important index of progenitor population dynamics. However, accurate and reproducible in situ quantification of cell proliferation within progenitor populations can be technically difficult to achieve due to the complexity and harsh tissue treatment required of certain protocols. Objective: To optimize and compare the performance of the 3 most accurate S phase–specific labeling methods used for in situ detection and quantification of nephron progenitor and ureteric bud cell proliferation in the developing kidney, namely, 5-bromo-2’-deoxyuridine (BrdU), 5-ethynyl-2’-deoxyuridine (EdU), and proliferating cell nuclear antigen (PCNA). Methods: Protocols for BrdU, EdU, and PCNA were optimized for fluorescence labeling on paraformaldehyde-fixed, paraffin-embedded mouse kidney tissue sections, with co-labeling of nephron progenitor cells and ureteric bud with Six2 and E-cadherin antibodies, respectively. Image processing and analysis, including quantification of proliferating cells, were carried out using free ImageJ software. Results: All 3 methods detect similar ratios of nephron progenitor and ureteric bud proliferating cells. The BrdU staining protocol is the lengthiest and most complex protocol to perform, requires tissue denaturation, and is most subject to interexperimental signal variability. In contrast, bound PCNA and EdU protocols are relatively more straightforward, consistently yield clear results, and far more easily lend themselves to co-staining; however, the bound PCNA protocol requires substantive additional postexperimental analysis to distinguish the punctate nuclear PCNA staining pattern characteristic of proliferating cells. Conclusions: All 3 markers exhibit distinct advantages and disadvantages in quantifying cell proliferation in kidney progenitor populations, with EdU and PCNA protocols being favored due to greater technical ease and reproducibility of results associated with these methods.
Collapse
Affiliation(s)
- Rosalie E O'Hara
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Michel G Arsenault
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Blanca P Esparza Gonzalez
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Ashley Patriquen
- Diagnostic Services, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Sunny Hartwig
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| |
Collapse
|
18
|
Pople JE, Bhogal RK, Moore AE, Jenkins G. Changes in epidermal morphology associated with dandruff. Int J Cosmet Sci 2019; 41:357-363. [PMID: 31087801 DOI: 10.1111/ics.12539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Dandruff is a very common scalp condition characterized by flaking and pruritus usually with no visible signs of inflammation, such as redness and erythema. Dandruff is considered a multifactorial condition with both microbial colonization and host factors such as sebum production thought to play a role. There is evidence of changes in epidermal morphology in the scalp skin of dandruff sufferers, with reports of an increase in mean thickness and more nucleated cell layers. The underlying mechanisms driving these morphological changes are currently unclear. The objective of this study was to fully characterize epidermal morphology in dandruff compared to healthy scalp skin and to evaluate potential mechanisms underlying any changes observed. METHODS Scalp skin biopsies were taken from 22 healthy female subjects and 21 dandruff sufferers, from both lesional and non-lesional sites. Samples were processed, sectioned and stained using haematoxylin and eosin (H&E). To fully characterize epidermal morphology, measurements were taken of epidermal thickness, the convolution of the dermal-epidermal junction and the depth of epidermal rete ridges. To analyse changes in epidermal proliferation immunohistochemical staining was performed using Ki67, a well-established marker of cell proliferation, and quantified using image analysis. RESULTS Histochemical analysis of skin sections revealed that in dandruff lesional samples, the epidermis was thicker, had a more convoluted dermal epidermal junction and the rete ridges were elongated, compared to healthy scalp skin. Similar directional changes in epidermal morphology, were observed in non-lesional dandruff samples, albeit to a lesser extent. Image analysis of Ki67 expression in the epidermis revealed dandruff lesional skin contained significantly more Ki67-positive proliferating keratinocytes than healthy controls samples. This suggests dandruff scalp skin epidermal keratinocytes are in a hyper-proliferative state. CONCLUSION There were significant changes in epidermal morphology in dandruff lesional skin compared to healthy scalp skin including increased epidermal thickness, a more convoluted dermal-epidermal junction and elongation of rete ridges. Interestingly, we found there was evidence of an increase in the percentage of epidermal Ki67-positive cells, which has not been reported previously, and demonstrates dandruff is a condition displaying epidermal hyper-proliferation.
Collapse
Affiliation(s)
- J E Pople
- Unilever R&D Colworth, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - R K Bhogal
- Unilever R&D Colworth, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - A E Moore
- Unilever R&D Colworth, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - G Jenkins
- Unilever R&D Colworth, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| |
Collapse
|
19
|
Circulating osteogenic precursor cells: Building bone from blood. EBioMedicine 2018; 39:603-611. [PMID: 30522933 PMCID: PMC6354620 DOI: 10.1016/j.ebiom.2018.11.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022] Open
Abstract
Circulating osteogenic precursor (COP) cells constitute a recently discovered population of circulating progenitor cells with the capacity to form not only bone but other mesenchymal tissues. There is a small, but growing body of literature exploring these cells, but with a great deal of disagreement and contradiction within it. This review explores the origins and biological characterization of these cells, including the identification strategies used to isolate these cells from the peripheral blood. It also examines the available knowledge on the in vitro and in vivo behaviour of these cells, in the areas of plastic adherence, differentiation capacity, proliferation, and cellular homing. We also review the implications for future use of COP cells in clinical practice, particularly in the area of regenerative medicine and the treatment and assessment of musculoskeletal disease. Circulating Osteogenic Precursors are circulating cells with characteristics of bone marrow mesenchymal stem cells. They are able to differentiate into bone, fat, cartilage and muscle, but many other characteristics remain unknown. They are heterogenous, with at least two specific populations present, with displaying different markers and behaviors.
Collapse
|
20
|
Takahashi S, Takebuchi R, Taniwaki H, Domon T. Recovery of atrophic parotid glands in rats fed a liquid diet by switching to a pellet diet. Arch Oral Biol 2018; 96:39-45. [DOI: 10.1016/j.archoralbio.2018.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 11/29/2022]
|
21
|
Jackson A, Dowsett K. Proliferating Cell Nuclear Antigen in the Equine Testis: Effects of Age and Experimental Treatment1. Biol Reprod 2018. [DOI: 10.1093/biolreprod/52.monograph_series1.631] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Anne Jackson
- Departments of Anatomical Sciences The University of Queensland, St. Lucia, Queensland, Australia, 4072
| | - Kerry Dowsett
- Farm Animal Medicine and Production The University of Queensland, St. Lucia, Queensland, Australia, 4072
| |
Collapse
|
22
|
McLennan D, Armstrong JD, Stewart DC, Mckelvey S, Boner W, Monaghan P, Metcalfe NB. Telomere elongation during early development is independent of environmental temperatures in Atlantic salmon. ACTA ACUST UNITED AC 2018; 221:jeb.178616. [PMID: 29636409 PMCID: PMC6031317 DOI: 10.1242/jeb.178616] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/29/2018] [Indexed: 12/31/2022]
Abstract
There is increasing evidence from endothermic vertebrates that telomeres, which cap the ends of chromosomes and play an important role in chromosome protection, decline in length during postnatal life and are a useful indicator of physiological state and expected lifespan. However, much less is currently known about telomere dynamics in ectothermic vertebrates, which are likely to differ from that of endotherms, at least in part due to the sensitivity of ectotherm physiology to environmental temperature. We report here on an experiment in which Atlantic salmon (Salmo salar) were reared through the embryonic and larval stages of development, and under differing temperatures, in order to examine the effects of environmental temperature during early life on telomere dynamics, oxidative DNA damage and cellular proliferation. Telomere length significantly increased between the embryonic and larval stages of development. Contrary to our expectations, variation in telomere length at the end of the larval stage was unrelated to either cell proliferation rate or the relative level of oxidative DNA damage, and did not vary between the temperature treatments. This study suggests that salmon are able to restore the length of their telomeres during early development, which may possibly help to buffer potentially harmful environmental effects experienced in early life. Summary: The authors show that, in salmon, telomeres significantly lengthen between the embryonic and larval stages of development, and that this is not influenced by environmental temperature.
Collapse
Affiliation(s)
- Darryl McLennan
- Institute of Biodiversity, Animal Health and Comparative Medicine, Graham Kerr Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - John D Armstrong
- Marine Scotland-Science, Freshwater Laboratory, Faskally, Pitlochry, PH16 5LB, UK
| | - David C Stewart
- Marine Scotland-Science, Freshwater Laboratory, Faskally, Pitlochry, PH16 5LB, UK
| | - Simon Mckelvey
- Cromarty Firth Fishery Trust, CKD Galbraith, Reay House, 17 Old Edinburgh Road, Inverness, IV2 3HF
| | - Winnie Boner
- Institute of Biodiversity, Animal Health and Comparative Medicine, Graham Kerr Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Pat Monaghan
- Institute of Biodiversity, Animal Health and Comparative Medicine, Graham Kerr Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Neil B Metcalfe
- Institute of Biodiversity, Animal Health and Comparative Medicine, Graham Kerr Building, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
23
|
Argüello RJ, Reverendo M, Mendes A, Camosseto V, Torres AG, Ribas de Pouplana L, van de Pavert SA, Gatti E, Pierre P. SunRiSE - measuring translation elongation at single-cell resolution by means of flow cytometry. J Cell Sci 2018; 131:jcs.214346. [PMID: 29700204 DOI: 10.1242/jcs.214346] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/04/2018] [Indexed: 12/30/2022] Open
Abstract
The rate at which ribosomes translate mRNAs regulates protein expression by controlling co-translational protein folding and mRNA stability. Many factors regulate translation elongation, including tRNA levels, codon usage and phosphorylation of eukaryotic elongation factor 2 (eEF2). Current methods to measure translation elongation lack single-cell resolution, require expression of multiple transgenes and have never been successfully applied ex vivo Here, we show, by using a combination of puromycilation detection and flow cytometry (a method we call 'SunRiSE'), that translation elongation can be measured accurately in primary cells in pure or heterogenous populations isolated from blood or tissues. This method allows for the simultaneous monitoring of multiple parameters, such as mTOR or S6K1/2 signaling activity, the cell cycle stage and phosphorylation of translation factors in single cells, without elaborated, costly and lengthy purification procedures. We took advantage of SunRiSE to demonstrate that, in mouse embryonic fibroblasts, eEF2 phosphorylation by eEF2 kinase (eEF2K) mostly affects translation engagement, but has a surprisingly small effect on elongation, except after proteotoxic stress induction.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Rafael J Argüello
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France
| | - Marisa Reverendo
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France
| | - Andreia Mendes
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France
| | - Voahirana Camosseto
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France
| | - Adrian G Torres
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Lluis Ribas de Pouplana
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain.,Catalan Institute for Research and Advanced Studies (ICREA), P/Lluis Companys 23, 08010 Barcelona, Catalonia, Spain
| | - Serge A van de Pavert
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France
| | - Evelina Gatti
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France.,Institute for Research in Biomedicine (iBiMED) and Ilidio Pinho Foundation, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Philippe Pierre
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288, Marseille Cedex 9, France .,Institute for Research in Biomedicine (iBiMED) and Ilidio Pinho Foundation, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
24
|
Hoffmann N, Weise SC, Marinaro F, Vogel T, De Pietri Tonelli D. DGCR8 Promotes Neural Progenitor Expansion and Represses Neurogenesis in the Mouse Embryonic Neocortex. Front Neurosci 2018; 12:281. [PMID: 29760646 PMCID: PMC5936999 DOI: 10.3389/fnins.2018.00281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/10/2018] [Indexed: 11/17/2022] Open
Abstract
DGCR8 and DROSHA are the minimal functional core of the Microprocessor complex essential for biogenesis of canonical microRNAs and for the processing of other RNAs. Conditional deletion of Dgcr8 and Drosha in the murine telencephalon indicated that these proteins exert crucial functions in corticogenesis. The identification of mechanisms of DGCR8- or DROSHA-dependent regulation of gene expression in conditional knockout mice are often complicated by massive apoptosis. Here, to investigate DGCR8 functions on amplification/differentiation of neural progenitors cells (NPCs) in corticogenesis, we overexpress Dgcr8 in the mouse telencephalon, by in utero electroporation (IUEp). We find that DGCR8 promotes the expansion of NPC pools and represses neurogenesis, in absence of apoptosis, thus overcoming the usual limitations of Dgcr8 knockout-based approach. Interestingly, DGCR8 selectively promotes basal progenitor amplification at later developmental stages, entailing intriguing implications for neocortical expansion in evolution. Finally, despite a 3- to 5-fold increase of DGCR8 level in the mouse telencephalon, the composition, target preference and function of the DROSHA-dependent Microprocessor complex remain unaltered. Thus, we propose that DGCR8-dependent modulation of gene expression in corticogenesis is more complex than previously known, and possibly DROSHA-independent.
Collapse
Affiliation(s)
- Nadin Hoffmann
- Neurobiology of miRNA Laboratory, Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Stefan C Weise
- Department of Molecular Embryology, Medical Faculty, Institute for Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Federica Marinaro
- Neurobiology of miRNA Laboratory, Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Tanja Vogel
- Department of Molecular Embryology, Medical Faculty, Institute for Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Davide De Pietri Tonelli
- Neurobiology of miRNA Laboratory, Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
25
|
|
26
|
Endoplasmic Reticulum Stress Contributes to the Loss of Newborn Hippocampal Neurons after Traumatic Brain Injury. J Neurosci 2018; 38:2372-2384. [PMID: 29386258 DOI: 10.1523/jneurosci.1756-17.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/17/2017] [Accepted: 01/24/2018] [Indexed: 01/24/2023] Open
Abstract
Adult hippocampal neurogenesis has been shown to be required for certain types of cognitive function. For example, studies have shown that these neurons are critical for pattern separation, the ability to store similar experiences as distinct memories. Although traumatic brain injury (TBI) has been shown to cause the loss of newborn hippocampal neurons, the signaling pathway(s) that triggers their death is unknown. Endoplasmic reticulum (ER) stress activates the PERK-eIF2α pathway that acts to restore ER function and improve cell survival. However, unresolved/intense ER stress activates C/EBP homologous protein (CHOP), leading to cell death. We show that TBI causes the death of hippocampal newborn neurons via CHOP. Using CHOP KO mice, we show that loss of CHOP markedly reduces newborn neuron loss after TBI. Injured CHOP mice performed significantly better in a context fear discrimination task compared with injured wild-type mice. In contrast, the PERK inhibitor GSK2606414 exacerbated doublecortin cell loss and worsened contextual discrimination. Administration of guanabenz (which reduces ER stress) to injured male rats reduced the loss of newborn neurons and improved one-trial contextual fear memory. Interestingly, we also found that the surviving newborn neurons in brain-injured animals had dendritic loss, which was not observed in injured CHOP KO mice or in animals treated with guanabenz. These results indicate that ER stress plays a key role in the death of newborn neurons after TBI. Further, these findings indicate that ER stress can alter dendritic arbors, suggesting a role for ER stress in neuroplasticity and dendritic pathologies.SIGNIFICANCE STATEMENT The hippocampus, a structure in the temporal lobe, is critical for learning and memory. The hippocampus is one of only two areas in which neurons are generated in the adult brain. These newborn neurons are required for certain types of memory, and are particularly vulnerable to traumatic brain injury (TBI). However, the mechanism(s) that causes the loss of these cells after TBI is poorly understood. We show that endoplasmic reticulum (ER) stress pathways are activated in newborn neurons after TBI, and that manipulation of the CHOP cascade improves newborn neuron survival and cognitive outcome. These results suggest that treatments that prevent/resolve ER stress may be beneficial in treating TBI-triggered memory dysfunction.
Collapse
|
27
|
Panaliappan TK, Wittmann W, Jidigam VK, Mercurio S, Bertolini JA, Sghari S, Bose R, Patthey C, Nicolis SK, Gunhaga L. Sox2 is required for olfactory pit formation and olfactory neurogenesis through BMP restriction and Hes5 upregulation. Development 2018; 145:145/2/dev153791. [PMID: 29352015 PMCID: PMC5825848 DOI: 10.1242/dev.153791] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
The transcription factor Sox2 is necessary to maintain pluripotency of embryonic stem cells, and to regulate neural development. Neurogenesis in the vertebrate olfactory epithelium persists from embryonic stages through adulthood. The role Sox2 plays for the development of the olfactory epithelium and neurogenesis within has, however, not been determined. Here, by analysing Sox2 conditional knockout mouse embryos and chick embryos deprived of Sox2 in the olfactory epithelium using CRISPR-Cas9, we show that Sox2 activity is crucial for the induction of the neural progenitor gene Hes5 and for subsequent differentiation of the neuronal lineage. Our results also suggest that Sox2 activity promotes the neurogenic domain in the nasal epithelium by restricting Bmp4 expression. The Sox2-deficient olfactory epithelium displays diminished cell cycle progression and proliferation, a dramatic increase in apoptosis and finally olfactory pit atrophy. Moreover, chromatin immunoprecipitation data show that Sox2 directly binds to the Hes5 promoter in both the PNS and CNS. Taken together, our results indicate that Sox2 is essential to establish, maintain and expand the neuronal progenitor pool by suppressing Bmp4 and upregulating Hes5 expression. Summary: Analysis of Sox2 mutant mouse and Sox2 CRISPR-targeted chick embryos reveals that Sox2 controls the establishment of sensory progenitors in the olfactory epithelium by suppressing Bmp4 and upregulating Hes5 expression.
Collapse
Affiliation(s)
| | - Walter Wittmann
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Vijay K Jidigam
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Sara Mercurio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Jessica A Bertolini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Soufien Sghari
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Raj Bose
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Cedric Patthey
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
28
|
Matatall KA, Kadmon CS, King KY. Detecting Hematopoietic Stem Cell Proliferation Using BrdU Incorporation. Methods Mol Biol 2018; 1686:91-103. [PMID: 29030815 PMCID: PMC6020038 DOI: 10.1007/978-1-4939-7371-2_7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cellular quiescence is a key component of hematopoietic stem cell (HSC) homeostasis; therefore, a reliable method to measure HSC cell division is critical in many studies. However, measuring the proliferation rate of largely quiescent and rare populations of cells can be challenging. Bromo-deoxyuridine (BrdU) incorporation into replicating DNA is a commonly used and highly reproducible method to detect cell division history. Here, we describe a protocol for BrdU incorporation analysis in hematopoietic stem and progenitor cells that can provide a sensitive measure of cell division even in rare cell populations. In combination with flow cytometry, this method can be generalized to analyze other cell populations and other tissues as identified by cell surface markers.
Collapse
Affiliation(s)
- Katie A Matatall
- Pediatric Infectious Diseases, Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Claudine S Kadmon
- Pediatric Infectious Diseases, Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Katherine Y King
- Pediatric Infectious Diseases, Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
- Texas Children's Hospital, 1102 Bates Street, Suite 1150, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Demir MK, Yapıcıer O, Hasanov T, Kilic D, Kilic T. A rapidly expanding calvarial Langerhans cell histiocytosis with low Ki-67 in an adult: a challenging diagnosis on magnetic resonance imaging. Neuroradiol J 2017; 31:390-394. [PMID: 29125031 DOI: 10.1177/1971400917741905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Langerhans cell histiocytosis may have a wide variety of clinical presentations. The growth of localised form is usually slow and it mainly involves bones such as the skull, femur, spine, ribs, mandible and pelvis in children. The spectrum of clinical manifestations and magnetic resonance imaging findings of the disease may sometimes mimic infections as well as benign and malignant tumours. In this report, we describe an unusual case of rapidly progressive calvarial Langerhans cell histiocytosis with a painful and tender mass in a 27-year-old man that created diagnostic and therapeutic challenges. We also discuss the relationship between Ki-67 proliferative index and the expansion of the disease. Magnetic resonance imaging studies revealed a bone defect on the left frontal bone with soft tissue components, showing a prominent expansion within a month. The clinical manifestation with the enhancement pattern of epidural soft tissue component on contrast-enhanced magnetic resonance imaging suggested an infection besides Langerhans cell histiocytosis. Immunohistochemical expression of Ki-67 antigen was 5%. The presence of a relatively low Ki-67 value indicates that the inflammatory response may have a more important role than local Langerhans cell proliferation in the aggressive clinical course and rapid expansion.
Collapse
Affiliation(s)
| | - Ozlem Yapıcıer
- 2 Department of Pathology, Bahçeşehir University School of Medicine, Turkey
| | | | - Deniz Kilic
- 4 Department of Neurosurgery, Bahçeşehir University School of Medicine, Turkey
| | - Turker Kilic
- 3 Bahçeşehir University School of Medicine, Turkey
| |
Collapse
|
30
|
Acute stress promotes post-injury brain regeneration in fish. Brain Res 2017; 1676:28-37. [PMID: 28916442 DOI: 10.1016/j.brainres.2017.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 01/14/2023]
Abstract
The central nervous system and the immune system, the two major players in homeostasis, operate in the ongoing bidirectional interaction. Stress is the third player that exerts strong effect on these two 'supersystems'; yet, its impact is studied much less. In this work employing carp model, we studied the influence of preliminary stress on neural and immune networks involved in post-injury brain regeneration. The relevant in vivo models of air-exposure stress and precisely directed cerebellum injury have been developed. Neuronal regeneration was evaluated by using specific tracers of cell proliferation and differentiation. Involvement of immune networks was accessed by monitoring the expression of selected T cells markers. Contrast difference between acute and chronic stress manifested in the fact that chronically stressed fish did not survive the brain injury. Neuronal regeneration appeared as a biphasic process whereas involvement of immune system proceeded as a monophasic route. In stressed fish, immune response was fast and accompanied or even preceded neuronal regeneration. In unstressed subjects, immune response took place on the second phase of neuronal regeneration. These findings imply an intrinsic regulatory impact of acute stress on neuronal and immune factors involved in post-injury brain regeneration. Stress activates both neuronal and immune defense mechanisms and thus contributes to faster regeneration. In this context, paradoxically, acute preliminary stress might be considered a distinct asset in speeding up the following post-injury brain regeneration.
Collapse
|
31
|
Forsberg D, Thonabulsombat C, Jäderstad J, Jäderstad LM, Olivius P, Herlenius E. Functional Stem Cell Integration into Neural Networks Assessed by Organotypic Slice Cultures. ACTA ACUST UNITED AC 2017; 42:2D.13.1-2D.13.30. [PMID: 28806855 DOI: 10.1002/cpsc.34] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Re-formation or preservation of functional, electrically active neural networks has been proffered as one of the goals of stem cell-mediated neural therapeutics. A primary issue for a cell therapy approach is the formation of functional contacts between the implanted cells and the host tissue. Therefore, it is of fundamental interest to establish protocols that allow us to delineate a detailed time course of grafted stem cell survival, migration, differentiation, integration, and functional interaction with the host. One option for in vitro studies is to examine the integration of exogenous stem cells into an existing active neural network in ex vivo organotypic cultures. Organotypic cultures leave the structural integrity essentially intact while still allowing the microenvironment to be carefully controlled. This allows detailed studies over time of cellular responses and cell-cell interactions, which are not readily performed in vivo. This unit describes procedures for using organotypic slice cultures as ex vivo model systems for studying neural stem cell and embryonic stem cell engraftment and communication with CNS host tissue. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Charoensri Thonabulsombat
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Section of Otorhinolaryngology, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Center for Hearing and Communication Research, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Johan Jäderstad
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Linda Maria Jäderstad
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Petri Olivius
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Section of Otorhinolaryngology, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Center for Hearing and Communication Research, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Matson JP, Cook JG. Cell cycle proliferation decisions: the impact of single cell analyses. FEBS J 2017; 284:362-375. [PMID: 27634578 PMCID: PMC5296213 DOI: 10.1111/febs.13898] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/23/2016] [Accepted: 09/13/2016] [Indexed: 12/16/2022]
Abstract
Cell proliferation is a fundamental requirement for organismal development and homeostasis. The mammalian cell division cycle is tightly controlled to ensure complete and precise genome duplication and segregation of replicated chromosomes to daughter cells. The onset of DNA replication marks an irreversible commitment to cell division, and the accumulated efforts of many decades of molecular and cellular studies have probed this cellular decision, commonly called the restriction point. Despite a long-standing conceptual framework of the restriction point for progression through G1 phase into S phase or exit from G1 phase to quiescence (G0), recent technical advances in quantitative single cell analysis of mammalian cells have provided new insights. Significant intercellular heterogeneity revealed by single cell studies and the discovery of discrete subpopulations in proliferating cultures suggests the need for an even more nuanced understanding of cell proliferation decisions. In this review, we describe some of the recent developments in the cell cycle field made possible by quantitative single cell experimental approaches.
Collapse
Affiliation(s)
- Jacob P. Matson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
| | - Jeanette G. Cook
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
| |
Collapse
|
33
|
de Greeff A, Resink JW, van Hees HMJ, Ruuls L, Klaassen GJ, Rouwers SMG, Stockhofe-Zurwieden N. Supplementation of piglets with nutrient-dense complex milk replacer improves intestinal development and microbial fermentation. J Anim Sci 2016; 94:1012-9. [PMID: 27065263 DOI: 10.2527/jas.2015-9481] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Weaning of piglets causes stress due to environmental, behavioral, and nutritional stressors and can lead to postweaning diarrhea and impaired gut development. The diet changes experienced during weaning require extensive adaptation of the digestive system. A well-developed piglet that had creep-feed experience before weaning performs better after weaning. In the current study, the effect of providing sow-fed piglets with a supplemental nutrient-dense complex milk replacer (NDM) on gut development and growth performance was studied. Litters of sows with similar parities (3.6 ± 0.8) and similar numbers of live born piglets (13.5 ± 0.3) were assigned to 1 of 2 groups: 1 group of piglets had ad libitum access to NDM from Day 2 through 21 after birth, whereas the other group was used as controls. Nutrient-dense complex milk replacer-fed piglets were shown to be significantly heavier after 21 d of supplementation compared with the control piglets. At Day 21, 3 piglets from each litter were euthanized for morphological and functional analyses of the intestinal tract. The small intestines of NDM-fed piglets had significantly higher weights (g) as well as significantly higher relative weight:length ratios (g//cm) compared with the small intestines of control piglets ( < 0.05). Morphometric analysis demonstrated that villi length and numbers of goblet cells did not differ between groups. However, NDM-fed piglets had deeper crypts ( < 0.001) and an increased expression of the cell-proliferation marker proliferating cell nuclear antigen in crypts ( < 0.05), suggesting higher cell-proliferation rates. The gene encoding IGF-1 showed a tendency to higher gene expression in the jejunum from NDM-fed piglets ( = 0.07) compared with the jejunum from control piglets, suggesting that IGF-1 might be involved in the regulation of cell proliferation and intestinal growth. Finally, as a result of dietary fiber in NDM, piglets showed significantly increased concentrations of metabolic fermentation products. This suggests differences in metabolic activity in the colon between treatment groups. In conclusion, providing sow-fed piglets with NDM before weaning stimulates intestinal proliferation, leading to increased circular growth. Nutrient-dense complex milk replacer supplementation might, therefore, help piglets through the transition period at weaning by increased BW and increased capacity for uptake of nutrients.
Collapse
|
34
|
Smith SC, Price E, Hewitt MJ, Symonds EM, Baker PN. Cellular Proliferation in the Placenta in Normal Human Pregnancy and Pregnancy Complicated by Intrauterine Growth Restriction. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769800500607] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Stephen C. Smith
- Department of Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom; School of Human Development, New Maternity Unit. City Hospital, Hucknall Road, Nottingham, United Kingdom, NG5 1PB
| | | | | | | | - Philip N. Baker
- Department of Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
35
|
Lynn AAA, King SA, LiVolsi VA. Utility of Proliferation Markers Ki-67 and Proliferating Cell Nuclear Antigen (PCNA) in the Evaluation of Uterine Papillary Serous Carcinomas. Int J Surg Pathol 2016. [DOI: 10.1177/106689699700400403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined proliferation markers Ki-67 and proliferating cell nuclear antigen (PCNA) to determine whether immunohistochemical staining results could predict outcome in 22 uterine papillary serous carcinomas (UPSC). Eighteen tumors (82%) had increased proliferation as demonstrated by Ki-67 immunostaining with 50% of these patients dying of disease. Twenty tumors (91%) showed high PCNA immunoreactivity; 60% of these patients died of disease. Most UPSC exhibit high immunoreactivity to Ki-67 and/or PCNA, which showed no correlation with outcome or stage. These results may reflect the aggressive clinical behavior of UPSC. We conclude that Ki-67 and PCNA immunohistochemistry have limited use as prognostic indicators in UPSC.
Collapse
Affiliation(s)
| | | | - Virginia A. LiVolsi
- University of Pennsylvania Medical Center, Department of Pathology and Laboratory Medicine and Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Philadelphia, Pennsylvania
| |
Collapse
|
36
|
Sinha S, Verma S, Chaturvedi MM. Differential Expression of SWI/SNF Chromatin Remodeler Subunits Brahma and Brahma-Related Gene During Drug-Induced Liver Injury and Regeneration in Mouse Model. DNA Cell Biol 2016; 35:373-84. [PMID: 27097303 DOI: 10.1089/dna.2015.3155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The chromatin remodeling activity of mammalian SWI/SNF complex is carried out by either Brahma (BRM) or Brahma-related gene (BRG-1). The BRG-1 regulates genes involved in cell proliferation, whereas BRM is associated with cell differentiation, and arrest of cell growth. Global modifications of histones and expression of genes of chromatin-remodeling subunits have not been studied in in vivo model systems. In the present study, we investigate epigenetic modifications of histones and the expression of genes in thioacetamide (TAA)-induced liver injury and regeneration in a mouse model. In the present study, we report that hepatocyte proliferation and H3S10 phosphorylation occur during 60 to 72 h post TAA treatment in mice. Furthermore, there was change in the H3K9 acetylation and H3K9 trimethylation pattern with respect to liver injury and regeneration phase. Looking into the expression pattern of Brg-1 and Brm, it is evident that they contribute substantially to the process of liver regeneration. The SWI/SNF remodeler might contain BRG-1 as its ATPase subunit during injury phase. Whereas, BRM-associated SWI/SNF remodeler might probably be predominant during decline of injury phase and initiation of regeneration phase. Furthermore, during the regeneration phase, BRG-1-containing remodeler again predominates. Considering all these observations, the present study depicts an interplay between chromatin interacting machineries in different phases of thioacetamide-induced liver injury and regeneration.
Collapse
Affiliation(s)
- Sonal Sinha
- 1 Laboratory for Chromatin Biology, Department of Zoology, University of Delhi , New Delhi, India
| | - Sudhir Verma
- 1 Laboratory for Chromatin Biology, Department of Zoology, University of Delhi , New Delhi, India
| | - Madan M Chaturvedi
- 1 Laboratory for Chromatin Biology, Department of Zoology, University of Delhi , New Delhi, India .,2 Cluster Innovation Center, Delhi University , Delhi, India
| |
Collapse
|
37
|
Mangiavini L, Merceron C, Schipani E. Analysis of Mouse Growth Plate Development. CURRENT PROTOCOLS IN MOUSE BIOLOGY 2016; 6:67-130. [PMID: 26928664 PMCID: PMC5493209 DOI: 10.1002/9780470942390.mo150094] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
To investigate skeletal development, pathophysiological mechanisms of cartilage and bone disease, and eventually assess innovative treatments, the mouse is a very important resource. During embryonic development, mesenchymal condensations are formed, and cells within these mesenchymal condensations either directly differentiate into osteoblasts and give origin to intramembranous bone, or differentiate into chondrocytes and form a cartilaginous anlage. The cartilaginous anlage or fetal growth plate is then replaced with bone. This process is also called endochondral bone development, and it is responsible for the generation of most of our skeleton. Here we discuss in detail the most common in vivo and in vitro techniques our laboratory is currently using for the analysis of the mouse fetal growth plate during development.
Collapse
Affiliation(s)
- Laura Mangiavini
- Department of Orthopaedic Surgery, Department of Medicine, Division of Endocrinology and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Christophe Merceron
- Department of Orthopaedic Surgery, Department of Medicine, Division of Endocrinology and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
- Inserm, UMRS 791-LIOAD, Centre for Osteoarticular and Dental Tissue Engineering, Group STEP ‘Skeletal Tissue Engineering and Physiopathology’, Nantes, France
- Faculty of Dental Surgery, LUNAM, Nantes University, Nantes, France
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, Department of Medicine, Division of Endocrinology and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
38
|
Osei-Sarfo K, Urvalek AM, Tang XH, Scognamiglio T, Gudas LJ. Initiation of esophageal squamous cell carcinoma (ESCC) in a murine 4-nitroquinoline-1-oxide and alcohol carcinogenesis model. Oncotarget 2016; 6:6040-52. [PMID: 25714027 PMCID: PMC4467420 DOI: 10.18632/oncotarget.3339] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/04/2015] [Indexed: 01/14/2023] Open
Abstract
Esophageal squamous cell carcinomas (ESCCs) are very common, aggressive tumors, and are often associated with alcohol and tobacco abuse. Because ESCCs exhibit high recurrence rates and are diagnosed at late stages, identification of prognostic and drug targets for prevention and treatment is critical. We used the 4-nitroquinoline-1-oxide (4-NQO) murine model of oral carcinogenesis and the Meadows-Cook model of alcohol abuse to assess changes in the expression of molecular markers during the initial stages of ESCC. Combining these two models, which mimic chronic alcohol and tobacco abuse in humans, we detected increased cellular proliferation (EGFR and Ki67 expression), increased canonical Wnt signaling and downstream elements (β-catenin, FoxM1, and S100a4 protein levels), changes in cellular adhesive properties (reduced E-cadherin in the basal layer of the esophageal epithelium), and increased levels of phosphorylated ERK1/2 and p38. Additionally, we found that treatment with ethanol alone increased the numbers of epithelial cells expressing solute carrier family 2 (facilitated glucose transporter, member 1) (SLC2A1) and carbonic anhydrase IX (CAIX), and increased the phosphorylation of p38. Thus, we identified both 4-NQO- and ethanol-specific targets in the initial stages of esophageal carcinogenesis, which should lead to the development of potential markers and therapeutic targets for human ESCC.
Collapse
Affiliation(s)
- Kwame Osei-Sarfo
- Department of Pharmacology, Weill Cornell Medical College, New York, USA
| | - Alison M Urvalek
- Department of Pharmacology, Weill Cornell Medical College, New York, USA
| | - Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medical College, New York, USA
| | | | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, USA.,The Meyer Cancer Center, Weill Cornell Medical College, New York, USA
| |
Collapse
|
39
|
Garnol T, Kučera O, Staňková P, Lotková H, Červinková Z. Does Simple Steatosis Affect Liver Regeneration after Partial Hepatectomy in Rats? ACTA MEDICA (HRADEC KRALOVE) 2016; 59:35-42. [PMID: 27526303 DOI: 10.14712/18059694.2016.87] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM The aim of our study was to assess whether simple steatosis impairs liver regeneration after partial hepatectomy (PHx) in rats. METHODS Male Sprague-Dawley rats were fed a standard diet (ST-1, 10% kcal fat) and high-fat diet (HFD, 71% kcal fat) for 6 weeks. Then the rats were submitted to 2/3 PHx and animals were sacrificed 24, 48 or 72 h after PHx. Serum biochemistry, respiration of mitochondria in liver homogenate, hepatic oxidative stress markers, selected cytokines and DNA content were measured, and histopathological samples were prepared. Liver regeneration was evaluated by incorporation of bromodeoxyuridine (BrdU) to hepatocyte DNA. RESULTS HFD induced simple microvesicular liver steatosis. PHx caused elevation of serum markers of liver injury in both groups; however, an increase in these parameters was delayed in HFD group. Hepatic content of reduced glutathione was significantly increased in both groups after PHx. There were no significant changes in activities of respiratory complexes I and II (state 3). Relative and absolute liver weights, total DNA content, and DNA synthesis exerted very similar changes in both ST-1 and HFD groups after PHx. CONCLUSION PHx-induced regeneration of the rat liver with simple steatosis was not significantly affected when compared to the lean liver.
Collapse
Affiliation(s)
- Tomáš Garnol
- Department of Physiology, Charles University in Prague, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Otto Kučera
- Department of Physiology, Charles University in Prague, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic.
| | - Pavla Staňková
- Department of Physiology, Charles University in Prague, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Halka Lotková
- Department of Physiology, Charles University in Prague, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Zuzana Červinková
- Department of Physiology, Charles University in Prague, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| |
Collapse
|
40
|
Oboti L, Ibarra-Soria X, Pérez-Gómez A, Schmid A, Pyrski M, Paschek N, Kircher S, Logan DW, Leinders-Zufall T, Zufall F, Chamero P. Pregnancy and estrogen enhance neural progenitor-cell proliferation in the vomeronasal sensory epithelium. BMC Biol 2015; 13:104. [PMID: 26621367 PMCID: PMC4665882 DOI: 10.1186/s12915-015-0211-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/16/2015] [Indexed: 12/02/2022] Open
Abstract
Background The hormonal state during the estrus cycle or pregnancy produces alterations on female olfactory perception that are accompanied by specific maternal behaviors, but it is unclear how sex hormones act on the olfactory system to enable these sensory changes. Results Herein, we show that the production of neuronal progenitors is stimulated in the vomeronasal organ (VNO) epithelium of female mice during a late phase of pregnancy. Using a wide range of molecular markers that cover the whole VNO cell maturation process in combination with Ca2+ imaging in early postmitotic neurons, we show that newly generated VNO cells adopt morphological and functional properties of mature sensory neurons. A fraction of these newly generated cells project their axons to the olfactory forebrain, extend dendrites that contact the VNO lumen, and can detect peptides and urinary proteins shown to contain pheromone activity. High-throughput RNA-sequencing reveals concomitant differences in gene expression in the VNO transcriptomes of pregnant females. These include relative increases in expression of 20 vomeronasal receptors, of which 17 belong to the V1R subfamily, and may therefore be considered as candidate receptors for mediating maternal behaviors. We identify the expression of several hormone receptors in the VNO of which estrogen receptor α (Esr1) is directly localized to neural progenitors. Administration of sustained high levels of estrogen, but not progesterone, is sufficient to stimulate vomeronasal progenitor cell proliferation in the VNO epithelium. Conclusions Peripheral olfactory neurogenesis driven by estrogen may contribute to modulate sensory perception and adaptive VNO-dependent behaviors during pregnancy and early motherhood. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0211-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Livio Oboti
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany. .,Present address: Center for Neuroscience Research, Children's National Health System, 20010, Washington, DC, USA.
| | - Ximena Ibarra-Soria
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Anabel Pérez-Gómez
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Andreas Schmid
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Martina Pyrski
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Nicole Paschek
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Sarah Kircher
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Darren W Logan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK. .,Monell Chemical Senses Center, 3500 Market Street, Philadelphia, Pennsylvania, 19104, USA.
| | - Trese Leinders-Zufall
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Frank Zufall
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany.
| | - Pablo Chamero
- Department of Physiology, and Center for Integrative Physiology and Molecular Medicine, University of Saarland School of Medicine, 66421, Homburg, Germany. .,Present address: Laboratoire de Physiologie de la Reproduction & des Comportments, UMR 7247 INRA-CNRS-Université François Rabelais, F-37380, Nouzilly, France.
| |
Collapse
|
41
|
Parham DM. Immunohistochemical Markers of Soft Tissue Tumors: Pathologic Diagnosis, Genetic Contributions, and Therapeutic Options. ANALYTICAL CHEMISTRY INSIGHTS 2015; 10:1-10. [PMID: 26549970 PMCID: PMC4627416 DOI: 10.4137/aci.s32730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 09/16/2015] [Indexed: 12/18/2022]
Abstract
After ~30 years of widespread usage, immunohistochemistry (IHC) has become a standard method of diagnosis for surgical pathology. Because of the plethora of diagnoses and often subtle nature of diagnostic criteria, IHC finds particular utility in soft tissue tumors. The use of progressively small amounts of tissue for diagnosis highlights the importance of this method. The sensitivity and crispness of IHC stains have progressively improved with the advent of new techniques. Traditionally, IHC detects cell-typic markers that characterize cell phenotypes, such as chromogranin for neuroectodermal tissue, myogenin for skeletal muscle, and cytokeratin for epithelium. However, the advent of genetic discoveries have led to IHC testing for detection of fusion gene products or overexpressed oncogenes associated with deletions and mutations. Proliferation-based markers such as Ki-67 can also be used for prognosis and grading, but more standardization is needed. Development of monoclonal antibody-based pharmaceuticals, such as imatinib or crizotinib, holds the promise of tailored anticancer therapy. IHC thus has assumed importance not only for diagnosis but also for guidance of personalized medicine.
Collapse
Affiliation(s)
- David M Parham
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA. ; Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
42
|
Bonventre JV. Maladaptive proximal tubule repair: cell cycle arrest. NEPHRON. CLINICAL PRACTICE 2015; 127:61-4. [PMID: 25343823 DOI: 10.1159/000363673] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Acute kidney injury (AKI) leads to worsening of chronic kidney disease (CKD), and CKD predisposes to the clinical entity of AKI. The tubules of the kidney play a central role in the fibrotic response, which ultimately leads to progressive kidney disease. The cellular mechanisms responsible for the epidemiological association between AKI and CKD are complex. In order to unravel characteristics of this direct involvement of the tubules, in particular the proximal tubules, we established a model to specifically target injury to the proximal tubule using a genetic approach to express the simian diphtheria toxin (DT) receptor in the proximal tubule. A single administration of DT to the proximal tubule resulted in inflammation, reversible injury, and adaptive repair. By contrast, thrice repeated injury led to maladaptive repair with sustained tubule injury, vascular rarefaction, proliferation of interstitial myofibroblasts, interstitial fibrosis, and glomerular sclerosis. An important feature of the maladaptive repair process after severe injury is the development of cell cycle arrest in G2/M. There is a subsequent activation of the DNA repair response with activation of a secretory phenotype whereby profibrotic factors are released. This insight introduces a number of potential new targets for therapeutic intervention to prevent and/or arrest CKD progression.
Collapse
Affiliation(s)
- Joseph V Bonventre
- Renal Division and Biomedical Engineering Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass., USA
| |
Collapse
|
43
|
Gamat M, Malinowski RL, Parkhurst LJ, Steinke LM, Marker PC. Ornithine Decarboxylase Activity Is Required for Prostatic Budding in the Developing Mouse Prostate. PLoS One 2015; 10:e0139522. [PMID: 26426536 PMCID: PMC4591331 DOI: 10.1371/journal.pone.0139522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/12/2015] [Indexed: 11/23/2022] Open
Abstract
The prostate is a male accessory sex gland that produces secretions in seminal fluid to facilitate fertilization. Prostate secretory function is dependent on androgens, although the mechanism by which androgens exert their effects is still unclear. Polyamines are small cationic molecules that play pivotal roles in DNA transcription, translation and gene regulation. The rate-limiting enzyme in polyamine biosynthesis is ornithine decarboxylase, which is encoded by the gene Odc1. Ornithine decarboxylase mRNA decreases in the prostate upon castration and increases upon administration of androgens. Furthermore, testosterone administered to castrated male mice restores prostate secretory activity, whereas administering testosterone and the ornithine decarboxylase inhibitor D,L-α-difluromethylornithine (DFMO) to castrated males does not restore prostate secretory activity, suggesting that polyamines are required for androgens to exert their effects. To date, no one has examined polyamines in prostate development, which is also androgen dependent. In this study, we showed that ornithine decarboxylase protein was expressed in the epithelium of the ventral, dorsolateral and anterior lobes of the adult mouse prostate. Ornithine decarboxylase protein was also expressed in the urogenital sinus (UGS) epithelium of the male and female embryo prior to prostate development, and expression continued in prostatic epithelial buds as they emerged from the UGS. Inhibiting ornithine decarboxylase using DFMO in UGS organ culture blocked the induction of prostatic buds by androgens, and significantly decreased expression of key prostate transcription factor, Nkx3.1, by androgens. DFMO also significantly decreased the expression of developmental regulatory gene Notch1. Other genes implicated in prostatic development including Sox9, Wif1 and Srd5a2 were unaffected by DFMO. Together these results indicate that Odc1 and polyamines are required for androgens to exert their effect in mediating prostatic bud induction, and are required for the expression of a subset of prostatic developmental regulatory genes including Notch1 and Nkx3.1.
Collapse
Affiliation(s)
- Melissa Gamat
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Rita L. Malinowski
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Linnea J. Parkhurst
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Laura M. Steinke
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Paul C. Marker
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
- * E-mail:
| |
Collapse
|
44
|
Wang J, Lu Z, Wang J, Cui M, Yeung BZ, Cole DJ, Wientjes MG, Au JLS. Paclitaxel tumor priming promotes delivery and transfection of intravenous lipid-siRNA in pancreatic tumors. J Control Release 2015; 216:103-10. [PMID: 26272765 DOI: 10.1016/j.jconrel.2015.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/14/2015] [Accepted: 08/05/2015] [Indexed: 01/05/2023]
Abstract
The major barrier for using small interfering RNA (siRNA) as cancer therapeutics is the inadequate delivery and transfection in solid tumors. We have previously shown that paclitaxel tumor priming, by inducing apoptosis, expands the tumor interstitial space, improves the penetration and dispersion of nanoparticles and siRNA-lipoplexes in 3-dimensional tumor histocultures, and promotes the delivery and transfection efficiency of siRNA-lipoplexes under the locoregional setting in vivo (i.e., intraperitoneal treatment of intraperitoneal tumors). The current study evaluated whether tumor priming is functional for systemically delivered siRNA via intravenous injection, which would subject siRNA to several additional delivery barriers and elimination processes. We used the same pegylated cationic (PCat)-siRNA lipoplexes as in the intraperitoneal study to treat mice bearing subcutaneous human pancreatic Hs766T xenograft tumors. The target gene was survivin, an inducible chemoresistance gene. The results show single agent paclitaxel delayed tumor growth but also significantly induced the survivin protein level in residual tumors, whereas addition of PCat-siSurvivin completely reversed the paclitaxel-induced survivin and enhanced the paclitaxel activity (p<0.05). In comparison, PCat-siSurvivin alone did not yield survivin knockdown or antitumor activity, indicating the in vivo effectiveness of intravenous siRNA-mediated gene silencing requires paclitaxel cotreatment. Additional in vitro studies showed that paclitaxel promoted the cytoplasmic release of siGLO, a 22 nucleotide double-stranded RNA that has no mRNA targets, from its PCat lipoplex and/or endosomes/lysosomes. Taken together, our earlier and current data show paclitaxel tumor priming, by promoting the interstitial transport and cytoplasmic release, is critical to promote the delivery and transfection of siRNA in vivo. In addition, because paclitaxel has broad spectrum activity and is used to treat multiple types of solid tumors including the hard-to-treat pancreatic cancer, the synergistic paclitaxel+siSurvivin combination represents a potentially useful chemo-gene therapy.
Collapse
Affiliation(s)
- Jie Wang
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Ze Lu
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Junfeng Wang
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Minjian Cui
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Bertrand Z Yeung
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | - David J Cole
- Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Jessie L-S Au
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA; Medical University of South Carolina, Charleston, SC 29425, USA; Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
45
|
Zhou L, Wang R, Yao C, Li X, Wang C, Zhang X, Xu C, Zeng A, Zhao D, Zhang F. Single-band upconversion nanoprobes for multiplexed simultaneous in situ molecular mapping of cancer biomarkers. Nat Commun 2015; 6:6938. [PMID: 25907226 PMCID: PMC4423208 DOI: 10.1038/ncomms7938] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/16/2015] [Indexed: 02/06/2023] Open
Abstract
The identification of potential diagnostic markers and target molecules among the plethora of tumour oncoproteins for cancer diagnosis requires facile technology that is capable of quantitatively analysing multiple biomarkers in tumour cells and tissues. Diagnostic and prognostic classifications of human tumours are currently based on the western blotting and single-colour immunohistochemical methods that are not suitable for multiplexed detection. Herein, we report a general and novel method to prepare single-band upconversion nanoparticles with different colours. The expression levels of three biomarkers in breast cancer cells were determined using single-band upconversion nanoparticles, western blotting and immunohistochemical technologies with excellent correlation. Significantly, the application of antibody-conjugated single-band upconversion nanoparticle molecular profiling technology can achieve the multiplexed simultaneous in situ biodetection of biomarkers in breast cancer cells and tissue specimens and produce more accurate results for the simultaneous quantification of proteins present at low levels compared with classical immunohistochemical technology.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Chemistry and Laboratory of Advanced Materials, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Rui Wang
- Department of Chemistry, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Chi Yao
- Department of Chemistry, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Xiaomin Li
- Department of Chemistry, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Chengli Wang
- Department of Chemistry, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Xiaoyan Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Aijun Zeng
- Shanghai Institute of Optics and Fine Mechanics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Dongyuan Zhao
- Department of Chemistry, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Fan Zhang
- Department of Chemistry, iChEm (Collaborative Innovation Center of Chemistry for Energy Materials), State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| |
Collapse
|
46
|
Karaca G, Xie G, Moylan C, Swiderska-Syn M, Guy CD, Krüger L, Machado MV, Choi SS, Michelotti GA, Burkly LC, Diehl AM. Role of Fn14 in acute alcoholic steatohepatitis in mice. Am J Physiol Gastrointest Liver Physiol 2015; 308:G325-34. [PMID: 25524063 PMCID: PMC4329478 DOI: 10.1152/ajpgi.00429.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TNF-like weak inducer of apoptosis (TWEAK) is a growth factor for bipotent liver progenitors that express its receptor, fibroblast growth factor-inducible 14 (Fn14), a TNF receptor superfamily member. Accumulation of Fn14(+) progenitors occurs in severe acute alcoholic steatohepatitis (ASH) and correlates with acute mortality. In patients with severe ASH, inhibition of TNF-α increases acute mortality. The aim of this study was to determine whether deletion of Fn14 improves the outcome of liver injury in alcohol-consuming mice. Wild-type (WT) and Fn14 knockout (KO) mice were fed control high-fat Lieber deCarli diet or high-fat Lieber deCarli diet with 2% alcohol (ETOH) and injected intraperitoneally with CCl₄ for 2 wk to induce liver injury. Mice were euthanized 3 or 10 days after CCl₄ treatment. Survival was assessed. Liver tissues were analyzed for cell death, inflammation, proliferation, progenitor accumulation, and fibrosis by quantitative RT-PCR, immunoblot, hydroxyproline content, and quantitative immunohistochemistry. During liver injury, Fn14 expression, apoptosis, inflammation, hepatocyte replication, progenitor and myofibroblast accumulation, and fibrosis increased in WT mice fed either diet. Mice fed either diet expressed similar TWEAK/Fn14 levels, but ETOH-fed mice had higher TNF-α expression. The ETOH-fed group developed more apoptosis, inflammation, fibrosis, and regenerative responses. Fn14 deletion did not reduce hepatic TNF-α expression but improved all injury parameters in mice fed the control diet. In ETOH-fed mice, Fn14 deletion inhibited TNF-α induction and increased acute mortality, despite improvement in liver injury. Fn14 mediates wound-healing responses that are necessary to survive acute liver injury during alcohol exposure.
Collapse
Affiliation(s)
- Gamze Karaca
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Guanhua Xie
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Cynthia Moylan
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Marzena Swiderska-Syn
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Cynthia D. Guy
- 2Department of Pathology, Duke University Medical Center, Durham, North Carolina;
| | - Leandi Krüger
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Mariana Verdelho Machado
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Steve S. Choi
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina; ,3Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina; and
| | - Gregory A. Michelotti
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Linda C. Burkly
- 4Department of Immunology, Biogen Idec, Inc., Cambridge, Massachusetts
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| |
Collapse
|
47
|
Ido Y, Duranton A, Lan F, Weikel KA, Breton L, Ruderman NB. Resveratrol prevents oxidative stress-induced senescence and proliferative dysfunction by activating the AMPK-FOXO3 cascade in cultured primary human keratinocytes. PLoS One 2015; 10:e0115341. [PMID: 25647160 PMCID: PMC4315597 DOI: 10.1371/journal.pone.0115341] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/21/2014] [Indexed: 01/27/2023] Open
Abstract
The aging process is perceived as resulting from a combination of intrinsic factors such as changes in intracellular signaling and extrinsic factors, most notably environmental stressors. In skin, the relationship between intrinsic changes and keratinocyte function is not clearly understood. Previously, we found that increasing the activity of AMP-activated protein kinase (AMPK) suppressed senescence in hydrogen peroxide (H2O2)-treated human primary keratinocytes, a model of oxidative stress-induced cellular aging. Using this model in the present study, we observed that resveratrol, an agent that increases the activities of both AMPK and sirtuins, ameliorated two age-associated phenotypes: cellular senescence and proliferative dysfunction. In addition, we found that treatment of keratinocytes with Ex527, a specific inhibitor of sirtuin 1 (SIRT1), attenuated the ability of resveratrol to suppress senescence. In keeping with the latter observation, we noted that compared to non-senescent keratinocytes, senescent cells lacked SIRT1. In addition to these effects on H2O2-induced senescence, resveratrol also prevented the H2O2-induced decrease in proliferation (as indicated by 3H-thymidine incorporation) in the presence of insulin. This effect was abrogated by inhibition of AMPK but not SIRT1. Compared to endothelium, we found that human keratinocytes expressed relatively high levels of Forkhead box O3 (FOXO3), a downstream target of both AMPK and SIRT1. Treatment of keratinocytes with resveratrol transactivated FOXO3 and increased the expression of its target genes including catalase. Resveratrol’s effects on both senescence and proliferation disappeared when FOXO3 was knocked down. Finally, we performed an exploratory study which showed that skin from humans over 50 years old had lower AMPK activity than skin from individuals under age 20. Collectively, these findings suggest that the effects of resveratrol on keratinocyte senescence and proliferation are regulated by the AMPK-FOXO3 pathway and in some situations, but not all, by SIRT1.
Collapse
Affiliation(s)
- Yasuo Ido
- Diabetes and Metabolism Unit, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| | | | - Fan Lan
- Endocrinology, Second Affiliated Hospital Chongqing Medical University, Chongqing, China
| | - Karen A. Weikel
- Diabetes and Metabolism Unit, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lionel Breton
- L’OREAL Research and Innovation, Aulnay sous bois, France
| | - Neil B. Ruderman
- Diabetes and Metabolism Unit, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
48
|
Kjosness KM, Hines JE, Lovejoy CO, Reno PL. The pisiform growth plate is lost in humans and supports a role for Hox in growth plate formation. J Anat 2014; 225:527-38. [PMID: 25279687 PMCID: PMC4292754 DOI: 10.1111/joa.12235] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 12/29/2022] Open
Abstract
The human pisiform is a small, nodular, although functionally significant, bone of the wrist. In most other mammals, including apes and Australopithecus afarensis, pisiforms are elongate. An underappreciated fact is that the typical mammalian pisiform forms from two ossification centers. We hypothesize that: (i) the presence of a secondary ossification center in mammalian pisiforms indicates the existence of a growth plate; and (ii) human pisiform reduction results from growth plate loss. To address these hypotheses, we surveyed African ape pisiform ossification and confirmed the presence of a late-forming secondary ossification center in chimpanzees and gorillas. Identification of the initial ossification center occurs substantially earlier in apes relative to humans, raising questions concerning the homology of the human pisiform and the two mammalian ossification centers. Second, we conducted histological and immunohistochemical analyses of pisiform ossification in mice. We confirm the presence of two ossification centers separated by organized columnar and hypertrophic chondrocyte zones. Flattened chondrocytes were highly mitotic, indicating the presence of a growth plate. Hox genes have been proposed to play a fundamental role in growth plate patterning. The existence of a pisiform growth plate presents an interesting test case for the association between Hox expression and growth plate formation, and could explain the severe effects on the pisiform observed in Hoxa11 and Hoxd11 knockout mice. Consistent with this hypothesis, we show that Hoxd11 is expressed adjacent to the pisiform in late-stage embryonic mouse limbs supporting a role for Hox genes in growth plate specification. This raises questions concerning the mechanisms regulating Hox expression in the developing carpus.
Collapse
Affiliation(s)
- Kelsey M Kjosness
- Department of Anthropology, The Pennsylvania State University, University Park, PA, USA
| | | | | | | |
Collapse
|
49
|
Schlage WK, Iskandar AR, Kostadinova R, Xiang Y, Sewer A, Majeed S, Kuehn D, Frentzel S, Talikka M, Geertz M, Mathis C, Ivanov N, Hoeng J, Peitsch MC. In vitro systems toxicology approach to investigate the effects of repeated cigarette smoke exposure on human buccal and gingival organotypic epithelial tissue cultures. Toxicol Mech Methods 2014; 24:470-87. [PMID: 25046638 PMCID: PMC4219813 DOI: 10.3109/15376516.2014.943441] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/20/2014] [Accepted: 06/29/2014] [Indexed: 11/13/2022]
Abstract
Smoking has been associated with diseases of the lung, pulmonary airways and oral cavity. Cytologic, genomic and transcriptomic changes in oral mucosa correlate with oral pre-neoplasia, cancer and inflammation (e.g. periodontitis). Alteration of smoking-related gene expression changes in oral epithelial cells is similar to that in bronchial and nasal epithelial cells. Using a systems toxicology approach, we have previously assessed the impact of cigarette smoke (CS) seen as perturbations of biological processes in human nasal and bronchial organotypic epithelial culture models. Here, we report our further assessment using in vitro human oral organotypic epithelium models. We exposed the buccal and gingival organotypic epithelial tissue cultures to CS at the air-liquid interface. CS exposure was associated with increased secretion of inflammatory mediators, induction of cytochrome P450s activity and overall weak toxicity in both tissues. Using microarray technology, gene-set analysis and a novel computational modeling approach leveraging causal biological network models, we identified CS impact on xenobiotic metabolism-related pathways accompanied by a more subtle alteration in inflammatory processes. Gene-set analysis further indicated that the CS-induced pathways in the in vitro buccal tissue models resembled those in the in vivo buccal biopsies of smokers from a published dataset. These findings support the translatability of systems responses from in vitro to in vivo and demonstrate the applicability of oral organotypical tissue models for an impact assessment of CS on various tissues exposed during smoking, as well as for impact assessment of reduced-risk products.
Collapse
Affiliation(s)
- Walter K. Schlage
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Anita R. Iskandar
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Radina Kostadinova
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Yang Xiang
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Alain Sewer
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Shoaib Majeed
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Diana Kuehn
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Stefan Frentzel
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Marcel Geertz
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Carole Mathis
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Nikolai Ivanov
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| | - Manuel C. Peitsch
- Philip Morris International R&D, Philip Morris Products S.A.NeuchâtelSwitzerland
| |
Collapse
|
50
|
DeWitt J, Pappas A, Nishi R. Ciliary neurotrophic factor reduces the proliferation and promotes the differentiation of TH- MYCN transformed sympathoadrenal progenitors. Dev Neurosci 2014; 36:422-31. [PMID: 25171250 DOI: 10.1159/000365281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/17/2014] [Indexed: 11/19/2022] Open
Abstract
Neuroblastoma is a childhood cancer caused by the transformation of sympathoadrenal progenitors. By following the formation of tumors in homozygous TH-MYCN mice, an established mouse model of neuroblastoma, we were able to capture transformed cells prior to the formation of large, vascularized tumors in order to determine the responsiveness of cells to neurotrophic factors. We discovered that the ciliary neurotrophic factor (CNTF) receptor is abundantly expressed in tumor cells from these mice. Furthermore, CNTF - but not nerve growth factor, brain-derived nerve growth factor, neurotrophin 3, or glial cell line-derived neurotrophic factor - promoted neuronal differentiation and withdrawal from the cell cycle. Thus, the transformation of sympathoadrenal progenitors by MYCN overexpression differentially affects responsiveness to neurotrophic molecules.
Collapse
Affiliation(s)
- John DeWitt
- Department of Neurological Sciences and Neuroscience Graduate Program, University of Vermont, Burlington, Vt., USA
| | | | | |
Collapse
|