1
|
Seong M, Bak-Gordon P, Liu Z, Canoll PD, Manley JL. Splicing dysregulation in glioblastoma alters the function of cell migration-related genes. Glia 2024. [PMID: 39448549 DOI: 10.1002/glia.24630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 09/09/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Glioblastoma (GBM) has a poor prognosis with a high recurrence and low survival rate. Previous RNA-seq analyses have revealed that alternative splicing (AS) plays a role in GBM progression. Here, we present a novel AS analysis method (Semi-Q) and describe its use to identify GBM-specific AS events. We analyzed RNA-seq data from normal brain (NB), normal human astrocytes (NHAs) and GBM samples, and found that comparison between NHA and GBM was especially informative. Importantly, this analysis revealed that genes encoding cell migration-related proteins, including filamins (FLNs) and actinins (ACTNs), were among those most affected by differential AS. Functional assays revealed that dysregulated AS of FLNA, B and C transcripts produced protein isoforms that not only altered transcription of cell proliferation-related genes but also led to enhanced cell migration, resistance to cell death and/or mitochondrial respiratory function, while a dysregulated AS isoform of ACTN4 enhanced cell migration. Together, our results indicate that cell migration and actin cytoskeleton-related genes are differentially regulated by AS in GBM, supporting a role for AS in facilitating tumor growth and invasiveness.
Collapse
Affiliation(s)
- Minu Seong
- Department of Biological Science, Columbia University, New York, New York, USA
| | - Pedro Bak-Gordon
- Department of Biological Science, Columbia University, New York, New York, USA
| | - Zhaoqi Liu
- Department of Systems Biology, Columbia University, New York, New York, USA
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Peter D Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - James L Manley
- Department of Biological Science, Columbia University, New York, New York, USA
| |
Collapse
|
2
|
Yazdi SAM, Farmani E, Shahvaisi S, Javadi AE, Nazar E. Prediction of villin expression and tumor behavior in colorectal cancer. Cancer Treat Res Commun 2024; 40:100825. [PMID: 38852262 DOI: 10.1016/j.ctarc.2024.100825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Colorectal cancer is one of the most common cancers and the leading cause of cancer-related deaths worldwide. The incidence is gradually increasing, and the mortality and recurrence rates of the disease remain high. METHODS This study was conducted as a cross-sectional study using tissue samples of 106 patients who underwent surgery at Sina Hospital from 2021 to 2022. After histopathological examination and identification of the pathological features of the tumor, the samples were subjected to immunohistochemical staining using a monoclonal antibody against villin. RESULTS In this study, we observed a significant association between villin expression and tumor depth, as well as a correlation between villin expression and tumor location (colon or rectum). However, no association was found between villin expression and the number of affected lymph nodes and age, sex, tumor grade, and size. Furthermore, there was no significant association between villin expression and tumor vascular or neural invasion. CONCLUSION The extent of local invasion and metastasis are important factors in disease progression and can lead to treatment failure. Therefore, new biomarkers are needed to identify patients at risk of local and distant metastases and to enable appropriate treatment of patients.
Collapse
Affiliation(s)
- Seyed Amir Miratashi Yazdi
- Department of General Surgery, Sina Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Farmani
- Department of Pathology, Sina Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Shahvaisi
- Department of Pathology, Sina Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezoo Eftekhar Javadi
- Department of Pathology, Sina Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Nazar
- Department of Pathology, Sina Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Esmaeilniakooshkghazi A, Pham E, George SP, Ahrorov A, Villagomez FR, Byington M, Mukhopadhyay S, Patnaik S, Conrad JC, Naik M, Ravi S, Tebbutt N, Mooi J, Reehorst CM, Mariadason JM, Khurana S. In colon cancer cells fascin1 regulates adherens junction remodeling. FASEB J 2023; 37:e22786. [PMID: 36786724 DOI: 10.1096/fj.202201454r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 01/10/2023] [Indexed: 02/15/2023]
Abstract
Adherens junctions (AJs) are a defining feature of all epithelial cells. They regulate epithelial tissue architecture and integrity, and their dysregulation is a key step in tumor metastasis. AJ remodeling is crucial for cancer progression, and it plays a key role in tumor cell survival, growth, and dissemination. Few studies have examined AJ remodeling in cancer cells consequently, it remains poorly understood and unleveraged in the treatment of metastatic carcinomas. Fascin1 is an actin-bundling protein that is absent from the normal epithelium but its expression in colon cancer is linked to metastasis and increased mortality. Here, we provide the molecular mechanism of AJ remodeling in colon cancer cells and identify for the first time, fascin1's function in AJ remodeling. We show that in colon cancer cells fascin1 remodels junctional actin and actomyosin contractility which makes AJs less stable but more dynamic. By remodeling AJs fascin1 drives mechanoactivation of WNT/β-catenin signaling and generates "collective plasticity" which influences the behavior of cells during cell migration. The impact of mechanical inputs on WNT/β-catenin activation in cancer cells remains poorly understood. Our findings highlight the role of AJ remodeling and mechanosensitive WNT/β-catenin signaling in the growth and dissemination of colorectal carcinomas.
Collapse
Affiliation(s)
| | - Eric Pham
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Sudeep P George
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Afzal Ahrorov
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Fabian R Villagomez
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Michael Byington
- Department of Chemical and Bimolecular Engineering, University of Houston, Houston, Texas, USA
| | - Srijita Mukhopadhyay
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Srinivas Patnaik
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Jacinta C Conrad
- Department of Chemical and Bimolecular Engineering, University of Houston, Houston, Texas, USA
| | - Monali Naik
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Saathvika Ravi
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Niall Tebbutt
- Gastrointestinal Cancers Programs, Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Jennifer Mooi
- Gastrointestinal Cancers Programs, Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Camilla M Reehorst
- Gastrointestinal Cancers Programs, Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - John M Mariadason
- Gastrointestinal Cancers Programs, Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Seema Khurana
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA.,School of Health Professions, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
4
|
Dum D, Lennartz M, Menz A, Kluth M, Hube-Magg C, Weidemann S, Fraune C, Luebke AM, Hornsteiner L, Bernreuther C, Simon R, Clauditz TS, Sauter G, Uhlig R, Hinsch A, Kind S, Jacobsen F, Möller K, Wilczak W, Steurer S, Minner S, Burandt E, Marx AH, Krech T, Lebok P. Villin expression in human tumours: a tissue microarray study on 14,398 tumours. Expert Rev Mol Diagn 2022; 22:665-675. [PMID: 35866621 DOI: 10.1080/14737159.2022.2104122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Villin is a protein of the brush border of epithelial cells which is used as an immunohistochemical marker for colorectal and gastrointestinal neoplasms. However, other tumor entities can also express villin. METHODS To comprehensively determine villin expression, tissue microarrays containing 14,398 samples from 118 different tumor types as well as 608 samples of 76 different normal tissues were analyzed by immunohistochemistry. RESULTS Villin was found in 54 of 118 tumor categories, including 36 tumor categories with strong staining. Villin expression was frequent in colorectal, upper gastrointestinal tract, pancreatobiliary, and renal tumors as well as in mucinous ovarian cancers, yolk sac tumors and in neuroendocrine neoplasms. Reduced villin expression was linked to advanced pT stage, lymph vessel invasion and microsatellite instability (p≤0.0006) in colorectal adenocarcinoma. In summary, our data demonstrate that villin expression is most common in gastrointestinal, pancreatobiliary, and neuroendocrine neoplasms, yolk sac tumors and mucinous ovarian cancers. CONCLUSION Our data support a high utility of villin immunohistochemistry for the identification of tumors with gastrointestinal, pancreatobiliary, and yolk sac tumor origin. However, considering that at least a weak villin positivity in some tumor cells occurred in 54 different tumor categories, villin immunohistochemistry should be applied as a part of a marker panel rather than as a stand-alone marker.
Collapse
Affiliation(s)
- David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Hornsteiner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Kind
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Pathology, Academic Hospital Fuerth, Fuerth Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Pathology, Academic Hospital Fuerth, Fuerth Germany
| |
Collapse
|
5
|
Association between Immunohistochemistry Markers and Tumor Features and Their Diagnostic and Prognostic Values in Intrahepatic Cholangiocarcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8367395. [PMID: 35529254 PMCID: PMC9071873 DOI: 10.1155/2022/8367395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 12/28/2022]
Abstract
This study investigated the expression of some frequently used immunohistochemistry (IHC) markers. Besides, we evaluated their correlations with the clinical features and outcomes of intrahepatic cholangiocarcinoma (ICC). Patients who underwent surgical removal of the ICC tumors were followed up for 4 years. The paraffin-embedded sections were used to obtain different markers, including CK7, CK19, CK20, CDX2, Glypican3, Hepa1, Ki-67, Villin, and SATB1. Overall survival in relation to IHC marker expression patterns and other clinical characteristics was evaluated by Kaplan-Meier survival curve and log-rank test, followed by the Cox proportional hazard model (to evaluate the relationship between multiple factors and the overall postoperative survival). A total of 122 ICC patients (67 males and 55 females, averagely aged 57.75) were included in this study. There were 44 cases with vascular invasion, 46 cases with lymphatic metastasis, and 13 cases with distant metastasis. CK7 was negatively correlated with lymphatic metastasis; and in distant-metastasis cases, the positive ratio of SATB1 was lower. Interestingly, SATB1 expression indicated a poorer survival, while Villin expression was associated with a better survival. The COX regression analysis showed that female was a protective factor versus male, Villin expression was a strong protective factor, and Ki-67 expression was correlated with a poor survival. Together, IHC markers are associated with tumor features and postoperative survival, especially for SATB1 as a risk factor and Villin as a protective marker, and female ICC patients may have better survival than males.
Collapse
|
6
|
|
7
|
Castro VMDD, Medeiros KCDP, Lemos LICD, Pedrosa LDFC, Ladd FVL, Carvalho TGD, Araújo Júnior RFD, Abreu BJ, Farias NBDS. S-methyl cysteine sulfoxide ameliorates duodenal morphological alterations in streptozotocin-induced diabetic rats. Tissue Cell 2021; 69:101483. [PMID: 33444959 DOI: 10.1016/j.tice.2020.101483] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/08/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus (DM) is a metabolic disease associated with several intestinal disorders. S-methyl cysteine sulfoxide (SMCS) is an amino acid present in Allium cepa L with hypoglycemic effects. However, the effects of SMCS on diabetic intestinal changes are unknown. Thus, we aimed to investigate the effects of SMCS on duodenal morphology and immunomodulatory markers in diabetic rats. Twenty-six rats were divided into three groups: control (C), diabetic (D) and diabetic +200 mg/kg SMCS (DSM). DM was induced by intraperitoneal injection of streptozotocin (50 mg/kg). After 30 days, duodenum samples were processed to assess histopathological and stereological alterations in volume, villus length, and immunohistochemical expression of NF-kB, IL-10, BCL-2, and caspase-3. SMCS reduced hyperglycemia and mitigated the increase in total reference volume of the duodenum, the absolute volume of the mucosa, and the length of the intestinal crypts in the DMS group when compared to D. IL-10 immunostaining was reduced in D when compared to C, while NF-kB was increased in D in comparison to the other groups. SMCS supplementation could decrease the NF-kB immunostaining observed in D. Positive staining for BCL-2 and caspase-3 were not statistically different between groups. In summary, SMCS decreased hyperglycemia and mitigated the morphological changes of the duodenum in diabetic animals, and these beneficial effects can be partially explained by NF-kB modulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bento João Abreu
- Department of Morphology, Federal University of Rio Grande do Norte, Natal, Brazil.
| | | |
Collapse
|
8
|
Cizkova K, Koubova K, Foltynkova T, Jiravova J, Tauber Z. Soluble Epoxide Hydrolase as an Important Player in Intestinal Cell Differentiation. Cells Tissues Organs 2021; 209:177-188. [PMID: 33588415 DOI: 10.1159/000512807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/05/2020] [Indexed: 11/19/2022] Open
Abstract
There is growing evidence that soluble epoxide hydrolase (sEH) may play a role in cell differentiation. sEH metabolizes biologically highly active and generally cytoprotective epoxyeicosatrienoic acids (EETs), generated from arachidonic acid metabolism by CYP epoxygenases (CYP2C and CYP2J subfamilies), to less active corresponding diols. We investigated the effect of sEH inhibitor (TPPU) on the expression of villin, CYP2C8, CYP2C9, CYP2J2, and sEH in undifferentiated and in vitro differentiated HT-29 and Caco2 cell lines. The administration of 10 μM TPPU on differentiated HT-29 and Caco2 cells resulted in a significant decrease in expression of villin, a marker for intestinal cell differentiation. It was accompanied by a disruption of the brush border when microvilli appeared sparse and short in atomic force microscope scans of HT-29 cells. Although inhibition of sEH in differentiated HT-29 and Caco2 cells led to an increase in sEH expression in both cell lines, this treatment had an opposite effect on CYP2J2 expression in HT-29 and Caco2 cells. In addition, tissue samples of colorectal carcinoma and adjacent normal tissues from 45 patients were immunostained for sEH and villin. We detected a significant decrease in the expression of both proteins in colorectal carcinoma in comparison to adjacent normal tissue, and the decrease in both sEH and villin expression revealed a moderate positive association. Taken together, our results showed that sEH is an important player in intestinal cell differentiation.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Katerina Koubova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Tereza Foltynkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Jana Jiravova
- Department of Medical Biophysics, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Zdenek Tauber
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia,
| |
Collapse
|
9
|
Vomhof-DeKrey EE, Lansing JT, Darland DC, Umthun J, Stover AD, Brown C, Basson MD. Loss of Slfn3 induces a sex-dependent repair vulnerability after 50% bowel resection. Am J Physiol Gastrointest Liver Physiol 2021; 320:G136-G152. [PMID: 33237796 PMCID: PMC7864235 DOI: 10.1152/ajpgi.00344.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023]
Abstract
Bowel resection accelerates enterocyte proliferation in the remaining gut with suboptimal absorptive and digestive capacity because of a proliferation-associated decrease in functional differentiation markers. We hypothesized that although schlafen 3 (Slfn3) is an important regulator of enterocytic differentiation, Slfn3 would have less impact on bowel resection adaptation, where accelerated proliferation takes priority over differentiation. We assessed proliferation, cell shedding, and enterocyte differentiation markers from resected and postoperative bowel of wild-type (WT) and Slfn3-knockout (Slfn3KO) mice. Villus length and crypt depth were increased in WT mice and were even longer in Slfn3KO mice. Mitotic marker, Phh3+, and the proliferation markers Lgr5, FoxL1, and platelet-derived growth factor-α (PDGFRα) were increased after resection in male WT, but this was blunted in male Slfn3KO mice. Cell-shedding regulators Villin1 and TNFα were downregulated in female mice and male WT mice only, whereas Gelsolin and EGFR increased expression in all mice. Slfn3 expression increased after resection in WT mice, whereas other Slfn family members 1, 2, 5, 8, and 9 had varied expressions that were affected also by sex difference and loss of Slfn3. Differentiation markers sucrase isomaltase, Dpp4, Glut2, and SGLT1 were all decreased, suggesting that enterocytic differentiation effort is incompatible with rapid proliferation shift in intestinal adaptation. Slfn3 absence potentiates villus length and crypt depth, suggesting that the differentiating stimulus of Slfn3 signaling may restrain mucosal mass increase through regulating Villin1, Gelsolin, EGFR, TNFα, and proliferation markers. Therefore, Slfn3 may be an important regulator not only of "normal" enterocytic differentiation but also in response to bowel resection.NEW & NOTEWORTHY The differentiating stimulus of Slfn3 signaling restrains an increase in mucosal mass after bowel resection, and there is a Slfn3-sex interaction regulating differentiation gene expression and intestinal adaptation. This current study highlights the combinatory effects of gender and Slfn3 genotype on the gene expression changes that contribute to the adaptation in intestinal cellular milleu (i.e. villus and crypt structure) which are utilized to compensate for the stress-healing response that the animals display in intestinal adaptation.
Collapse
Affiliation(s)
- Emilie E Vomhof-DeKrey
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Jack T Lansing
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | - Josey Umthun
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | - Allie D Stover
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Christopher Brown
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Marc D Basson
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| |
Collapse
|
10
|
Involvement of Actin and Actin-Binding Proteins in Carcinogenesis. Cells 2020; 9:cells9102245. [PMID: 33036298 PMCID: PMC7600575 DOI: 10.3390/cells9102245] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
The actin cytoskeleton plays a crucial role in many cellular processes while its reorganization is important in maintaining cell homeostasis. However, in the case of cancer cells, actin and ABPs (actin-binding proteins) are involved in all stages of carcinogenesis. Literature has reported that ABPs such as SATB1 (special AT-rich binding protein 1), WASP (Wiskott-Aldrich syndrome protein), nesprin, and villin take part in the initial step of carcinogenesis by regulating oncogene expression. Additionally, changes in actin localization promote cell proliferation by inhibiting apoptosis (SATB1). In turn, migration and invasion of cancer cells are based on the formation of actin-rich protrusions (Arp2/3 complex, filamin A, fascin, α-actinin, and cofilin). Importantly, more and more scientists suggest that microfilaments together with the associated proteins mediate tumor vascularization. Hence, the presented article aims to summarize literature reports in the context of the potential role of actin and ABPs in all steps of carcinogenesis.
Collapse
|
11
|
Batista VL, da Silva TF, de Jesus LCL, Coelho-Rocha ND, Barroso FAL, Tavares LM, Azevedo V, Mancha-Agresti P, Drumond MM. Probiotics, Prebiotics, Synbiotics, and Paraprobiotics as a Therapeutic Alternative for Intestinal Mucositis. Front Microbiol 2020; 11:544490. [PMID: 33042054 PMCID: PMC7527409 DOI: 10.3389/fmicb.2020.544490] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal mucositis, a cytotoxic side effect of the antineoplastic drug 5-fluorouracil (5-FU), is characterized by ulceration, inflammation, diarrhea, and intense abdominal pain, making it an important issue for clinical medicine. Given the seriousness of the problem, therapeutic alternatives have been sought as a means to ameliorate, prevent, and treat this condition. Among the alternatives available to address this side effect of treatment with 5-FU, the most promising has been the use of probiotics, prebiotics, synbiotics, and paraprobiotics. This review addresses the administration of these "biotics" as a therapeutic alternative for intestinal mucositis caused by 5-FU. It describes the effects and benefits related to their use as well as their potential for patient care.
Collapse
Affiliation(s)
- Viviane Lima Batista
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Tales Fernando da Silva
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Pamela Mancha-Agresti
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Faculdade de Minas, FAMINAS-BH, Belo Horizonte, Brazil
| | - Mariana Martins Drumond
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte, Brazil
| |
Collapse
|
12
|
George SP, Esmaeilniakooshkghazi A, Roy S, Khurana S. F-actin-bundling sites are conserved in proteins with villin-type headpiece domains. Mol Biol Cell 2020; 31:1857-1866. [PMID: 32520642 PMCID: PMC7525818 DOI: 10.1091/mbc.e20-02-0158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/15/2020] [Accepted: 06/04/2020] [Indexed: 11/17/2022] Open
Abstract
Villin is a major actin-bundling protein that assembles the brush border of intestinal and renal epithelial cells. The villin "headpiece" domain and the actin-binding residues within it regulate its actin-bundling function. Substantial experimental and theoretical information about the three-dimensional structure of the isolated villin headpiece, including a description of the actin-binding residues within the headpiece, is available. Despite that, the actin-bundling site in the full-length (FL) villin protein remains unidentified. We used this existing villin headpiece nuclear magnetic resonance data and performed mutational analysis and functional assays to identify the actin-bundling site in FL human villin protein. By careful evaluation of these conserved actin-binding residues in human advillin protein, we demonstrate their functional significance in the over 30 proteins that contain a villin-type headpiece domain. Our study is the first that combines the available structural data on villin headpiece with functional assays to identify the actin-binding residues in FL villin that regulate its filament-bundling activity. Our findings could have wider implications for other actin-bundling proteins that contain a villin-type headpiece domain.
Collapse
Affiliation(s)
- Sudeep P. George
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77044
| | | | - Swati Roy
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77044
| | - Seema Khurana
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77044
- Department of Allied Health, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
13
|
Mouse intestinal tuft cells express advillin but not villin. Sci Rep 2020; 10:8877. [PMID: 32483224 PMCID: PMC7264147 DOI: 10.1038/s41598-020-65469-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 03/06/2020] [Indexed: 12/26/2022] Open
Abstract
Tuft (or brush) cells are solitary chemosensory cells scattered throughout the epithelia of the respiratory and alimentary tract. The actin-binding protein villin (Vil1) is used as a marker of tuft cells and the villin promoter is frequently used to drive expression of the Cre recombinase in tuft cells. While there is widespread agreement about the expression of villin in tuft cells there are several disagreements related to tuft cell lineage commitment and function. We now show that many of these inconsistencies could be resolved by our surprising finding that intestinal tuft cells, in fact, do not express villin protein. Furthermore, we show that a related actin-binding protein, advillin which shares 75% homology with villin, has a tuft cell restricted expression in the gastrointestinal epithelium. Our study identifies advillin as a marker of tuft cells and provides a mechanism for driving gene expression in tuft cells but not in other epithelial cells of the gastrointestinal tract. Our findings fundamentally change the way we identify and study intestinal tuft cells.
Collapse
|
14
|
Tian R, Liu X, Luo Y, Jiang S, Liu H, You F, Zheng C, Wu J. Apoptosis Exerts a Vital Role in the Treatment of Colitis-Associated Cancer by Herbal Medicine. Front Pharmacol 2020; 11:438. [PMID: 32410986 PMCID: PMC7199713 DOI: 10.3389/fphar.2020.00438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/20/2020] [Indexed: 12/24/2022] Open
Abstract
Colitis-associated cancer (CAC) is known as inflammatory bowel disease (IBD)-developed colorectal cancer, the pathogenesis of which involves the occurrence of apoptosis. Western drugs clinically applied to CAC are often single-targeted and exert many adverse reactions after long-term administration, so it is urgent to develop new drugs for the treatment of CAC. Herbal medicines commonly have multiple components with multiple targets, and most of them are low-toxicity. Some herbal medicines have been reported to ameliorate CAC through inducing apoptosis, but there is still a lack of systematic review. In this work, we reviewed articles published in Sci Finder, Web of Science, PubMed, Google Scholar, CNKI, and other databases in recent years by setting the keywords as apoptosis in combination with colitis-associated cancer. We summarized the herbal medicine extracts or their compounds that can prevent CAC by modulating apoptosis and analyzed the mechanism of action. The results show the following. (1) Herbal medicines regulate both the mitochondrial apoptosis pathway and death receptor apoptosis pathway. (2) Herbal medicines modulate the above two apoptotic pathways by affecting signal transductions of IL-6/STAT3, MAPK/NF-κ B, Oxidative stress, Non-canonical TGF-β1, WNT/β-catenin, and Cell cycle, thereby ameliorating CAC. We conclude that following. (1) Studies on the role of herbal medicine in regulating apoptosis through the Ras/Raf/ERK, WNT/β-catenin, and Cell cycle pathways have not yet been carried out in sufficient depth. (2) The active constituents of reported anti-CAC herbal medicine mainly include polyphenols, terpenoids, and saccharide. Also, we identified other herbal medicines with the constituents mentioned above as their main components, aiming to provide a reference for the clinical use of herbal medicine in the treatment of CAC. (3) New dosage forms can be utilized to elevate the targeting and reduce the toxicity of herbal medicine.
Collapse
Affiliation(s)
- Ruimin Tian
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacology, North Sichuan Medical College, Nanchong, China
| | - Xianfeng Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanqin Luo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengnan Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiasi Wu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
The influence of selected gastrointestinal parasites on apoptosis in intestinal epithelial cells. Biomolecules 2020; 10:biom10050674. [PMID: 32349424 PMCID: PMC7277436 DOI: 10.3390/biom10050674] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Studies on the parasite–host interaction may provide valuable information concerning the modulation of molecular mechanisms as well as of the host immune system during infection. To date, it has been demonstrated that intestinal parasites may affect, among others, the processes of digestion in the gastrointestinal system of the host, thus limiting the elimination of the parasite, the immune response as well as inflammation. However, the most recent studies suggest that intestinal parasites may also affect modulation of the apoptosis pathway of the host. The present paper presents the latest scientific information on the influence of intestinal parasite species (Blastocystis sp., Giardia sp., Cryptosporidium sp., Trichuris sp., Entamoeba histolytica, Nippostrongylus brasiliensis, Heligmosomoides polygyrus) on the molecular mechanisms of apoptosis in intestinal epithelial cells. This paper stresses that the interdependency between the intestinal parasite and the host results from the direct effect of the parasite and the host’s defense reactions, which lead to modulation of the apoptosis pathways (intrinsic and extrinsic). Moreover, the present paper presents the role of proteins involved in the mechanisms of apoptosis as well as the physiological role of apoptosis in the host’s intestinal epithelial cells.
Collapse
|
16
|
Verdile N, Pasquariello R, Scolari M, Scirè G, Brevini TAL, Gandolfi F. A Detailed Study of Rainbow Trout ( Onchorhynchus mykiss) Intestine Revealed That Digestive and Absorptive Functions Are Not Linearly Distributed along Its Length. Animals (Basel) 2020; 10:ani10040745. [PMID: 32344584 PMCID: PMC7223369 DOI: 10.3390/ani10040745] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Aquaculture is the fastest growing food-producing sector due to the increase of fish intended for human consumption. However, aquaculture growth generates concerns, since carnivorous fish are extensively fed using fish-meal and fish-oil. This constitutes a severe limit to the aquaculture industry, questioning its sustainability. Consequently, alternative feeds are continuously searched through extensive in vivo feeding trials. Undoubtedly, to evaluate their impact on the gastrointestinal tract health, detailed knowledge of the intestine morphology and physiology is required. To date, extensive studies have been performed in several livestock species; however, available information on fish is limited nowadays, most importantly because their alimentary canal is able to easily adapt to external stimuli, and their intestinal morphology is affected by external factors. Therefore, it is essential to establish accurate reference values, especially along the productive cycle of animals raised in standardized conditions. Here, we performed a detailed characterization of the epithelial cells lining the intestinal mucosa in rainbow trout along the first year of development. We studied the absorptive and secretory activity as well as its ability to self-renewal. Our results indicate that, in this species, both digestive and absorptive functions are not linearly distributed along the intestinal length. Abstract To increase the sustainability of trout farming, the industry requires alternatives to fish-based meals that do not compromise animal health and growth performances. To develop new feeds, detailed knowledge of intestinal morphology and physiology is required. We performed histological, histochemical, immunohistochemical and morphometric analysis at typical time points of in vivo feeding trials (50, 150 and 500 g). Only minor changes occurred during growth whereas differences characterized two compartments, not linearly distributed along the intestine. The first included the pyloric caeca, the basal part of the complex folds and the villi of the distal intestine. This was characterized by a significantly smaller number of goblet cells with smaller mucus vacuoles, higher proliferation and higher apoptotic rate but a smaller extension of fully differentiated epithelial cells and by the presence of numerous pinocytotic vacuolization. The second compartment was formed by the proximal intestine and the apical part of the posterior intestine complex folds. Here we observed more abundant goblet cells with bigger vacuoles, low proliferation rate, few round apoptotic cells, a more extended area of fully differentiated cells and no pinocytotic vacuoles. Our results suggest that rainbow trout intestine is physiologically arranged to mingle digestive and absorptive functions along its length.
Collapse
Affiliation(s)
- Nicole Verdile
- Department of Agricultural and Environmental Sciences, University of Milan, 20133 Milano, Italy;
- Correspondence: (N.V.); (F.G.); Tel.: +39-02-5031-6449 (N.V.); +39-02-5031-7990 (F.G.)
| | - Rolando Pasquariello
- Department of Agricultural and Environmental Sciences, University of Milan, 20133 Milano, Italy;
| | - Marco Scolari
- Skretting Aquaculture Research Centre, 37100 Verona, Italy; (M.S.); (G.S.)
| | - Giulia Scirè
- Skretting Aquaculture Research Centre, 37100 Verona, Italy; (M.S.); (G.S.)
| | - Tiziana A. L. Brevini
- Department of Health, Animal Science and Food Safety, University of Milan, 20133 Milano, Italy;
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences, University of Milan, 20133 Milano, Italy;
- Correspondence: (N.V.); (F.G.); Tel.: +39-02-5031-6449 (N.V.); +39-02-5031-7990 (F.G.)
| |
Collapse
|
17
|
Parker A, Vaux L, Patterson AM, Modasia A, Muraro D, Fletcher AG, Byrne HM, Maini PK, Watson AJM, Pin C. Elevated apoptosis impairs epithelial cell turnover and shortens villi in TNF-driven intestinal inflammation. Cell Death Dis 2019; 10:108. [PMID: 30728350 PMCID: PMC6365534 DOI: 10.1038/s41419-018-1275-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/05/2018] [Accepted: 12/03/2018] [Indexed: 12/27/2022]
Abstract
The intestinal epithelial monolayer, at the boundary between microbes and the host immune system, plays an important role in the development of inflammatory bowel disease (IBD), particularly as a target and producer of pro-inflammatory TNF. Chronic overexpression of TNF leads to IBD-like pathology over time, but the mechanisms driving early pathogenesis events are not clear. We studied the epithelial response to inflammation by combining mathematical models with in vivo experimental models resembling acute and chronic TNF-mediated injury. We found significant villus atrophy with increased epithelial cell death along the crypt-villus axis, most dramatically at the villus tips, in both acute and chronic inflammation. In the acute model, we observed overexpression of TNF receptor I in the villus tip rapidly after TNF injection and concurrent with elevated levels of intracellular TNF and rapid shedding at the tip. In the chronic model, sustained villus atrophy was accompanied by a reduction in absolute epithelial cell turnover. Mathematical modelling demonstrated that increased cell apoptosis on the villus body explains the reduction in epithelial cell turnover along the crypt-villus axis observed in chronic inflammation. Cell destruction in the villus was not accompanied by changes in proliferative cell number or division rate within the crypt. Epithelial morphology and immunological changes in the chronic setting suggest a repair response to cell damage although the villus length is not recovered. A better understanding of how this state is further destabilised and results in clinical pathology resembling IBD will help identify suitable pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Aimée Parker
- Gut Health and Food Safety Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Laura Vaux
- Gut Health and Food Safety Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Angela M Patterson
- Gut Health and Food Safety Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Amisha Modasia
- Gut Health and Food Safety Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | | | - Alexander G Fletcher
- School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom.,Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | | - Carmen Pin
- Gut Health and Food Safety Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom. .,Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom.
| |
Collapse
|
18
|
Space radiation triggers persistent stress response, increases senescent signaling, and decreases cell migration in mouse intestine. Proc Natl Acad Sci U S A 2018; 115:E9832-E9841. [PMID: 30275302 PMCID: PMC6196540 DOI: 10.1073/pnas.1807522115] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Coordinated epithelial cell migration is key to maintaining functional integrity and preventing pathological processes in gastrointestinal tissue, and is essential for astronauts’ health and space mission success. Here we show that energetic heavy ions, which are more prevalent in deep space relative to low-Earth orbit, could persistently decrease intestinal epithelial cell migration, alter cytoskeletal remodeling, and increase cell proliferation with ongoing DNA damage and cell senescence, even a year after irradiation. Our study has provided the molecular underpinnings for energetic heavy-ion 56Fe radiation-induced cell migration alterations, and raises a potentially serious concern, particularly for long-term deep-space manned missions. Proliferative gastrointestinal (GI) tissue is radiation-sensitive, and heavy-ion space radiation with its high-linear energy transfer (high-LET) and higher damaging potential than low-LET γ-rays is predicted to compromise astronauts’ GI function. However, much uncertainty remains in our understanding of how heavy ions affect coordinated epithelial cell migration and extrusion, which are essential for GI homeostasis. Here we show using mouse small intestine as a model and BrdU pulse labeling that cell migration along the crypt–villus axis is persistently decreased after a low dose of heavy-ion 56Fe radiation relative to control and γ-rays. Wnt/β-catenin and its downstream EphrinB/EphB signaling are key to intestinal epithelial cell (IEC) proliferation and positioning during migration, and both are up-regulated after 56Fe radiation. Conversely, factors involved in cell polarity and adhesion and cell–extracellular matrix interactions were persistently down-regulated after 56Fe irradiation—potentially altering cytoskeletal remodeling and cell extrusion. 56Fe radiation triggered a time-dependent increase in γH2AX foci and senescent cells but without a noticeable increase in apoptosis. Some senescent cells acquired the senescence-associated secretory phenotype, and this was accompanied by increased IEC proliferation, implying a role for progrowth inflammatory factors. Collectively, this study demonstrates a unique phenomenon of heavy-ion radiation-induced persistently delayed IEC migration involving chronic sublethal genotoxic and oncogenic stress-induced altered cytoskeletal dynamics, which were seen even a year later. When considered along with changes in barrier function and nutrient absorption factors as well as increased intestinal tumorigenesis, our in vivo data raise a serious concern for long-duration deep-space manned missions.
Collapse
|
19
|
Fu Y, Yu W, Cai H, Lu A. Forecast of actin-binding proteins as the oncotarget in osteosarcoma - a review of mechanism, diagnosis and therapy. Onco Targets Ther 2018; 11:1553-1561. [PMID: 29593421 PMCID: PMC5865567 DOI: 10.2147/ott.s159894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone malignant tumor with a high rate of lung metastasis and principally emerges in children and adolescents. Although neoadjuvant chemotherapy is widely used around the world, a high rate of chemoresistance occurs and frequently generates a poor prognosis. Therefore, finding a new appropriate prognostic marker for OS is a valuable research direction, which will give patients a better chance to receive proper therapy. Actin-binding proteins (ABPs) are a group of proteins that interact with actin cytoskeleton and play a crucial role in the regulation of the cell motility and morphology in eukaryotes. Meanwhile, ABPs also act as a bridge between the cytomembrane and nucleus, which transmit the outside-in and inside-out signals in cytoplasm. Furthermore, ABPs alter the dynamic structure of actin and regulate the invasion and metastasis of cancer. Hence, ABPs have a wide application in predicting the prognosis, and may be new targets, in tumor therapy. This review focuses on a series of ABPs and discusses their modulatory functions. It provides a new insight into the classification of ABPs’ functions in the process of invasion and metastasis in OS and illuminates the potential ability in predicting the prognosis of OS patients.
Collapse
Affiliation(s)
- Yucheng Fu
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Wei Yu
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Hongliu Cai
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Anwei Lu
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
20
|
Patterson AM, Watson AJM. Deciphering the Complex Signaling Systems That Regulate Intestinal Epithelial Cell Death Processes and Shedding. Front Immunol 2017; 8:841. [PMID: 28769935 PMCID: PMC5513916 DOI: 10.3389/fimmu.2017.00841] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/04/2017] [Indexed: 01/16/2023] Open
Abstract
Intestinal epithelial cells play a fundamental role in maintaining homeostasis. Shedding of intestinal cells in a controlled manner is critical to maintenance of barrier function. Barrier function is maintained during this shedding process by a redistribution of tight junctional proteins to facilitate closure of the gap left by the shedding cell. However, despite the obvious importance of epithelial cell shedding to gut health, a central question is how the extrusion of epithelial cells is achieved, enabling barrier integrity to be maintained in the healthy gut and restored during inflammation remains largely unanswered. Recent studies have provided evidence that excessive epithelial cell shedding and loss of epithelial barrier integrity is triggered by exposure to lipopolysaccharide or tumor necrosis factor alpha. Subsequent studies have provided evidence of the involvement of specific cellular components and signaling mechanisms as well as the functionality of microbiota that can be either detrimental or beneficial for intestinal barrier integrity. This review will focus on the evidence and decipher how the signaling systems through which the mucosal immune system and microbiota can regulate epithelial cell shedding and how these mechanisms interact to preserve the viability of the epithelium.
Collapse
Affiliation(s)
- Angela M Patterson
- Quadram Institute, Norwich Research Park, Norwich, United Kingdom.,Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Alastair J M Watson
- Quadram Institute, Norwich Research Park, Norwich, United Kingdom.,Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
21
|
O'Reilly EL, Burchmore RJ, Sparks NH, Eckersall PD. The effect of microbial challenge on the intestinal proteome of broiler chickens. Proteome Sci 2017; 15:10. [PMID: 28572745 PMCID: PMC5450085 DOI: 10.1186/s12953-017-0118-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 05/02/2017] [Indexed: 12/12/2022] Open
Abstract
Background In poultry production intestinal health and function is paramount to achieving efficient feed utilisation and growth. Uncovering the localised molecular mechanisms that occur during the early and important periods of growth that allow birds to grow optimally is important for this species. The exposure of young chicks to used litter from older flocks, containing mixed microbial populations, is a widely utilised model in poultry research. It rarely causes mortality but effects an immunogenic stimulation sufficient enough to cause reduced and uneven growth that is reflective of a challenging growing environment. Methods A mixed microbial challenge was delivered as used litter containing Campylobacter jejuni and coccidial oocysts to 120 male Ross 308 broiler chicks, randomly divided into two groups: control and challenged. On day 12, 15, 18 and 22 (pre- and 3, 6 and 10 days post-addition of the used litter) the proximal jejunum was recovered from 6 replicates per group and differentially abundant proteins identified between groups and over time using 2D DiGE. Results The abundance of cytoskeletal proteins of the chicken small intestinal proteome, particularly actin and actin associated proteins, increased over time in both challenged and control birds. Villin-1, an actin associated anti-apoptotic protein, was reduced in abundance in the challenged birds indicating that many of the changes in cytoskeletal protein abundance in the challenged birds were as a result of an increased rate of apoptosis. A number of heat shock proteins decreased in abundance over time in the intestine and this was more pronounced in the challenged birds. Conclusions The small intestinal proteome sampled from 12 to 22 days of age showed considerable developmental change, comparable to other species indicating that many of the changes in protein abundance in the small intestine are conserved among vertebrates. Identifying and distinguishing the changes in proteins abundance and molecular pathways that occur as a result of normal growth from those that occur as a result of a challenging microbial environment is important in this major food producing animal.
Collapse
Affiliation(s)
- Emily L O'Reilly
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, Glasgow University, Glasgow, UK
| | - Richard J Burchmore
- Glasgow Polyomics Facility, College of Medical, Veterinary and Life Sciences, Glasgow University, Glasgow, UK
| | - Nicholas H Sparks
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Easter Bush, Midlothian, EH25 9RG UK
| | - P David Eckersall
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, Glasgow University, Glasgow, UK
| |
Collapse
|