151
|
Wang L, Yang X, Zhou S, Lyu T, Shi L, Dong Y, Zhang H. Comparative transcriptome analysis revealed omnivorous adaptation of the small intestine of Melinae. Sci Rep 2021; 11:19162. [PMID: 34580368 PMCID: PMC8476558 DOI: 10.1038/s41598-021-98561-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/09/2021] [Indexed: 01/04/2023] Open
Abstract
As the main digestive organ, the small intestine plays a vital role in the digestion of animals. At present, most of the research on animal feeding habits focuses on carnivores and herbivores. However, the mechanism of feeding and digestion in omnivores remains unclear. This study aims to reveal the molecular basis of the omnivorous adaptive evolution of Melinae by comparing the transcriptome of the small intestines of Asian Badgers (Meles leucurus) and Northern Hog Badgers (Arctonyx albogularis). We obtained high-quality small intestinal transcriptome data from these two species. Key genes and signalling pathways were analysed through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and other databases. Research has mainly found that orthologous genes related to six enzymes have undergone adaptive evolution. In addition, the study also found three digestion-related pathways (cGMP-PKG, cAMP, and Hippo). They are related to the digestion and absorption of nutrients, the secretion of intestinal fluids, and the transport of food through the small intestine, which may help omnivorous animals adapt to an omnivorous diet. Our study provides insight into the adaptation of Melinae to omnivores and affords a valuable transcriptome resource for future research.
Collapse
Affiliation(s)
| | | | | | | | - Lupeng Shi
- Qufu Normal University, Qufu, 273165, China
| | | | | |
Collapse
|
152
|
Female developmental environment delays development of male honeybee (Apis mellifera). BMC Genomics 2021; 22:699. [PMID: 34579651 PMCID: PMC8477528 DOI: 10.1186/s12864-021-08014-1] [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: 07/12/2020] [Accepted: 09/15/2021] [Indexed: 11/10/2022] Open
Abstract
Background Nutrition and cell size play an important role in the determination of caste differentiation in queen and worker of honeybees (Apis mellifera), whereas the haploid genome dominates the differentiation of drones. However, the effects of female developmental environment on the development of males remain unclear. In this study, young drone larvae were transferred into worker cells (WCs) or remained in drone cells (DCs) to rear drones. The drone larvae were also grafted into queen cells (QCs) for 48 h and then transplanted into drone cells until emerging. Morphological indexes and reproductive organs of these three types of newly emerged drones were measured. Newly emerged drones and third instar drone larvae from WCs, DCs and QCs were sequenced by RNA sequencing (RNA-Seq). Results The amount of food remaining in cells of the QC and WC groups was significantly different to that in the DC group at the early larval stage. Morphological results showed that newly emerged DC drones had bigger body sizes and more well-developed reproductive tissues than WC and QC drones, whereas the reproductive tissues of QC drones were larger than those of WC drones. Additionally, whole body gene expression results showed a clear difference among three groups. At larval stage there were 889, 1761 and 1927 significantly differentially expressed genes (DEGs) in WC/DC, QC/DC and WC/QC comparisons, respectively. The number of DEGs decreased in adult drones of these three comparisons [678 (WC/DC), 338 (QC/DC) and 518 (WC/QC)]. A high number of DEGs were involved in sex differentiation, growth, olfaction, vision, mammalian target of rapamycin (mTOR), Wnt signaling pathways, and other processes. Conclusions This study demonstrated that the developmental environment of honeybee females can delay male development, which may serve as a model for understanding the regulation of sex differentiation and male development in social insects by environmental factors. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08014-1.
Collapse
|
153
|
Kim SY, Park SY, Jang HS, Park YD, Kee SH. Yes-Associated Protein Is Required for ZO-1-Mediated Tight-Junction Integrity and Cell Migration in E-Cadherin-Restored AGS Gastric Cancer Cells. Biomedicines 2021; 9:biomedicines9091264. [PMID: 34572450 PMCID: PMC8467433 DOI: 10.3390/biomedicines9091264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 01/04/2023] Open
Abstract
Yes-associated protein (YAP) regulates numerous cellular homeostasis processes and malignant transformation. We found that YAP influences ZO-1-mediated cell migration using E-cadherin-restored EC96 cells derived from gastric malignant AGS cells. Ectopic expression of E-cadherin enhanced straightforward migration of cells, in comparison to the meandering movement of parental AGS cells. In EC96 cells, YAP and ZO-1 expression increased but nuclear YAP levels and activity were reduced. Nuclear factor-κB (NF-κB) mediated the increase in ZO-1 expression, possibly stabilizing cytoplasmic YAP post-translationally. Downregulation of YAP expression using siYAP RNA or stable knock-down inhibited straightforward cell migration by fragmenting ZO-1 containing tight junctions (TJs) but not adherens junctions, implying involvement of YAP in ZO-1-mediated cell migration. The association of YAP with ZO-1 was mediated by angiomotin (AMOT) because downregulation of AMOT dissociated YAP from ZO-1 and reduced cell migration. E-cadherin restoration in malignant cancer cells induced NF-κB signaling to enhance ZO-1 expression and subsequently stabilize YAP. At high expression levels, YAP associates with ZO-1 via AMOT at TJs, influencing ZO-1-mediated cell migration and maintaining TJ integrity.
Collapse
Affiliation(s)
- Seon-Young Kim
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.K.); (S.-Y.P.)
| | - Song-Yi Park
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.K.); (S.-Y.P.)
| | - Hwan-Seok Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (H.-S.J.); (Y.-D.P.)
| | - Yong-Doo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (H.-S.J.); (Y.-D.P.)
| | - Sun-Ho Kee
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.K.); (S.-Y.P.)
- Correspondence: ; Tel.: +82-2-2286-1460
| |
Collapse
|
154
|
Park H, Furtado J, Poulet M, Chung M, Yun S, Lee S, Sessa WC, Franco CA, Schwartz MA, Eichmann A. Defective Flow-Migration Coupling Causes Arteriovenous Malformations in Hereditary Hemorrhagic Telangiectasia. Circulation 2021; 144:805-822. [PMID: 34182767 PMCID: PMC8429266 DOI: 10.1161/circulationaha.120.053047] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/15/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Activin receptor-like kinase 1 (ALK1) is an endothelial transmembrane serine threonine kinase receptor for BMP family ligands that plays a critical role in cardiovascular development and pathology. Loss-of-function mutations in the ALK1 gene cause type 2 hereditary hemorrhagic telangiectasia, a devastating disorder that leads to arteriovenous malformations. Here, we show that ALK1 controls endothelial cell polarization against the direction of blood flow and flow-induced endothelial migration from veins through capillaries into arterioles. METHODS Using Cre lines that recombine in different subsets of arterial, capillary-venous, or endothelial tip cells, we show that capillary-venous Alk1 deletion was sufficient to induce arteriovenous malformation formation in the postnatal retina. RESULTS ALK1 deletion impaired capillary-venous endothelial cell polarization against the direction of blood flow in vivo and in vitro. Mechanistically, ALK1-deficient cells exhibited increased integrin signaling interaction with vascular endothelial growth factor receptor 2, which enhanced downstream YAP/TAZ nuclear translocation. Pharmacologic inhibition of integrin or YAP/TAZ signaling rescued flow migration coupling and prevented vascular malformations in Alk1-deficient mice. CONCLUSIONS Our study reveals ALK1 as an essential driver of flow-induced endothelial cell migration and identifies loss of flow-migration coupling as a driver of arteriovenous malformation formation in hereditary hemorrhagic telangiectasia disease. Integrin-YAP/TAZ signaling blockers are new potential targets to prevent vascular malformations in patients with hereditary hemorrhagic telangiectasia.
Collapse
Affiliation(s)
- Hyojin Park
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
| | - Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
| | - Mathilde Poulet
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
| | - Minhwan Chung
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
| | - Sanguk Yun
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
| | - Sungwoon Lee
- Yale University School of Medicine, Department of Pharmacology, New Haven CT, USA
| | - William C Sessa
- Yale University School of Medicine, Department of Pharmacology, New Haven CT, USA
| | - Claudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Martin A Schwartz
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Yale University School of Medicine, Departments of Cell Biology and Biomedical Engineering, New Haven CT, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Yale University School of Medicine, Department of Molecular and Cellular Physiology, New Haven, CT, USA
- Université de Paris, PARCC, INSERM, F-75006 Paris, France
| |
Collapse
|
155
|
Shi T, Zhu Y, Liu P, Ye L, Jiang X, Cao H, Yu L. Age and Behavior-Dependent Differential miRNAs Expression in the Hypopharyngeal Glands of Honeybees ( Apis mellifera L.). INSECTS 2021; 12:insects12090764. [PMID: 34564204 PMCID: PMC8466209 DOI: 10.3390/insects12090764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 12/22/2022]
Abstract
This study aims to investigate the expression differences of miRNAs in the hypopharyngeal glands (HPGs) of honeybees at three developmental stages and to explore their regulation functions in the HPGs development. Small RNA sequencing was employed to analyze the miRNA profiles of HPGs in newly-emerged bees (NEB), nurse bees (NB), and forager bees (FB). Results showed that a total of 153 known miRNAs were found in the three stages, and ame-miR-276-3p, ame-miR-375-3p, ame-miR-14-3p, ame-miR-275-3p, and ame-miR-3477-5p were the top five most abundant ones. Furthermore, the expression of 11 miRNAs, 17 miRNAs, and 18 miRNAs were significantly different in NB vs. FB comparison, NB vs. NEB comparison, and in FB vs. NEB comparison, respectively, of which ame-miR-184-3p and ame-miR-252a-5p were downregulated in NB compared with that in both the FB and NEB, while ame-miR-11-3p, ame-miR-281-3p, and ame-miR-31a-5p had lower expression levels in FB compared with that in both the NB and NEB. Bioinformatic analysis showed that the potential target genes of the differentially expressed miRNAs (DEMs) were mainly enriched in several key signaling pathways, including mTOR signaling pathway, MAPK signaling pathway-fly, FoxO signaling pathway, Hippo signaling pathway-fly. Overall, our study characterized the miRNA profiles in the HPGs of honeybees at three different developmental stages and provided a basis for further study of the roles of miRNAs in HPGs development.
Collapse
Affiliation(s)
- Tengfei Shi
- School of Plant Protection, Anhui Agricultural University, Hefei 230036, China; (T.S.); (P.L.); (L.Y.); (X.J.); (H.C.)
| | - Yujie Zhu
- School of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China;
| | - Peng Liu
- School of Plant Protection, Anhui Agricultural University, Hefei 230036, China; (T.S.); (P.L.); (L.Y.); (X.J.); (H.C.)
| | - Liang Ye
- School of Plant Protection, Anhui Agricultural University, Hefei 230036, China; (T.S.); (P.L.); (L.Y.); (X.J.); (H.C.)
| | - Xingchuan Jiang
- School of Plant Protection, Anhui Agricultural University, Hefei 230036, China; (T.S.); (P.L.); (L.Y.); (X.J.); (H.C.)
| | - Haiqun Cao
- School of Plant Protection, Anhui Agricultural University, Hefei 230036, China; (T.S.); (P.L.); (L.Y.); (X.J.); (H.C.)
| | - Linsheng Yu
- School of Plant Protection, Anhui Agricultural University, Hefei 230036, China; (T.S.); (P.L.); (L.Y.); (X.J.); (H.C.)
- Correspondence:
| |
Collapse
|
156
|
Mak T, Jones AW, Nurse P. The TOR-dependent phosphoproteome and regulation of cellular protein synthesis. EMBO J 2021; 40:e107911. [PMID: 34296454 PMCID: PMC8365262 DOI: 10.15252/embj.2021107911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/28/2021] [Accepted: 06/09/2021] [Indexed: 01/21/2023] Open
Abstract
Cell growth is orchestrated by a number of interlinking cellular processes. Components of the TOR pathway have been proposed as potential regulators of cell growth, but little is known about their immediate effects on protein synthesis in response to TOR-dependent growth inhibition. Here, we present a resource providing an in-depth characterisation of Schizosaccharomyces pombe phosphoproteome in relation to changes observed in global cellular protein synthesis upon TOR inhibition. We find that after TOR inhibition, the rate of protein synthesis is rapidly reduced and that notable phosphorylation changes are observed in proteins involved in a range of cellular processes. We show that this reduction in protein synthesis rates upon TOR inhibition is not dependent on S6K activity, but is partially dependent on the S. pombe homologue of eIF4G, Tif471. Our study demonstrates the impact of TOR-dependent phospho-regulation on the rate of protein synthesis and establishes a foundational resource for further investigation of additional TOR-regulated targets both in fission yeast and other eukaryotes.
Collapse
Affiliation(s)
- Tiffany Mak
- Cell Cycle LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKgs. LyngbyDenmark
| | - Andrew W Jones
- Cell Cycle LaboratoryThe Francis Crick InstituteLondonUK
- Protein Analysis and Proteomics PlatformThe Francis Crick InstituteLondonUK
| | - Paul Nurse
- Cell Cycle LaboratoryThe Francis Crick InstituteLondonUK
- Laboratory of Yeast Genetics and Cell BiologyRockefeller UniversityNew YorkNYUSA
| |
Collapse
|
157
|
Yuan T, Annamalai K, Naik S, Lupse B, Geravandi S, Pal A, Dobrowolski A, Ghawali J, Ruhlandt M, Gorrepati KDD, Azizi Z, Lim DS, Maedler K, Ardestani A. The Hippo kinase LATS2 impairs pancreatic β-cell survival in diabetes through the mTORC1-autophagy axis. Nat Commun 2021; 12:4928. [PMID: 34389720 PMCID: PMC8363615 DOI: 10.1038/s41467-021-25145-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetes results from a decline in functional pancreatic β-cells, but the molecular mechanisms underlying the pathological β-cell failure are poorly understood. Here we report that large-tumor suppressor 2 (LATS2), a core component of the Hippo signaling pathway, is activated under diabetic conditions and induces β-cell apoptosis and impaired function. LATS2 deficiency in β-cells and primary isolated human islets as well as β-cell specific LATS2 ablation in mice improves β-cell viability, insulin secretion and β-cell mass and ameliorates diabetes development. LATS2 activates mechanistic target of rapamycin complex 1 (mTORC1), a physiological suppressor of autophagy, in β-cells and genetic and pharmacological inhibition of mTORC1 counteracts the pro-apoptotic action of activated LATS2. We further show a direct interplay between Hippo and autophagy, in which LATS2 is an autophagy substrate. On the other hand, LATS2 regulates β-cell apoptosis triggered by impaired autophagy suggesting an existence of a stress-sensitive multicomponent cellular loop coordinating β-cell compensation and survival. Our data reveal an important role for LATS2 in pancreatic β-cell turnover and suggest LATS2 as a potential therapeutic target to improve pancreatic β-cell survival and function in diabetes.
Collapse
Affiliation(s)
- Ting Yuan
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Karthika Annamalai
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Shruti Naik
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Blaz Lupse
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Shirin Geravandi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Anasua Pal
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Jaee Ghawali
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Marina Ruhlandt
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Zahra Azizi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dae-Sik Lim
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.
| | - Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
158
|
Nieto-Torres JL, Shanahan SL, Chassefeyre R, Chaiamarit T, Zaretski S, Landeras-Bueno S, Verhelle A, Encalada SE, Hansen M. LC3B phosphorylation regulates FYCO1 binding and directional transport of autophagosomes. Curr Biol 2021; 31:3440-3449.e7. [PMID: 34146484 PMCID: PMC8439105 DOI: 10.1016/j.cub.2021.05.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 01/07/2021] [Accepted: 05/25/2021] [Indexed: 01/22/2023]
Abstract
Macroautophagy (hereafter referred to as autophagy) is a conserved process that promotes cellular homeostasis through the degradation of cytosolic components, also known as cargo. During autophagy, cargo is sequestered into double-membrane vesicles called autophagosomes, which are predominantly transported in the retrograde direction to the perinuclear region to fuse with lysosomes, thus ensuring cargo degradation.1 The mechanisms regulating directional autophagosomal transport remain unclear. The ATG8 family of proteins associates with autophagosome membranes2 and plays key roles in autophagy, including the movement of autophagosomes. This is achieved via the association of ATG8 with adaptor proteins like FYCO1, involved in the anterograde transport of autophagosomes toward the cell periphery.1,3-5 We previously reported that phosphorylation of LC3B/ATG8 on threonine 50 (LC3B-T50) by the Hippo kinase STK4/MST1 is required for autophagy through unknown mechanisms.6 Here, we show that STK4-mediated phosphorylation of LC3B-T50 reduces the binding of FYCO1 to LC3B. In turn, impairment of LC3B-T50 phosphorylation decreases starvation-induced perinuclear positioning of autophagosomes as well as their colocalization with lysosomes. Moreover, a significantly higher number of LC3B-T50A-positive autophagosomes undergo aberrant anterograde movement to axonal tips in mammalian neurons and toward the periphery of mammalian cells. Our data support a role of a nutrient-sensitive STK4-LC3B-FYCO1 axis in the regulation of the directional transport of autophagosomes, a key step of the autophagy process, via the post-translational modification of LC3B.
Collapse
Affiliation(s)
- Jose L Nieto-Torres
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sean-Luc Shanahan
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Romain Chassefeyre
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tai Chaiamarit
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sviatlana Zaretski
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sara Landeras-Bueno
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adriaan Verhelle
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sandra E Encalada
- Department of Molecular Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Malene Hansen
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
159
|
Gharehdaghi L, Bakhtiarizadeh MR, He K, Harkinezhad T, Tahmasbi G, Li F. Diet-derived transmission of MicroRNAs from host plant into honey bee Midgut. BMC Genomics 2021; 22:587. [PMID: 34344297 PMCID: PMC8336336 DOI: 10.1186/s12864-021-07916-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/22/2021] [Indexed: 11/24/2022] Open
Abstract
Background MicroRNA (miRNA) is a class of small noncoding RNAs, which targets on thousands of mRNA and thus plays important roles in many biological processes. It has been reported that miRNA has cross-species regulation functions between parasitoid-host, or plant-animal, etc. For example, several plant miRNAs enter into the honey bees and regulate gene expression. However, whether cross-species regulation function of miRNAs is a universal mechanism remains a debate question. Results We have evaluated transmission of miRNAs from sunflower and sedr plants into the midgut of honey bee using RNA-Seq analyses complemented with confirmation by RT-qPCR. The results showed that at least 11 plant miRNAs were found in the midgut of honey bee feeding by sunflower and sedr pollen. Among which, nine miRNAs, including miR-30d, miR-143, miR-148a, miR-21, let-7 g, miR-26a, miR-126, miR-27a, and miR-203, were shared between the sunflower- and sedr-fed honey bees, suggesting they might have essential roles in plant-insect interactions. Moreover, existence of these co-shared miRNAs presents a strong evidence to support the successful transmission of miRNAs into the midgut of the insect. In total, 121 honeybee mRNAs were predicted to be the target of these 11 plant-derived miRNAs. Interestingly, a sedr-derived miRNA, miR-206, targets on 53 honeybee genes. Kyoto Encyclopedia of Genes and Genome (KEGG) analyses showed that these target genes are significantly involved in hippo signaling pathway-fly, Wnt signaling pathway, and N-Glycan biosynthesis. Conclusions In summary, these results provide evidence of cross-species regulation function of miRNA between honeybee and flowering host plants, extending our understanding of the molecular interactions between plants and animals. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07916-4.
Collapse
Affiliation(s)
- Leila Gharehdaghi
- Department of Animal Science, Faculty of Agriculture, University of Zanjan, Zanjan, Iran
| | | | - Kang He
- Ministry of Agriculture and Rural Affairs Key Lab of Molecular Biology of Crop Pathogens and Insects/Institute of Insect Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Taher Harkinezhad
- Department of Animal Science, Faculty of Agriculture, University of Zanjan, Zanjan, Iran
| | - Gholamhosein Tahmasbi
- Department of Honeybee, Agricultural Research, Education and Extension Organization (AREEO), Animal Science Research Institute of Iran, Karaj, Iran
| | - Fei Li
- Ministry of Agriculture and Rural Affairs Key Lab of Molecular Biology of Crop Pathogens and Insects/Institute of Insect Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China.
| |
Collapse
|
160
|
Integrated Analysis of lncRNA and mRNA in Subcutaneous Adipose Tissue of Ningxiang Pig. BIOLOGY 2021; 10:biology10080726. [PMID: 34439958 PMCID: PMC8389317 DOI: 10.3390/biology10080726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022]
Abstract
Simple Summary This study shows the transcription profiles and the functional network in lncRNA and mRNA in the subcutaneous adipose tissue of Ningxiang piglets in four stages of development (piglets, nursery pigs, early fattening, and late fattening). A total of 2872 novel lncRNAs have now been determined. A total of 10,084 DEmRNAs and 931 DElncRNAs were determined. Interestingly, most DEmRNAs were up-regulated in the piglet stage and, in contrast, DElncRNAs were up-regulated in the late fattening stage. A complicated interaction between mRNAs and lncRNAs was determined via STEM and WGCNA, demonstrating that lncRNAs are an essential regulatory component in mRNAs. Modules 2 and 5 shows a similar mode of transcriptions for both mRNA and lncRNA, which are mainly involved in steroid biosynthesis, glycosphingolipid biosynthesis, metabolic pathways, and glycerolipid metabolism. The transcription levels of mRNAs and lncRNAs for both modules were higher in the early and late fattening stage. This may be explained by the active fatty acids, sterols, steroids, and lipid-related metabolic activity in the subcutaneous adipose tissue during the early and late fattening stage. Abstract Ningxiang pigs, a Chinese bred pig known for its tender meat and high quality unsaturated fatty acids. This study discovers the transcription profiles and functional networks in long non-coding RNA (lncRNA) and messenger RNA (mRNA) in subcutaneous adipose tissue. Subcutaneous adipose tissue was collected from piglet, nursery pig, early fattening, and late fattening stage of Ningxiang piglets, and lncRNA and mRNA transcription of each stage was profiled. A total of 339,204,926 (piglet), 315,609,246 (nursery), 266,798,202 (early fattening), and 343,740,308 (late fattening) clean reads were generated, and 2872 novel lncRNAs were identified. Additionally, 10,084 differential mRNAs (DEmRNAs) and 931 differential lncRNAs were determined. Most DEmRNAs were up-regulated in the piglet stage, while they were down-regulated in late fattening stage. A complicated interaction between mRNAs and lncRNAs was identified via STEM and WGCNA, demonstrated that lncRNAs are a significant regulatory component in mRNAs. The findings showed that modules 2 and 5 have a similar mode of transcription for both mRNA and lncRNA, and were mainly participated in steroid biosynthesis, glycosphingolipid biosynthesis, metabolic pathways, and glycerolipid metabolism. The mRNAs and lncRNAs transcription levels of both modules was higher in the early and late fattening stage, which may be due to the active activity of the metabolism in relation to fatty acids, sterols, steroids, and lipids in the subcutaneous adipose tissue during the early and late fattening stage. These findings could be expected to result in further research of the functional properties of lncRNA from subcutaneous adipose tissue at different stages of development in Ningxiang pigs.
Collapse
|
161
|
Zhou Z, Yuan Z, Hong W, Wang W. Editorial: A Hippo's View: From Molecular Basis to Translational Medicine. Front Cell Dev Biol 2021; 9:729155. [PMID: 34368170 PMCID: PMC8343012 DOI: 10.3389/fcell.2021.729155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (ASTAR), Singapore, Singapore
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
162
|
Li FL, Guan KL. The two sides of Hippo pathway in cancer. Semin Cancer Biol 2021; 85:33-42. [PMID: 34265423 DOI: 10.1016/j.semcancer.2021.07.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 02/08/2023]
Abstract
The Hippo signaling pathway was originally characterized by genetic studies in Drosophila to regulate tissue growth and organ size, and the core components of this pathway are highly conserved in mammals. Studies over the past two decades have revealed critical physiological and pathological functions of the Hippo tumor-suppressor pathway, which is tightly regulated by a broad range of intracellular and extracellular signals. These properties enable the Hippo pathway to serve as an important controller in organismal development and adult tissue homeostasis. Dysregulation of the Hippo signaling has been observed in many cancer types, suggesting the possibility of cancer treatment by targeting the Hippo pathway. The general consensus is that Hippo has tumor suppressor function. However, growing evidence also suggests that the function of the Hippo pathway in malignancy is cancer context dependent as recent studies indicating tumor promoting function of LATS. This article surveys the Hippo pathway signaling mechanisms and then reviews both the tumor suppressing and promoting function of this pathway. A comprehensive understanding of the dual roles of the Hippo pathway in cancer will benefit future therapeutic targeting of the Hippo pathway for cancer treatment.
Collapse
Affiliation(s)
- Fu-Long Li
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Kun-Liang Guan
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
163
|
Fernandez-Guerrero M, Zdral S, Castilla-Ibeas A, Lopez-Delisle L, Duboule D, Ros MA. Time-sequenced transcriptomes of developing distal mouse limb buds: A comparative tissue layer analysis. Dev Dyn 2021; 251:1550-1575. [PMID: 34254395 DOI: 10.1002/dvdy.394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The development of the amniote limb has been an important model system to study patterning mechanisms and morphogenesis. For proper growth and patterning, it requires the interaction between the distal sub-apical mesenchyme and the apical ectodermal ridge (AER) that involve the separate implementation of coordinated and tissue-specific genetic programs. RESULTS Here, we produce and analyze the transcriptomes of both distal limb mesenchymal progenitors and the overlying ectodermal cells, following time-coursed dissections that cover from limb bud initiation to fully patterned limbs. The comparison of transcriptomes within each layer as well as between layers over time, allowed the identification of specific transcriptional signatures for each of the developmental stages. Special attention was given to the identification of genes whose transcription dynamics suggest a previously unnoticed role in the context of limb development and also to signaling pathways enriched between layers. CONCLUSION We interpret the transcriptomic data in light of the known development pattern and we conclude that a major transcriptional transition occurs in distal limb buds between E9.5 and E10.5, coincident with the switch from an early phase continuation of the signature of trunk progenitors, related to the initial proximo distal specification, to a late intrinsic phase of development.
Collapse
Affiliation(s)
- Marc Fernandez-Guerrero
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-University of Cantabria-SODERCAN), Santander, Spain
| | - Sofia Zdral
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-University of Cantabria-SODERCAN), Santander, Spain
| | - Alejandro Castilla-Ibeas
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-University of Cantabria-SODERCAN), Santander, Spain
| | | | - Denis Duboule
- School of Life Sciences, Federal Institute of Technology, Lausanne, Switzerland.,Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Collège de France, Paris, France
| | - Marian A Ros
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-University of Cantabria-SODERCAN), Santander, Spain.,Facultad de Medicina, Departamento de Anatomía y Biología Celular, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
164
|
Takeda T, Tsubaki M, Genno S, Matsuda T, Yamamoto Y, Kimura A, Shimizu N, Nishida S. Inhibition of yes-associated protein suppresses migration, invasion, and metastasis in non-small cell lung cancer in vitro and in vivo. Clin Exp Med 2021; 22:221-228. [PMID: 34196881 DOI: 10.1007/s10238-021-00738-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is a highly aggressive cancer with one of the most prevalent malignant tumors. Metastasis in NSCLC is the major cause of treatment failure and cancer-related deaths. Yes-associated protein (YAP) is a transcriptional coactivator regulated by the evolutionarily conserved Hippo signaling pathway that regulates organ size, growth, and regeneration. YAP is highly expressed in several malignant tumor types. Furthermore, YAP promotes tumor initiation and/or progression in various types of cancer. However, it is unclear whether YAP contributes to the metastasis in NSCLC and serves as a useful therapeutic target. Here, we investigated whether levels of YAP correlate with metastatic phenotype in NSCLC cells and serve as a useful therapeutic target. We found that high levels of YAP associate with high cell migration, invasion, and metastasis in NSCLC cell lines. Furthermore, YAP siRNA decreased the migration and invasion in NSCLC cells. Additionally, verteporfin, an agent used for the treatment of symptomatic polypoidal choroidal vasculopathy, decreased the expression of YAP and inhibited migration, invasion, and metastasis in NSCLC cells. Thus, the study suggests that targeting YAP may present a new avenue to develop therapeutics against metastasis in NSCLC and that verteporfin has potential molecular therapeutic strategy for the treatment of metastatic NSCLC.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Shuji Genno
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yuuta Yamamoto
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Akihiro Kimura
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Nao Shimizu
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
165
|
Pojer JM, Saiful Hilmi AJ, Kondo S, Harvey KF. Crumbs and the apical spectrin cytoskeleton regulate R8 cell fate in the Drosophila eye. PLoS Genet 2021; 17:e1009146. [PMID: 34097697 PMCID: PMC8211197 DOI: 10.1371/journal.pgen.1009146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 06/17/2021] [Accepted: 05/11/2021] [Indexed: 12/31/2022] Open
Abstract
The Hippo pathway is an important regulator of organ growth and cell fate. In the R8 photoreceptor cells of the Drosophila melanogaster eye, the Hippo pathway controls the fate choice between one of two subtypes that express either the blue light-sensitive Rhodopsin 5 (Hippo inactive R8 subtype) or the green light-sensitive Rhodopsin 6 (Hippo active R8 subtype). The degree to which the mechanism of Hippo signal transduction and the proteins that mediate it are conserved in organ growth and R8 cell fate choice is currently unclear. Here, we identify Crumbs and the apical spectrin cytoskeleton as regulators of R8 cell fate. By contrast, other proteins that influence Hippo-dependent organ growth, such as the basolateral spectrin cytoskeleton and Ajuba, are dispensable for the R8 cell fate choice. Surprisingly, Crumbs promotes the Rhodopsin 5 cell fate, which is driven by Yorkie, rather than the Rhodopsin 6 cell fate, which is driven by Warts and the Hippo pathway, which contrasts with its impact on Hippo activity in organ growth. Furthermore, neither the apical spectrin cytoskeleton nor Crumbs appear to regulate the Hippo pathway through mechanisms that have been observed in growing organs. Together, these results show that only a subset of Hippo pathway proteins regulate the R8 binary cell fate decision and that aspects of Hippo signalling differ between growing organs and post-mitotic R8 cells.
Collapse
Affiliation(s)
- Jonathan M. Pojer
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdul Jabbar Saiful Hilmi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Shu Kondo
- Laboratory of Invertebrate Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Kieran F. Harvey
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
166
|
Shi W, Wang Q, Wang J, Yan X, Feng W, Zhang Q, Zhai C, Chai L, Li S, Xie X, Li M. Activation of yes-associated protein mediates sphingosine-1-phosphate-induced proliferation and migration of pulmonary artery smooth muscle cells and its potential mechanisms. J Cell Physiol 2021; 236:4694-4708. [PMID: 33283886 DOI: 10.1002/jcp.30193] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/30/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
The aims of the present study were to examine the molecular mechanisms underlying sphingosine-1-phosphate (S1P)-induced rat pulmonary artery smooth muscle cells (PASMCs) proliferation/migration and to determine the effect of yes-associated protein (YAP) activation on S1P-induced PASMCs proliferation/migration and its potential mechanisms. S1P induced YAP dephosphorylation and nuclear translocation, upregulated microRNA-130a/b (miR-130a/b) expression, reduced bone morphogenetic protein receptor 2 (BMPR2), and inhibitor of DNA binding 1(Id1) expression, and promoted PASMCs proliferation and migration. Pretreatment of cells with Rho-associated protein kinase (ROCK) inhibitor Y27632 suppressed S1P-induced YAP activation, miR-130a/b upregulation, BMPR2/Id1 downregulation, and PASMCs proliferation/migration. Knockdown of YAP using small interfering RNA also suppressed S1P-induced alterations of miR-130a/b, BMPR2, Id1, and PASMCs behavior. In addition, luciferase reporter assay indicated that miR-130a/b directly regulated BMPR2 expression in PASMCs. Inhibition of miR-130a/b functions by anti-miRNA oligonucleotides attenuated S1P-induced BMPR2/Id1 downregulation and the proliferation and migration of PASMCs. Taken together, our study indicates that S1P induces activation of YAP through ROCK signaling and subsequently increases miR-130a/b expression, which, in turn, downregulates BMPR2 and Id1 leading to PASMCs proliferation and migration.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Inhibitor of Differentiation Protein 1/metabolism
- Intracellular Signaling Peptides and Proteins/metabolism
- Lysophospholipids/pharmacology
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- YAP-Signaling Proteins
- rho-Associated Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| |
Collapse
|
167
|
Mohajan S, Jaiswal PK, Vatanmakarian M, Yousefi H, Sankaralingam S, Alahari SK, Koul S, Koul HK. Hippo pathway: Regulation, deregulation and potential therapeutic targets in cancer. Cancer Lett 2021; 507:112-123. [PMID: 33737002 PMCID: PMC10370464 DOI: 10.1016/j.canlet.2021.03.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/25/2023]
Abstract
Hippo pathway is a master regulator of development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size control. Hippo pathway relays signals from different extracellular and intracellular events to regulate cell behavior and functions. Hippo pathway is conserved from Protista to eukaryotes. Deregulation of the Hippo pathway is associated with numerous cancers. Alteration of the Hippo pathway results in cell invasion, migration, disease progression, and therapy resistance in cancers. However, the function of the various components of the mammalian Hippo pathway is yet to be elucidated in detail especially concerning tumor biology. In the present review, we focused on the Hippo pathway in different model organisms, its regulation and deregulation, and possible therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Suman Mohajan
- Department of Biochemistry and Molecular Biology, LSUHSC, Shreveport, USA
| | - Praveen Kumar Jaiswal
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA; Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA
| | - Mousa Vatanmakarian
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | | | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA; Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA
| | - Sweaty Koul
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA
| | - Hari K Koul
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA; Urology, LSUHSC, School of Medicine, New Orleans, USA; Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA.
| |
Collapse
|
168
|
Brücher VC, Egbring C, Plagemann T, Nedvetsky PI, Höffken V, Pavenstädt H, Eter N, Kremerskothen J, Heiduschka P. Lack of WWC2 Protein Leads to Aberrant Angiogenesis in Postnatal Mice. Int J Mol Sci 2021; 22:5321. [PMID: 34070186 PMCID: PMC8158494 DOI: 10.3390/ijms22105321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/03/2023] Open
Abstract
The WWC protein family is an upstream regulator of the Hippo signalling pathway that is involved in many cellular processes. We examined the effect of an endothelium-specific WWC1 and/or WWC2 knock-out on ocular angiogenesis. Knock-outs were induced in C57BL/6 mice at the age of one day (P1) and evaluated at P6 (postnatal mice) or induced at the age of five weeks and evaluated at three months of age (adult mice). We analysed morphology of retinal vasculature in retinal flat mounts. In addition, in vivo imaging and functional testing by electroretinography were performed in adult mice. Adult WWC1/2 double knock-out mice differed neither functionally nor morphologically from the control group. In contrast, the retinas of the postnatal WWC knock-out mice showed a hyperproliferative phenotype with significantly enlarged areas of sprouting angiogenesis and a higher number of tip cells. The branching and end points in the peripheral plexus were significantly increased compared to the control group. The deletion of the WWC2 gene was decisive for these effects; while knocking out WWC1 showed no significant differences. The results hint strongly that WWC2 is an essential regulator of ocular angiogenesis in mice. As an activator of the Hippo signalling pathway, it prevents excessive proliferation during physiological angiogenesis. In adult animals, WWC proteins do not seem to be important for the maintenance of the mature vascular plexus.
Collapse
Affiliation(s)
- Viktoria Constanze Brücher
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Charlotte Egbring
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Tanja Plagemann
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Pavel I. Nedvetsky
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Verena Höffken
- Medical Cell Biology, Medical Clinic D, University of Münster Medical School, 48149 Münster, Germany;
| | - Hermann Pavenstädt
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Nicole Eter
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Joachim Kremerskothen
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Peter Heiduschka
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| |
Collapse
|
169
|
Teixeira SA, Marques DBD, Costa TC, Oliveira HC, Costa KA, Carrara ER, da Silva W, Guimarães JD, Neves MM, Ibelli AMG, Cantão ME, Ledur MC, Peixoto JO, Guimarães SEF. Transcription Landscape of the Early Developmental Biology in Pigs. Animals (Basel) 2021; 11:ani11051443. [PMID: 34069910 PMCID: PMC8157595 DOI: 10.3390/ani11051443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/13/2022] Open
Abstract
Since pre- and postnatal development are programmed during early prenatal life, studies addressing the complete transcriptional landscape during organogenesis are needed. Therefore, we aimed to disentangle differentially expressed (DE) genes between fetuses (at 35 days old) and embryos (at 25 days old) through RNA-sequencing analysis using the pig as model. In total, 1705 genes were DE, including the top DE IBSP, COL6A6, HBE1, HBZ, HBB, and NEUROD6 genes, which are associated with developmental transition from embryos to fetuses, such as ossification, skeletal muscle development, extracellular matrix organization, cardiovascular system, erythrocyte differentiation, and neuronal system. In pathway analysis, embryonic development highlighted those mainly related to morphogenic signaling and cell interactions, which are crucial for transcriptional control during the establishment of the main organs in early prenatal development, while pathways related to myogenesis, neuronal development, and cardiac and striated muscle contraction were enriched for fetal development, according to the greater complexity of organs and body structures at this developmental stage. Our findings provide an exploratory and informative transcriptional landscape of pig organogenesis, which might contribute to further studies addressing specific developmental events in pigs and in other mammals.
Collapse
Affiliation(s)
- Susana A. Teixeira
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - Daniele B. D. Marques
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - Thaís C. Costa
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - Haniel C. Oliveira
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - Karine A. Costa
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - Eula R. Carrara
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - Walmir da Silva
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
| | - José D. Guimarães
- Department of Veterinary Medicine, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil;
| | - Mariana M. Neves
- Department of General Biology, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil;
| | - Adriana M. G. Ibelli
- Embrapa Suínos e Aves, Concordia 89715-899, SC, Brazil; (A.M.G.I.); (M.E.C.); (M.C.L.); (J.O.P.)
| | - Maurício E. Cantão
- Embrapa Suínos e Aves, Concordia 89715-899, SC, Brazil; (A.M.G.I.); (M.E.C.); (M.C.L.); (J.O.P.)
| | - Mônica C. Ledur
- Embrapa Suínos e Aves, Concordia 89715-899, SC, Brazil; (A.M.G.I.); (M.E.C.); (M.C.L.); (J.O.P.)
| | - Jane O. Peixoto
- Embrapa Suínos e Aves, Concordia 89715-899, SC, Brazil; (A.M.G.I.); (M.E.C.); (M.C.L.); (J.O.P.)
| | - Simone E. F. Guimarães
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa 36570-000, MG, Brazil; (S.A.T.); (D.B.D.M.); (T.C.C.); (H.C.O.); (K.A.C.); (E.R.C.); (W.d.S.)
- Correspondence: ; Tel.: +55-31-36124671
| |
Collapse
|
170
|
Wu Y, Aegerter P, Nipper M, Ramjit L, Liu J, Wang P. Hippo Signaling Pathway in Pancreas Development. Front Cell Dev Biol 2021; 9:663906. [PMID: 34079799 PMCID: PMC8165189 DOI: 10.3389/fcell.2021.663906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo signaling pathway is a vital regulator of pancreatic development and homeostasis, directing cell fate decisions, morphogenesis, and adult pancreatic cellular plasticity. Through loss-of-function research, Hippo signaling has been found to play key roles in maintaining the proper balance between progenitor cell renewal, proliferation, and differentiation in pancreatic organogenesis. Other studies suggest that overactivation of YAP, a downstream effector of the pathway, promotes ductal cell development and suppresses endocrine cell fate specification via repression of Ngn3. After birth, disruptions in Hippo signaling have been found to lead to de-differentiation of acinar cells and pancreatitis-like phenotype. Further, Hippo signaling directs pancreatic morphogenesis by ensuring proper cell polarization and branching. Despite these findings, the mechanisms through which Hippo governs cell differentiation and pancreatic architecture are yet to be fully understood. Here, we review recent studies of Hippo functions in pancreatic development, including its crosstalk with NOTCH, WNT/β-catenin, and PI3K/Akt/mTOR signaling pathways.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States.,Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pauline Aegerter
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Michael Nipper
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Logan Ramjit
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jun Liu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Pei Wang
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
171
|
Okamoto T, Park EJ, Kawamoto E, Usuda H, Wada K, Taguchi A, Shimaoka M. Endothelial connexin-integrin crosstalk in vascular inflammation. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166168. [PMID: 33991620 DOI: 10.1016/j.bbadis.2021.166168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/18/2021] [Accepted: 05/02/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases including blood vessel disorders represent a major cause of death globally. The essential roles played by local and systemic vascular inflammation in the pathogenesis of cardiovascular diseases have been increasingly recognized. Vascular inflammation triggers the aberrant activation of endothelial cells, which leads to the functional and structural abnormalities in vascular vessels. In addition to humoral mediators such as pro-inflammatory cytokines and prostaglandins, the alteration of physical and mechanical microenvironment - including vascular stiffness and shear stress - modify the gene expression profiles and metabolic profiles of endothelial cells via mechano-transduction pathways, thereby contributing to the pathogenesis of vessel disorders. Notably, connexins and integrins crosstalk each other in response to the mechanical stress, and, thereby, play an important role in regulating the mechano-transduction of endothelial cells. Here, we provide an overview on how the inter-play between connexins and integrins in endothelial cells unfold during the mechano-transduction in vascular inflammation.
Collapse
Affiliation(s)
- Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan.
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Haruki Usuda
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan.
| |
Collapse
|
172
|
Abstract
AbstractIn the developing Drosophila CNS, two pools of neural stem cells, the symmetrically dividing progenitors in the neuroepithelium (NE) and the asymmetrically dividing neuroblasts (NBs) generate the majority of the neurons that make up the adult central nervous system (CNS). The generation of a correct sized brain depends on maintaining the fine balance between neural stem cell self-renewal and differentiation, which are regulated by cell-intrinsic and cell-extrinsic cues. In this review, we will discuss our current understanding of how self-renewal and differentiation are regulated in the two neural stem cell pools, and the consequences of the deregulation of these processes.
Collapse
Affiliation(s)
- Francesca Froldi
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
| | - Milán Szuperák
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
| | - Louise Y. Cheng
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
173
|
Estella C, Baonza A. Cell proliferation control by Notch signalling during imaginal discs development in Drosophila. AIMS GENETICS 2021. [DOI: 10.3934/genet.2015.1.70] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AbstractThe Notch signalling pathway is evolutionary conserved and participates in numerous developmental processes, including the control of cell proliferation. However, Notch signalling can promote or restrain cell division depending on the developmental context, as has been observed in human cancer where Notch can function as a tumor suppressor or an oncogene. Thus, the outcome of Notch signalling can be influenced by the cross-talk between Notch and other signalling pathways. The use of model organisms such as Drosophila has been proven to be very valuable to understand the developmental role of the Notch pathway in different tissues and its relationship with other signalling pathways during cell proliferation control. Here we review recent studies in Drosophila that shed light in the developmental control of cell proliferation by the Notch pathway in different contexts such as the eye, wing and leg imaginal discs. We also discuss the autonomous and non-autonomous effects of the Notch pathway on cell proliferation and its interactions with different signalling pathways.
Collapse
Affiliation(s)
- Carlos Estella
- Departamento de Biología Molecular and Centro de Biología Molecular SeveroOchoa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Antonio Baonza
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM) c/Nicolás Cabrera 1, 28049, Madrid, Spain
| |
Collapse
|
174
|
Yang S, Xu W, Liu C, Jin J, Li X, Jiang Y, Zhang L, Meng X, Zhan J, Zhang H. LATS1 K751 acetylation blocks activation of Hippo signalling and switches LATS1 from a tumor suppressor to an oncoprotein. SCIENCE CHINA-LIFE SCIENCES 2021; 65:129-141. [PMID: 33945069 DOI: 10.1007/s11427-020-1914-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/16/2021] [Indexed: 11/28/2022]
Abstract
Large tumor suppressor 1 (LATS1) is the key kinase controlling activation of Hippo signalling pathway. Post-translational modifications of LATS1 modulate its kinase activity. However, detailed mechanism underlying LATS1 stability and activation remains elusive. Here we report that LATS1 is acetylated by acetyltransferase CBP at K751 and is deacetylated by deacetylases SIRT3 and SIRT4. Acetylation at K751 stabilized LATS1 by decreasing LATS1 ubiquitination and inhibited LATS1 activation by reducing its phosphorylation. Mechanistically, LATS1 acetylation resulted in inhibition of YAP phosphorylation and degradation, leading to increased YAP nucleus translocation and promoted target gene expression. Functionally, LATS1-K751Q, the acetylation mimic mutant potentiated lung cancer cell migration, invasion and tumor growth, whereas LATS1-K751R, the acetylation deficient mutant inhibited these functions. Taken together, we demonstrated a previously unidentified post-translational modification of LATS1 that converts LATS1 from a tumor suppressor to a tumor promoter by suppression of Hippo signalling through acetylation of LATS1.
Collapse
Affiliation(s)
- Siyuan Yang
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Weizhi Xu
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Cheng Liu
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Jiaqi Jin
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Xueying Li
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Yuhan Jiang
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Lei Zhang
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jun Zhan
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Hongquan Zhang
- Department of Human Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
175
|
Iversen M, Mulugeta T, West AC, Jørgensen EH, Martin SAM, Sandve SR, Hazlerigg D. Photoperiod-dependent developmental reprogramming of the transcriptional response to seawater entry in Atlantic salmon (Salmo salar). G3-GENES GENOMES GENETICS 2021; 11:6169000. [PMID: 33710311 PMCID: PMC8049429 DOI: 10.1093/g3journal/jkab072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/28/2021] [Indexed: 01/22/2023]
Abstract
The developmental transition of juvenile salmon from a freshwater resident morph (parr) to a seawater (SW) migratory morph (smolt), known as smoltification, entails a reorganization of gill function to cope with the altered water environment. Recently, we used RNAseq to characterize the breadth of transcriptional change which takes place in the gill in the FW phase of smoltification. This highlighted the importance of extended exposure to short, winter-like photoperiods (SP) followed by a subsequent increase in photoperiod for completion of transcriptional reprogramming in FW and efficient growth following transfer to SW. Here, we extend this analysis to examine the consequences of this photoperiodic history-dependent reprogramming for subsequent gill responses upon exposure to SW. We use RNAseq to analyze gill samples taken from fish raised on the photoperiod regimes we used previously and then challenged by SW exposure for 24 hours. While fish held on constant light (LL) throughout were able to hypo-osmoregulate during a 24 hours SW challenge, the associated gill transcriptional response was highly distinctive from that in fish which had experienced a 7-week period of exposure to SP followed by a return to LL (SPLL) and had consequently acquired the characteristics of fully developed smolts. Fish transferred from LL to SP, and then held on SP for the remainder of the study was unable to hypo-osmoregulate, and the associated gill transcriptional response to SW exposure featured many transcripts apparently regulated by the glucocorticoid stress axis and by the osmo-sensing transcription factor NFAT5. The importance of these pathways for the gill transcriptional response to SW exposure appears to diminish as a consequence of photoperiod mediated induction of the smolt phenotype, presumably reflecting preparatory developmental changes taking place during this process.
Collapse
Affiliation(s)
- Marianne Iversen
- Department of Arctic and Marine Biology, UiT -The Arctic University of Norway, Tromsø NO-9037, Norway
| | - Teshome Mulugeta
- Department of Animal and Aquaculture Sciences, Norwegian University of Life Sciences, Ås NO-1432, Norway
| | - Alexander C West
- Department of Arctic and Marine Biology, UiT -The Arctic University of Norway, Tromsø NO-9037, Norway
| | - Even H Jørgensen
- Department of Arctic and Marine Biology, UiT -The Arctic University of Norway, Tromsø NO-9037, Norway
| | - Samuel A M Martin
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK
| | - Simen Rød Sandve
- Centre for Integrative Genetics, Department of Animal and Aquaculture Sciences, Norwegian University of Life Sciences, Ås NO-1432, Norway
| | - David Hazlerigg
- Department of Arctic and Marine Biology, UiT -The Arctic University of Norway, Tromsø NO-9037, Norway
| |
Collapse
|
176
|
Schmid D, Warnken U, Latzer P, Hoffmann DC, Roth J, Kutschmann S, Jaschonek H, Rübmann P, Foltyn M, Vollmuth P, Winkler F, Seliger C, Felix M, Sahm F, Haas J, Reuss D, Bendszus M, Wildemann B, von Deimling A, Wick W, Kessler T. Diagnostic biomarkers from proteomic characterization of cerebrospinal fluid in patients with brain malignancies. J Neurochem 2021; 158:522-538. [PMID: 33735443 DOI: 10.1111/jnc.15350] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 12/23/2022]
Abstract
Recent technological advances in molecular diagnostics through liquid biopsies hold the promise to repetitively monitor tumor evolution and treatment response of brain malignancies without the need of invasive surgical tissue accrual. Here, we implemented a mass spectrometry-based protein analysis pipeline which identified hundreds of proteins in 251 cerebrospinal fluid (CSF) samples from patients with four types of brain malignancies (glioblastoma, lymphoma, brain metastasis, and leptomeningeal disease [LMD]) and from healthy individuals with a focus on glioblastoma in a retrospective and confirmatory prospective observational study. CSF proteome deregulation via disruption of the blood brain barrier appeared to be largely conserved across brain tumor entities. CSF analysis of glioblastoma patients identified two proteomic clusters that correlated with tumor size and patient survival. By integrating CSF data with proteomic analyses of matching glioblastoma tumor tissue and primary glioblastoma cells, we identified potential CSF biomarkers for glioblastoma, in particular chitinase-3-like protein 1 (CHI3L1) and glial fibrillary acidic protein (GFAP). Key findings were validated in a prospective cohort consisting of 35 glioma patients. Finally, in LMD patients who frequently undergo repeated CSF work-up, we explored our proteomic pipeline as a mean to profile consecutive CSF samples. Therefore, proteomic analysis of CSF in brain malignancies has the potential to reveal biomarkers for diagnosis and therapy monitoring.
Collapse
Affiliation(s)
- Dominic Schmid
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany.,Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Uwe Warnken
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Functional Proteome Analysis, DKFZ, Heidelberg, Germany
| | - Pauline Latzer
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Judith Roth
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Kutschmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hannah Jaschonek
- Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Petra Rübmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martha Foltyn
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Corinna Seliger
- Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Marius Felix
- Clinical Cooperation Unit Neuropathology, DKTK, DKFZ, Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, DKTK, DKFZ, Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Haas
- Molecular Neuroimmunology, Heidelberg University Hospital, Heidelberg, Germany
| | - David Reuss
- Clinical Cooperation Unit Neuropathology, DKTK, DKFZ, Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Brigitte Wildemann
- Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany.,Molecular Neuroimmunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology, DKTK, DKFZ, Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neurooncology Program at the National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
177
|
Wang M, Xu T, Feng W, Liu J, Wang Z. Advances in Understanding the LncRNA-Mediated Regulation of the Hippo Pathway in Cancer. Onco Targets Ther 2021; 14:2397-2415. [PMID: 33854336 PMCID: PMC8039192 DOI: 10.2147/ott.s283157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are a class of RNA molecules that are longer than 200 nucleotides and cannot encode proteins. Over the past decade, lncRNAs have been defined as regulatory elements of multiple biological processes, and their aberrant expression contributes to the development and progression of various malignancies. Recent studies have shown that lncRNAs are involved in key cancer-related signaling pathways, including the Hippo signaling pathway, which plays a prominent role in controlling organ size and tissue homeostasis by regulating cell proliferation, apoptosis, and differentiation. However, dysregulation of this pathway is associated with pathological conditions, especially cancer. Accumulating evidence has revealed that lncRNAs can modulate the Hippo signaling pathway in cancer. In this review, we elaborate on the role of the Hippo signaling pathway and the advances in the understanding of its lncRNA-mediated regulation in cancer. This review provides additional insight into carcinogenesis and will be of great clinical value for developing novel early detection and treatment strategies for this deadly disease.
Collapse
Affiliation(s)
- Mengwei Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Tianwei Xu
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wenyan Feng
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Junxia Liu
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Zhaoxia Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
178
|
Circ-ITCH sponges miR-214 to promote the osteogenic differentiation in osteoporosis via upregulating YAP1. Cell Death Dis 2021; 12:340. [PMID: 33795657 PMCID: PMC8016856 DOI: 10.1038/s41419-021-03586-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/14/2022]
Abstract
Osteoporosis is the most prevailing primary bone disease and a growing health care burden. The aim of this study was to clarify the functional roles and mechanisms of the circ-ITCH regulating osteogenic differentiation of osteoporosis. Circ-ITCH and yes-associated protein 1 (YAP1) levels were downregulated, but the miR‐214 level was upregulated in osteoporotic mice and patients. Knockdown of circ-ITCH inhibited the alkaline phosphatase (ALP) activity, mineralized nodule formation, and expression of runt-related transcription factor 2 (RUNX2), osteopontin (OPN), and osteocalcin (OCN) during osteogenic induction. Furthermore, miR-214 was a target of circ-ITCH, knockdown of miR-214 could impede the regulatory effects of sh-circ-ITCH on osteogenic differentiation. Moreover, miR-214 suppressed hBMSCs osteogenic differentiation by downregulating YAP1. Finally, in vivo experiments indicated that overexpression of circ-ITCH could improve osteogenesis in ovariectomized mice. In conclusion, circ-ITCH upregulated YAP1 expression to promote osteogenic differentiation in osteoporosis via sponging miR-214. Circ-ITCH could act as a novel therapeutic target for osteoporosis.
Collapse
|
179
|
Cho YS, Jiang J. Hippo-Independent Regulation of Yki/Yap/Taz: A Non-canonical View. Front Cell Dev Biol 2021; 9:658481. [PMID: 33869224 PMCID: PMC8047194 DOI: 10.3389/fcell.2021.658481] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022] Open
Abstract
Initially identified in Drosophila, the Hippo signaling pathway has emerged as an evolutionarily conserved tumor suppressor pathway that controls tissue growth and organ size by simultaneously inhibiting cell proliferation and promoting cell death. Deregulation of Hippo pathway activity has been implicated in a wide range of human cancers. The core Hippo pathway consists of a kinase cascade: an upstream kinase Hippo (Hpo)/MST1/2 phosphorylates and activates a downstream kinase Warts (Wts)/Lats1/2, leading to phosphorylation and inactivation of a transcriptional coactivator Yki/YAP/Taz. Many upstream signals, including cell adhesion, polarity, mechanical stress, and soluble factors, regulate Hippo signaling through the kinase cascade, leading to change in the cytoplasmic/nuclear localization of Yki/YAP/Taz. However, recent studies have uncovered other mechanisms that regulate Yki/YAP/Taz subcellular localization, stability, and activity independent of the Hpo kinase cascade. These mechanisms provide additional layers of pathway regulation, nodes for pathway crosstalk, and opportunities for pathway intervention in cancer treatment and regenerative medicine.
Collapse
Affiliation(s)
- Yong Suk Cho
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Jin Jiang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States.,Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
180
|
O-GlcNAcylation and O-GlcNAc Cycling Regulate Gene Transcription: Emerging Roles in Cancer. Cancers (Basel) 2021; 13:cancers13071666. [PMID: 33916244 PMCID: PMC8037238 DOI: 10.3390/cancers13071666] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary O-linked β-N-acetylglucosamine (O-GlcNAc) is a post-translational modification (PTM) linking nutrient flux through the hexosamine biosynthetic pathway (HBP) to gene transcription. Mounting experimental and clinical data implicates aberrant O-GlcNAcylation in the development and progression of cancer. Herein, we discuss how alteration of O-GlcNAc-regulated transcriptional mechanisms leads to atypical gene expression in cancer. We discuss the challenges associated with studying O-GlcNAc function and present several new approaches for studies of O-GlcNAc-regulated transcription. Abstract O-linked β-N-acetylglucosamine (O-GlcNAc) is a single sugar post-translational modification (PTM) of intracellular proteins linking nutrient flux through the Hexosamine Biosynthetic Pathway (HBP) to the control of cis-regulatory elements in the genome. Aberrant O-GlcNAcylation is associated with the development, progression, and alterations in gene expression in cancer. O-GlcNAc cycling is defined as the addition and subsequent removal of the modification by O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA) provides a novel method for cells to regulate various aspects of gene expression, including RNA polymerase function, epigenetic dynamics, and transcription factor activity. We will focus on the complex relationship between phosphorylation and O-GlcNAcylation in the regulation of the RNA Polymerase II (RNAP II) pre-initiation complex and the regulation of the carboxyl-terminal domain of RNAP II via the synchronous actions of OGT, OGA, and kinases. Additionally, we discuss how O-GlcNAcylation of TATA-box binding protein (TBP) alters cellular metabolism. Next, in a non-exhaustive manner, we will discuss the current literature on how O-GlcNAcylation drives gene transcription in cancer through changes in transcription factor or chromatin remodeling complex functions. We conclude with a discussion of the challenges associated with studying O-GlcNAcylation and present several new approaches for studying O-GlcNAc regulated transcription that will advance our understanding of the role of O-GlcNAc in cancer.
Collapse
|
181
|
Peng Y, Qing X, Lin H, Huang D, Li J, Tian S, Liu S, Lv X, Ma K, Li R, Rao Z, Bai Y, Chen S, Lei M, Quan D, Shao Z. Decellularized Disc Hydrogels for hBMSCs tissue-specific differentiation and tissue regeneration. Bioact Mater 2021; 6:3541-3556. [PMID: 33842740 PMCID: PMC8022111 DOI: 10.1016/j.bioactmat.2021.03.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue specificity, a key factor in the decellularized tissue matrix (DTM), has shown bioactive functionalities in tuning cell fate-e.g., the differentiation of mesenchymal stem cells. Notably, cell fate is also determined by the living microenvironment, including material composition and spatial characteristics. Herein, two neighboring tissues within intervertebral discs, the nucleus pulposus (NP) and annulus fibrosus (AF), were carefully processed into DTM hydrogels (abbreviated DNP-G and DAF-G, respectively) to determine the tissue-specific effects on stem cell fate, such as specific components and different culturing methods, as well as in vivo regeneration. Distinct differences in their protein compositions were identified by proteomic analysis. Interestingly, the fate of human bone marrow mesenchymal stem cells (hBMSCs) also responds to both culturing methods and composition. Generally, hBMSCs cultured with DNP-G (3D) differentiated into NP-like cells, while hBMSCs cultured with DAF-G (2D) underwent AF-like differentiation, indicating a close correlation with the native microenvironments of NP and AF cells, respectively. Furthermore, we found that the integrin-mediated RhoA/LATS/YAP1 signaling pathway was activated in DAF-G (2D)-induced AF-specific differentiation. Additionally, the activation of YAP1 determined the tendency of NP- or AF-specific differentiation and played opposite regulatory effects. Finally, DNP-G and DAF-G specifically promoted tissue regeneration in NP degeneration and AF defect rat models, respectively. In conclusion, DNP-G and DAF-G can specifically determine the fate of stem cells through the integrin-mediated RhoA/LATS/YAP1 signaling pathway, and this tissue specificity is both compositional and spatial, supporting the utilization of tissue-specific DTM in advanced treatments of intervertebral disc degeneration.
Collapse
Key Words
- 2D, two-dimensional
- 3D, three-dimensional
- AF, annulus fibrosus
- Col I–S, collagen type I solution
- DAF, decellularized annulus fibrosus
- DAF-G, decellularized annulus fibrosus hydrogel
- DAF-S, decellularized annulus fibrosus solution
- DNP, decellularized nucleus pulposus
- DNP-G, decellularized nucleus pulposus hydrogel
- DNP-S, decellularized nucleus pulposus solution
- DTM, decellularized tissue matrix
- Decellularized tissue matrix
- Differentiation
- ECM, extracellular matrix
- FAF, fresh annulus fibrosus
- FNP, fresh nucleus pulposus
- IDD, intervertebral disc degeneration
- Intervertebral disc
- MSC, mesenchymal stem cell
- NP, nucleus pulposus
- Tissue specificity
- YAP1
- YAP1, yes-associated protein 1
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Donghua Huang
- Musculoskeletal Tumor Center, Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinye Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaige Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Li
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China
| | - Zilong Rao
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China
| | - Ying Bai
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510127, China
| | - Songfeng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, China
| | - Ming Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Corresponding author.
| | - Daping Quan
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China,School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510127, China,Corresponding author. School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Corresponding author.
| |
Collapse
|
182
|
Kim G, Bhattarai PY, Lim SC, Kim JY, Choi HS. PIN1 facilitates ubiquitin-mediated degradation of serine/threonine kinase 3 and promotes melanoma development via TAZ activation. Cancer Lett 2021; 499:164-174. [PMID: 33253791 DOI: 10.1016/j.canlet.2020.11.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/10/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
The Hippo signaling pathway controls cellular processes including growth, homeostasis, and apoptosis. The kinase STK3 acts upstream in this pathway to activate LATS1/2 kinase, which phosphorylates and inactivates the transcriptional coactivators YAP/TAZ. The dysregulation of Hippo signaling leads to human diseases including cancer; however, the molecular mechanisms underlying its dysregulation in melanoma are unknown. We aimed to determine the role of the PIN1 in Hippo signaling dysregulation and melanoma tumorigenesis. We report that PIN1 interacts with STK3 and induces ubiquitination-dependent proteasomal degradation of STK3. Furthermore, PIN1 plays a critical role in the nuclear translocation of TAZ, which forms a complex with TEAD to increase CTGF expression. PIN1 ablation blocks TAZ/TEAD complex formation and decreases CTGF expression. PIN1-mediated STK3 degradation is associated with enhanced cell growth, induction of cell transformation, and increased tumorigenicity. In clinical context, PIN1 and STK3 levels are inversely correlated in patient melanoma tissues. These findings indicate that PIN1-mediated STK3 destabilization contributes to the dysregulation of Hippo signaling, leading to oncogenic signaling and melanoma tumorigenesis. Our data suggest that inhibition of the PIN1-STK3 axis could be a novel treatment strategy for malignant melanoma.
Collapse
Affiliation(s)
- Garam Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | | | - Sung-Chul Lim
- Department of Pathology, School of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jin-Young Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hong Seok Choi
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
183
|
Kumar B, Ahmad R, Giannico GA, Zent R, Talmon GA, Harris RC, Clark PE, Lokeshwar V, Dhawan P, Singh AB. Claudin-2 inhibits renal clear cell carcinoma progression by inhibiting YAP-activation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:77. [PMID: 33622361 PMCID: PMC7901196 DOI: 10.1186/s13046-021-01870-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/08/2021] [Indexed: 12/28/2022]
Abstract
Background Claudin-2 expression is upregulated in multiple cancers and promotes cancer malignancy. Remarkably, the regulation of claudin-2 expression in kidney cell lines contrasts its reported regulation in other organs. However, claudin-2 role in renal clear cell carcinoma (RCC) remains unknown despite its predominant expression in the proximal tubular epithelium (PTE), the site of RCC origin. Methods Publicly available and independent patient databases were examined for claudin-2 association with RCC. The novel protein function was validated in vitro and in vivo by gain or loss of function assays. Mechanistic results were concluded by Mass spectroscopy, immunoprecipitation and mutational studies, and functional evaluations. Results We show that the significant decrease in claudin-2 expression characterized PTE cells and Ex-vivo cultured mouse kidney subjected to dedifferentiation. Inhibition of claudin-2 was enough to induce mesenchymal plasticity and invasive mobility in these models. Further, a progressive loss of claudin-2 expression associated with the RCC progression and poor patient survival. Overexpression of claudin-2 in RCC-derived cancer cells inhibited tumorigenic abilities and xenograft tumor growth. These data supported a novel tumor-suppressive role of claudin-2 in RCC. Mechanistic insights further revealed that claudin-2 associates with YAP-protein and modulates its phosphorylation (S127) and nuclear expression. The tumor suppressive effects of claudin-2 expression were lost upon deletion of its PDZ-binding motif emphasizing the critical role of the PDZ-domain in claudin-2 interaction with YAP in regulating RCC malignancy. Conclusions Our results demonstrate a novel kidney specific tumor suppressive role for claudin-2 protein and further demonstrate that claudin-2 co-operates with the YAP signaling in regulating the RCC malignancy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01870-5.
Collapse
Affiliation(s)
- Balawant Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Giovanna A Giannico
- Department of Pathology, Microbiology and Immunology, Vanderbilt Medical Center, Nashville, TN, USA
| | - Roy Zent
- Department of Medicine, Vanderbilt Medical Center, Nashville, TN, USA
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Raymond C Harris
- Department of Medicine, Vanderbilt Medical Center, Nashville, TN, USA
| | | | - Vinata Lokeshwar
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA.,Member, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA. .,Member, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA. .,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
184
|
Ngo MHT, Jeng HY, Kuo YC, Nanda JD, Brahmadhi A, Ling TY, Chang TS, Huang YH. The Role of IGF/IGF-1R Signaling in Hepatocellular Carcinomas: Stemness-Related Properties and Drug Resistance. Int J Mol Sci 2021; 22:ijms22041931. [PMID: 33669204 PMCID: PMC7919800 DOI: 10.3390/ijms22041931] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin-like Growth Factor (IGF)/IGF-1 Receptor (IGF-1R) signaling is known to regulate stem cell pluripotency and differentiation to trigger cell proliferation, organ development, and tissue regeneration during embryonic development. Unbalanced IGF/IGF-1R signaling can promote cancer cell proliferation and activate cancer reprogramming in tumor tissues, especially in the liver. Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death, with a high incidence and mortality rate in Asia. Most patients with advanced HCC develop tyrosine kinase inhibitor (TKI)-refractoriness after receiving TKI treatment. Dysregulation of IGF/IGF-1R signaling in HCC may activate expression of cancer stemness that leads to TKI refractoriness and tumor recurrence. In this review, we summarize the evidence for dysregulated IGF/IGF-1R signaling especially in hepatitis B virus (HBV)-associated HCC. The regulation of cancer stemness expression and drug resistance will be highlighted. Current clinical treatments and potential therapies targeting IGF/IGF-1R signaling for the treatment of HCC will be discussed.
Collapse
Affiliation(s)
- Mai-Huong Thi Ngo
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Han-Yin Jeng
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-Y.J.); (Y.-C.K.)
| | - Yung-Che Kuo
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-Y.J.); (Y.-C.K.)
| | - Josephine Diony Nanda
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
| | - Ageng Brahmadhi
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 11031, Taiwan
- Correspondence: (T.-Y.L.); (T.-S.C.); (Y.-H.H.); Tel.: +886-2-2312-3456 (ext. 8-8322) (T.-Y.L.); +886-5-3621-000 (ext. 2242) (T.-S.C.); +886-2-2736-1661 (ext. 3150) (Y.-H.H.)
| | - Te-Sheng Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33382, Taiwan
- Division of Internal Medicine, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
- Correspondence: (T.-Y.L.); (T.-S.C.); (Y.-H.H.); Tel.: +886-2-2312-3456 (ext. 8-8322) (T.-Y.L.); +886-5-3621-000 (ext. 2242) (T.-S.C.); +886-2-2736-1661 (ext. 3150) (Y.-H.H.)
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-Y.J.); (Y.-C.K.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (T.-Y.L.); (T.-S.C.); (Y.-H.H.); Tel.: +886-2-2312-3456 (ext. 8-8322) (T.-Y.L.); +886-5-3621-000 (ext. 2242) (T.-S.C.); +886-2-2736-1661 (ext. 3150) (Y.-H.H.)
| |
Collapse
|
185
|
Strutt H, Strutt D. How do the Fat-Dachsous and core planar polarity pathways act together and independently to coordinate polarized cell behaviours? Open Biol 2021; 11:200356. [PMID: 33561385 PMCID: PMC8061702 DOI: 10.1098/rsob.200356] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Planar polarity describes the coordinated polarization of cells within the plane of a tissue. This is controlled by two main pathways in Drosophila: the Frizzled-dependent core planar polarity pathway and the Fat–Dachsous pathway. Components of both of these pathways become asymmetrically localized within cells in response to long-range upstream cues, and form intercellular complexes that link polarity between neighbouring cells. This review examines if and when the two pathways are coupled, focusing on the Drosophila wing, eye and abdomen. There is strong evidence that the pathways are molecularly coupled in tissues that express a specific isoform of the core protein Prickle, namely Spiny-legs. However, in other contexts, the linkages between the pathways are indirect. We discuss how the two pathways act together and independently to mediate a diverse range of effects on polarization of cell structures and behaviours.
Collapse
Affiliation(s)
- Helen Strutt
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David Strutt
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
186
|
Li T, Liu J, Feng J, Liu Z, Liu S, Zhang M, Zhang Y, Hou Y, Wu D, Li C, Chen Y, Chen H, Lu X. Variation in the life history strategy underlies functional diversity of tumors. Natl Sci Rev 2021; 8:nwaa124. [PMID: 34691566 PMCID: PMC8288455 DOI: 10.1093/nsr/nwaa124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022] Open
Abstract
Classical r- vs. K-selection theory describes the trade-offs between high reproductive output and competitiveness and guides research in evolutionary ecology. While its impact has waned in the recent past, cancer evolution may rekindle it. Herein, we impose r- or K-selection on cancer cell lines to obtain strongly proliferative r cells and highly competitive K cells to test ideas on life-history strategy evolution. RNA-seq indicates that the trade-offs are associated with distinct expression of genes involved in the cell cycle, adhesion, apoptosis, and contact inhibition. Both empirical observations and simulations based on an ecological competition model show that the trade-off between cell proliferation and competitiveness can evolve adaptively. When the r and K cells are mixed, they exhibit strikingly different spatial and temporal distributions. Due to this niche separation, the fitness of the entire tumor increases. The contrasting selective pressure may operate in a realistic ecological setting of actual tumors.
Collapse
Affiliation(s)
- Tao Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jialin Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Feng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenzhen Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sixue Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minjie Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuezheng Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yali Hou
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Dafei Wu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chunyan Li
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering and Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
| | - Yongbin Chen
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Hua Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuemei Lu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
187
|
Wang C, Lin W, Wang Y, Fu L. Suppression of Hippo Pathway by Food Allergen Exacerbates Intestinal Epithelia Instability and Facilitates Hypersensitivity. Mol Nutr Food Res 2021; 65:e2000593. [PMID: 33245584 DOI: 10.1002/mnfr.202000593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/15/2020] [Indexed: 12/30/2022]
Abstract
SCOPE Hippo signaling is a crucial pathway in innate immune responses, but the relationship between food allergy and Hippo pathway is unknown. The aim of this work is to investigate the regulation of food allergy by Hippo pathway and reveal the molecular mechanisms. METHODS AND RESULTS Two food allergens tropomyosin and ovalbumin are used to challenge a mouse model and CMT93 intestinal epithelia cell model. The allergic responses and the activation of Hippo pathway are tested in these models. In the mouse model, both allergens trigged significant allergic responses, and Hippo pathway is suppressed after allergen challenge. In CMT93, both allergens upregulate the expression of allergic cytokines thymic stromal lymphopoietin, interleukin (IL)-25, and IL-33. In TAZ KD CMT93, the Hippo pathway is blocked, and the expression of allergenic cytokines are also suppressed. CONCLUSIONS Both in vivo and in vitro data demonstrate that the two food allergens suppressed Hippo pathway by downregulating TAZ expression, resulting in intestinal epithelia instability, and finally leading to hypersensitivity reactions. These findings provide potential therapeutic targets and molecular markers for food allergy, and provide dietary guidelines for allergenic individuals.
Collapse
Affiliation(s)
- Chong Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Wanglei Lin
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| |
Collapse
|
188
|
Li L, Ugalde AP, Scheele CLGJ, Dieter SM, Nagel R, Ma J, Pataskar A, Korkmaz G, Elkon R, Chien MP, You L, Su PR, Bleijerveld OB, Altelaar M, Momchev L, Manber Z, Han R, van Breugel PC, Lopes R, ten Dijke P, van Rheenen J, Agami R. A comprehensive enhancer screen identifies TRAM2 as a key and novel mediator of YAP oncogenesis. Genome Biol 2021; 22:54. [PMID: 33514403 PMCID: PMC7845134 DOI: 10.1186/s13059-021-02272-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Frequent activation of the co-transcriptional factor YAP is observed in a large number of solid tumors. Activated YAP associates with enhancer loci via TEAD4-DNA-binding protein and stimulates cancer aggressiveness. Although thousands of YAP/TEAD4 binding-sites are annotated, their functional importance is unknown. Here, we aim at further identification of enhancer elements that are required for YAP functions. RESULTS We first apply genome-wide ChIP profiling of YAP to systematically identify enhancers that are bound by YAP/TEAD4. Next, we implement a genetic approach to uncover functions of YAP/TEAD4-associated enhancers, demonstrate its robustness, and use it to reveal a network of enhancers required for YAP-mediated proliferation. We focus on EnhancerTRAM2, as its target gene TRAM2 shows the strongest expression-correlation with YAP activity in nearly all tumor types. Interestingly, TRAM2 phenocopies the YAP-induced cell proliferation, migration, and invasion phenotypes and correlates with poor patient survival. Mechanistically, we identify FSTL-1 as a major direct client of TRAM2 that is involved in these phenotypes. Thus, TRAM2 is a key novel mediator of YAP-induced oncogenic proliferation and cellular invasiveness. CONCLUSIONS YAP is a transcription co-factor that binds to thousands of enhancer loci and stimulates tumor aggressiveness. Using unbiased functional approaches, we dissect YAP enhancer network and characterize TRAM2 as a novel mediator of cellular proliferation, migration, and invasion. Our findings elucidate how YAP induces cancer aggressiveness and may assist diagnosis of cancer metastasis.
Collapse
Affiliation(s)
- Li Li
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Alejandro P. Ugalde
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Colinda L. G. J. Scheele
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Sebastian M. Dieter
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Remco Nagel
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Jin Ma
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Abhijeet Pataskar
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Gozde Korkmaz
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Miao-Ping Chien
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Li You
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pin-Rui Su
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Onno B. Bleijerveld
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Maarten Altelaar
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvt Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584CH Utrecht, The Netherlands
| | - Lyubomir Momchev
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Zohar Manber
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruiqi Han
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Pieter C. van Breugel
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Rui Lopes
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Peter ten Dijke
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Reuven Agami
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Erasmus MC, Rotterdam University, Rotterdam, The Netherlands
| |
Collapse
|
189
|
Kumar A, Ali M, Kumar R, Kumar M, Sagar P, Pandey RK, Akhouri V, Kumar V, Anand G, Niraj PK, Rani R, Kumar S, Kumar D, Bishwapriya A, Ghosh AK. Arsenic exposure in Indo Gangetic plains of Bihar causing increased cancer risk. Sci Rep 2021; 11:2376. [PMID: 33504854 PMCID: PMC7841152 DOI: 10.1038/s41598-021-81579-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/21/2020] [Indexed: 01/30/2023] Open
Abstract
Reportedly, 300 million people worldwide are affected by the consumption of arsenic contaminated groundwater. India prominently figures amongst them and the state of Bihar has shown an upsurge in cases affected by arsenic poisoning. Escalated arsenic content in blood, leaves 1 in every 100 human being highly vulnerable to being affected by the disease. Uncontrolled intake may lead to skin, kidney, liver, bladder, or lung related cancer but even indirect forms of cancer are showing up on a regular basis with abnormal arsenic levels as the probable cause. But despite the apparent relation, the etiology has not been understood clearly. Blood samples of 2000 confirmed cancer patients were collected from pathology department of our institute. For cross-sectional design, 200 blood samples of subjects free from cancer from arsenic free pockets of Patna urban agglomeration, were collected. Blood arsenic levels in carcinoma patients as compared to sarcomas, lymphomas and leukemia were found to be higher. The geospatial map correlates the blood arsenic with cancer types and the demographic area of Gangetic plains. Most of the cancer patients with high blood arsenic concentration were from the districts near the river Ganges. The raised blood arsenic concentration in the 2000 cancer patients strongly correlates the relationship of arsenic with cancer especially the carcinoma type which is more vulnerable. The average arsenic concentration in blood of the cancer patients in the Gangetic plains denotes the significant role of arsenic which is present in endemic proportions. Thus, the study significantly correlates and advocates a strong relation of the deleterious element with the disease. It also underlines the need to address the problem by deciphering the root cause of the elevated cancer incidences in the Gangetic basin of Bihar and its association with arsenic poisoning.
Collapse
Affiliation(s)
- Arun Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India.
| | - Mohammad Ali
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Ranjit Kumar
- Department of Animal Sciences, Central University of Himachal Pradesh, Kangra, Himachal Pradesh, India
| | - Mukesh Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Prity Sagar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Ritu Kumari Pandey
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Vivek Akhouri
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Vikas Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Gautam Anand
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Pintoo Kumar Niraj
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Rita Rani
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| | - Santosh Kumar
- Department of Applied Geoscience and Engineering, Delft University of Technology, Delft, The Netherlands
| | - Dhruv Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, India
| | | | - Ashok Kumar Ghosh
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar, 801505, India
| |
Collapse
|
190
|
Lee J, Jung Y, Jeong SW, Jeong GH, Moon GT, Kim M. Inhibition of Hippo Signaling Improves Skin Lesions in a Rosacea-Like Mouse Model. Int J Mol Sci 2021; 22:ijms22020931. [PMID: 33477764 PMCID: PMC7832320 DOI: 10.3390/ijms22020931] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 12/14/2022] Open
Abstract
The Hippo signaling pathway plays a key role in regulating organ size and tissue homeostasis. Hippo and two of its main effectors, yes-associated protein (YAP) and WWTR1 (WW domain-containing transcription regulator 1, commonly listed as TAZ), play critical roles in angiogenesis. This study investigated the role of the Hippo signaling pathway in the pathogenesis of rosacea. We performed immunohistochemical analyses to compare the expression levels of YAP and TAZ between rosacea skin and normal skin in humans. Furthermore, we used a rosacea-like BALB/c mouse model induced by LL-37 injections to determine the roles of YAP and TAZ in rosacea in vivo. We found that the expression levels of YAP and TAZ were upregulated in patients with rosacea. In the rosacea-like mouse model, we observed that the clinical features of rosacea, including telangiectasia and erythema, improved after the injection of a YAP/TAZ inhibitor. Additionally, treatment with a YAP/TAZ inhibitor reduced the expression levels of YAP and TAZ and diminished vascular endothelial growth factor (VEGF) immunoreactivity in the rosacea-like mouse model. Our findings suggest that YAP/TAZ inhibitors can attenuate angiogenesis associated with the pathogenesis of rosacea and that both YAP and TAZ are potential therapeutic targets for patients with rosacea.
Collapse
Affiliation(s)
- Jihyun Lee
- Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, #222 Banpo-daero, Seocho-gu, Seoul 06591, Korea;
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, #222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (G.H.J.); (G.T.M.)
| | - Yujin Jung
- Department of Dermatology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, #10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea; (Y.J.); (S.w.J.)
| | - Seo won Jeong
- Department of Dermatology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, #10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea; (Y.J.); (S.w.J.)
| | - Ga Hee Jeong
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, #222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (G.H.J.); (G.T.M.)
| | - Gue Tae Moon
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, #222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (G.H.J.); (G.T.M.)
| | - Miri Kim
- Department of Dermatology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, #10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea; (Y.J.); (S.w.J.)
- Correspondence: ; Tel.: +82-3779-1056
| |
Collapse
|
191
|
Masliantsev K, Karayan-Tapon L, Guichet PO. Hippo Signaling Pathway in Gliomas. Cells 2021; 10:184. [PMID: 33477668 PMCID: PMC7831924 DOI: 10.3390/cells10010184] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
The Hippo signaling pathway is a highly conserved pathway involved in tissue development and regeneration that controls organ size through the regulation of cell proliferation and apoptosis. The core Hippo pathway is composed of a block of kinases, MST1/2 (Mammalian STE20-like protein kinase 1/2) and LATS1/2 (Large tumor suppressor 1/2), which inhibits nuclear translocation of YAP/TAZ (Yes-Associated Protein 1/Transcriptional co-activator with PDZ-binding motif) and its downstream association with the TEAD (TEA domain) family of transcription factors. This pathway was recently shown to be involved in tumorigenesis and metastasis in several cancers such as lung, breast, or colorectal cancers but is still poorly investigated in brain tumors. Gliomas are the most common and the most lethal primary brain tumors representing about 80% of malignant central nervous system neoplasms. Despite intensive clinical protocol, the prognosis for patients remains very poor due to systematic relapse and treatment failure. Growing evidence demonstrating the role of Hippo signaling in cancer biology and the lack of efficient treatments for malignant gliomas support the idea that this pathway could represent a potential target paving the way for alternative therapeutics. Based on recent advances in the Hippo pathway deciphering, the main goal of this review is to highlight the role of this pathway in gliomas by a state-of-the-art synthesis.
Collapse
Affiliation(s)
- Konstantin Masliantsev
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| | - Lucie Karayan-Tapon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| | - Pierre-Olivier Guichet
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| |
Collapse
|
192
|
Yang S, Jiang W, Yang W, Yang C, Yang X, Chen K, Hu Y, Shen G, Lu L, Cheng F, Zhang F, Rao J, Wang X. Epigenetically modulated miR-1224 suppresses the proliferation of HCC through CREB-mediated activation of YAP signaling pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:944-958. [PMID: 33614242 PMCID: PMC7868928 DOI: 10.1016/j.omtn.2021.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Mounting evidence has demonstrated that microRNA-1224 (miR-1224) is commonly downregulated and serves as a tumor suppressor in multiple malignancies. However, the role and mechanisms responsible for miR-1224 in hepatocellular carcinoma (HCC) remain unclear. In this study, we found that the expression of miR-1224 was downregulated in HCC. Low miR-1224 expression was associated with poor clinicopathologic features and short overall survival. Moreover, the methylation status of putative CpG islands was also found to be an important part in the modulation of miR-1224 expression. miR-1224 could induce HCC cells to arrest in G0/G1 phase and inhibited the proliferation of HCC cells both in vitro and in vivo. Mechanistic investigation showed that by binding with cyclic AMP (cAMP)-response element binding protein (CREB) miR-1224 could repress the transcription and the activation of Yes-associated protein (YAP) signaling pathway. Furthermore, the expression of miR-1224 was inhibited by CREB through EZH2-mediated histone 3 lysine 27 (H3K27me3) on miR-1224 promoter, thus forming a positive feedback circuit. Our findings identify a miR-1224/CREB feedback loop for HCC progression and that blocking this circuit may represent a promising target for HCC treatment.
Collapse
Affiliation(s)
- Shikun Yang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Wei Jiang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 210500, China
| | - Wenjie Yang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Chao Yang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Xinchen Yang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Keyan Chen
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Yuanchang Hu
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Gefenqiang Shen
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Ling Lu
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Feng Cheng
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Feng Zhang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Jianhua Rao
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| | - Xuehao Wang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing 210029, China
| |
Collapse
|
193
|
Wang W, Todorov P, Isachenko E, Rahimi G, Mallmann P, Wang M, Isachenko V. In vitro activation of cryopreserved ovarian tissue: A single-arm meta-analysis and systematic review. Eur J Obstet Gynecol Reprod Biol 2021; 258:258-264. [PMID: 33485262 DOI: 10.1016/j.ejogrb.2021.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/22/2020] [Accepted: 01/10/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Primordial follicles in premature ovarian failure (POF) patients are very difficult to be activated spontaneously, so that mature oocytes are difficult to be obtained for in vitro fertilization. The aim of our review is to analyze and to systematize the published data regarding effectiveness of different strategies for in vitro activation of cryopreserved ovarian tissue. STUDY DESIGN According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a review of the literature was performed for all relevant full-text articles published in PubMed in English. Meta-analysis conducted using STATA 14.0. The random-effects model was used to combine 8 study results because the examination of heterogeneity was minimal. RESULTS One hundred and seventy seven patients after in vitro activation treatment (IVA) of ovarian tissue had accumulatively 26 pregnancies through IVF or natural pregnancy and then produced 18 live births. The random-effects model showed that the total clinical pregnancy and baby born rates reported in 8 studies evidence about effectiveness of IVA. CONCLUSION In vitro activation of primordial follicles as a new potential treatment for ovarian disorder patients, can be a promising option for fertility preservation. Drug-free activation of ovarian tissue in comparison with drug-included activation seemed to be more efficient.
Collapse
Affiliation(s)
- Wanxue Wang
- Research Group for Reproductive Medicine, University of Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
| | - Plamen Todorov
- Institute of Biology and Immunology of Reproduction, Tzarigradsko Shosse 73, 1113, Sofia, Bulgaria.
| | - Evgenia Isachenko
- Research Group for Reproductive Medicine, University of Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
| | - Gohar Rahimi
- Research Group for Reproductive Medicine, University of Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
| | - Peter Mallmann
- Research Group for Reproductive Medicine, University of Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
| | - Mengying Wang
- Research Group for Reproductive Medicine, University of Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
| | - Vladimir Isachenko
- Research Group for Reproductive Medicine, University of Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
| |
Collapse
|
194
|
Cwiklinski K, Robinson MW, Donnelly S, Dalton JP. Complementary transcriptomic and proteomic analyses reveal the cellular and molecular processes that drive growth and development of Fasciola hepatica in the host liver. BMC Genomics 2021; 22:46. [PMID: 33430759 PMCID: PMC7797711 DOI: 10.1186/s12864-020-07326-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background The major pathogenesis associated with Fasciola hepatica infection results from the extensive tissue damage caused by the tunnelling and feeding activity of immature flukes during their migration, growth and development in the liver. This is compounded by the pathology caused by host innate and adaptive immune responses that struggle to simultaneously counter infection and repair tissue damage. Results Complementary transcriptomic and proteomic approaches defined the F. hepatica factors associated with their migration in the liver, and the resulting immune-pathogenesis. Immature liver-stage flukes express ~ 8000 transcripts that are enriched for transcription and translation processes reflective of intensive protein production and signal transduction pathways. Key pathways that regulate neoblast/pluripotent cells, including the PI3K-Akt signalling pathway, are particularly dominant and emphasise the importance of neoblast-like cells for the parasite’s rapid development. The liver-stage parasites display different secretome profiles, reflecting their distinct niche within the host, and supports the view that cathepsin peptidases, cathepsin peptidase inhibitors, saposins and leucine aminopeptidases play a central role in the parasite’s destructive migration, and digestion of host tissue and blood. Immature flukes are also primed for countering immune attack by secreting immunomodulating fatty acid binding proteins (FABP) and helminth defence molecules (FhHDM). Combined with published host microarray data, our results suggest that considerable immune cell infiltration and subsequent fibrosis of the liver tissue exacerbates oxidative stress within parenchyma that compels the expression of a range of antioxidant molecules within both host and parasite. Conclusions The migration of immature F. hepatica parasites within the liver is associated with an increase in protein production, expression of signalling pathways and neoblast proliferation that drive their rapid growth and development. The secretion of a defined set of molecules, particularly cathepsin L peptidases, peptidase-inhibitors, saponins, immune-regulators and antioxidants allow the parasite to negotiate the liver micro-environment, immune attack and increasing levels of oxidative stress. This data contributes to the growing F. hepatica -omics information that can be exploited to understand parasite development more fully and for the design of novel control strategies to prevent host liver tissue destruction and pathology.
Collapse
Affiliation(s)
- Krystyna Cwiklinski
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland.
| | - Mark W Robinson
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sheila Donnelly
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland.,The School of Life Sciences, University of Technology, Sydney, Australia
| | - John P Dalton
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
195
|
Chen Q, Zhou XW, Zhang AJ, He K. ACTN1 supports tumor growth by inhibiting Hippo signaling in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:23. [PMID: 33413564 PMCID: PMC7791991 DOI: 10.1186/s13046-020-01821-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/20/2020] [Indexed: 11/18/2022]
Abstract
Background Alpha actinins (ACTNs) are major cytoskeletal proteins and exhibit many non-muscle functions. Emerging evidence have uncovered the regulatory role of ACTNs in tumorigenesis, however, the expression pattern, biological functions, and underlying mechanism of ACTN1 in hepatocellular carcinoma (HCC) remain largely unexplored. Methods Immunohistochemical analysis of a HCC tissue microarray (n = 157) was performed to determine the expression pattern and prognostic value of ACTN1 in HCC. In vitro loss-of-function study in HCC cells were carried out to investigate ACTN1 knockdown on cell proliferation. In vivo subcutaneous xenograft model and intrahepatic transplantation model were generated to decipher the contribution of ACTN1 in the tumor growth of HCC. Gene set enrichment analysis, quantitative real-time PCR, Co-immunoprecipitation, immunofluorescence and western blotting were performed to identify the underlying molecular mechanism. Results It was found that ACTN1 was significantly upregulated in HCC tissues and closely related to llpha-fetoprotein level, tumor thrombus, tumor size, TNM stage and patient prognoses. Knockdown of ACTN1 suppressed in vitro cell proliferation and in vivo tumor growth of HCC cells. Mechanistically, knockdown of ACTN1 increased Hippo signaling pathway activity and decreased Rho GTPases activities. Mechanistically, ACTN1 could competitively interact with MOB1 and decrease the phosphorylation of LATS1 and YAP. The growth-promoting effect induced by ACTN1 was significantly abrogated by pharmacological inhibition of YAP with verteporfin or super-TDU. Conclusions ACTN1 is highly expressed in HCC tissues and acts as a tumor promoter by suppressing Hippo signaling via physical interaction with MOB1. ACTN1 may serve as a potential prognostic marker and therapeutic target for HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-020-01821-6.
Collapse
Affiliation(s)
- Qian Chen
- Reproductive Medical Center, Department of Obstetrics and Gynecology of Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Xiao-Wei Zhou
- Reproductive Medical Center, Department of Obstetrics and Gynecology of Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Ai-Jun Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology of Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Kang He
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
196
|
Hu M, Wang H, Li S, Yan F, Fu C, Li L, Yu Y, Xiong J, Dong B. Yes-Associated Protein is Involved in Myocardial Fibrosis in Rats with Diabetic Cardiomyopathy. Diabetes Metab Syndr Obes 2021; 14:2133-2143. [PMID: 34012279 PMCID: PMC8128377 DOI: 10.2147/dmso.s302466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/01/2021] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION Recent studies have shown that YAP is closely related to the pathological process of cardiovascular diseases. But the role of YAP in cardiac injury of diabetic cardiomyopathy (DCM) is still unclear. METHODS Diabetic cardiomyopathy rat model was established and divided into control group, DCM group, LV-SC-shRNA group and LV-YAP-shRNA group. LV-SC-shRNA group and LV-YAP-shRNA group were injected with lentivirus expressing SC-shRNA and YAP-shRNA via tail vein, respectively. Primary rat cardiac fibroblasts (CFs) were stimulated with high concentration of glucose and treated with recombinant lentivirus expressing either SC-shRNA or YAP-shRNA to observe the expression of CTGF and fibronectin, so as to observe the effect of inhibiting YAP on the pathogenesis of DCM. RESULTS Compared with control group, high glucose markedly increased YAP mRNA and protein expression in DCM and CFs. Inhibition of YAP decreased myocardial fibrosis and improved cardiac function in the DCM model and decreased the expression of CTGF and fibronectin in CFs. The result suggested that YAP plays a key role in the pathological progression of DCM, and the underlying mechanisms may be associated with TEAD and CTGF. DISCUSSION We found that the expression of YAP was increased both in vivo and in vitro, suggesting that YAP is closely related to DCM, and YAP knockdown can reduce myocardial fibrosis in rat model of DCM by reducing the expression of PAI-1, collagen I, collagen III, CTGF and profilin, as well as the expression of CTGF and fibronectin in CFs. This study revealed that YAP plays an important role in the pathological process of diabetic cardiomyopathy, and down-regulation of YAP expression may provide a new therapeutic target for DCM.
Collapse
Affiliation(s)
- Maomao Hu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| | - Han Wang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Shengnan Li
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| | - Feng Yan
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| | - Changning Fu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| | - Lei Li
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| | - Yalin Yu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
- Department of Cardiology, Shandong Traditional Chinese Medicine University, Jinan, People’s Republic of China
| | - Jie Xiong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
- Department of Cardiology, Shandong Traditional Chinese Medicine University, Jinan, People’s Republic of China
- Correspondence: Bo Dong; Jie Xiong Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, 250012, People’s Republic of China Email ;
| |
Collapse
|
197
|
Liver regeneration: biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol 2021; 18:40-55. [PMID: 32764740 DOI: 10.1038/s41575-020-0342-4] [Citation(s) in RCA: 522] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 02/08/2023]
Abstract
The liver is the only solid organ that uses regenerative mechanisms to ensure that the liver-to-bodyweight ratio is always at 100% of what is required for body homeostasis. Other solid organs (such as the lungs, kidneys and pancreas) adjust to tissue loss but do not return to 100% of normal. The current state of knowledge of the regenerative pathways that underlie this 'hepatostat' will be presented in this Review. Liver regeneration from acute injury is always beneficial and has been extensively studied. Experimental models that involve partial hepatectomy or chemical injury have revealed extracellular and intracellular signalling pathways that are used to return the liver to equivalent size and weight to those prior to injury. On the other hand, chronic loss of hepatocytes, which can occur in chronic liver disease of any aetiology, often has adverse consequences, including fibrosis, cirrhosis and liver neoplasia. The regenerative activities of hepatocytes and cholangiocytes are typically characterized by phenotypic fidelity. However, when regeneration of one of the two cell types fails, hepatocytes and cholangiocytes function as facultative stem cells and transdifferentiate into each other to restore normal liver structure. Liver recolonization models have demonstrated that hepatocytes have an unlimited regenerative capacity. However, in normal liver, cell turnover is very slow. All zones of the resting liver lobules have been equally implicated in the maintenance of hepatocyte and cholangiocyte populations in normal liver.
Collapse
|
198
|
Huang M, Dong J, Guo H, Wang D. Effects of Dinotefuran on Brain miRNA Expression Profiles in Young Adult Honey Bees (Hymenopptera: Apidae). JOURNAL OF INSECT SCIENCE (ONLINE) 2021; 21:3. [PMID: 33400795 PMCID: PMC7785045 DOI: 10.1093/jisesa/ieaa131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 05/05/2023]
Abstract
Honey bees are important pollinators of wild plants and crops. MicroRNAs (miRNAs) are endogenous regulators of gene expression. In this study, we initially determined that the lethal concentration 50 (LC50) of dinotefuran was 0.773 mg/l. Then, the expression profiles and differentially expressed miRNAs (DE miRNAs) in honey bee brains after 1, 5, and 10 d of treatment with the lethal concentration 10 (LC10) of dinotefuran were explored via deep small-RNA sequencing and bioinformatics. In total, 2, 23, and 27 DE miRNAs were identified after persistent exposure to the LC10 of dinotefuran for 1, 5, and 10 d, respectively. Some abundant miRNAs, such as ame-miR-375-3p, ame-miR-281-5p, ame-miR-3786-3p, ame-miR-10-5p, and ame-miR-6037-3p, were extremely significantly differentially expressed. Enrichment analysis suggested that the candidate target genes of the DE miRNAs are involved in the regulation of biological processes, cellular processes, and behaviors. These results expand our understanding of the regulatory roles of miRNAs in honey bee Apis mellifera (Hymenopptera: Apidae) responses to neonicotinoid insecticides and facilitate further studies on the functions of miRNAs in honey bees.
Collapse
Affiliation(s)
- Minjie Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jie Dong
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Haikun Guo
- Institute of Quality and Standard for Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Deqian Wang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
199
|
Qian X, He L, Hao M, Li Y, Li X, Liu Y, Jiang H, Xu L, Li C, Wu W, Du L, Yin X, Lu Q. YAP mediates the interaction between the Hippo and PI3K/Akt pathways in mesangial cell proliferation in diabetic nephropathy. Acta Diabetol 2021; 58:47-62. [PMID: 32816106 DOI: 10.1007/s00592-020-01582-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022]
Abstract
AIMS Glomerular mesangial cell (MC) proliferation is one of the main pathological changes in diabetic nephropathy (DN), but its mechanism needs further elaboration. The Hippo and PI3K/Akt signalling pathways are involved in the regulation of MC proliferation, but their relationship in hyperglycaemia-induced MC proliferation has not been reported. METHODS We used db/db mice and high-glucose-cultured mesangial cells to generate a diabetic nephropathy model. An MST1-knockdown plasmid was used to identify whether the PI3K/Akt pathway is linked to the Hippo pathway through MST1. LY294002 and SC79 were used to verify the role of the PI3K/Akt signalling pathway in MC cells. RNA silencing and overexpression were performed by using YAP and PTEN-expression/knockdown plasmids to investigate the function of YAP and PTEN, respectively, in the Hippo and PI3K/Akt signalling pathways. RESULTS By examining a potential feedback loop, we found decreased phosphorylation of MST1 and Lats1 and increased PI3K/Akt activation in db/db mice and high glucose-treated MCs, along with increased MC proliferation. The results of our gene silencing experiment proved PI3K/Akt-mediated intervention in the Hippo pathway and the regulatory effect of YAP on PI3K/Akt through PTEN. CONCLUSIONS The Hippo pathway is inhibited under diabetic conditions, leading to YAP activation and promoting MC proliferation. The PI3K/Akt pathway is activated through the inhibitory effect of YAP on its repressor, PTEN. Finally, activation of the PI3K/Akt pathway inhibits the Hippo pathway, resulting in nuclear YAP accumulation and accelerating MC proliferation and DN formation.
Collapse
Affiliation(s)
- Xuan Qian
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Linlin He
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xizhi Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yiqi Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liu Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chengcheng Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenya Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
200
|
Baker K, Kwok E, Reardon P, Rodriguez DJ, Rolland AD, Wilson JW, Prell JS, Nyarko A. Yorkie-Warts Complexes are an Ensemble of Interconverting Conformers Formed by Multivalent Interactions. J Mol Biol 2020; 433:166776. [PMID: 33383033 DOI: 10.1016/j.jmb.2020.166776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 11/17/2022]
Abstract
Multiple copies of WW domains and PPXY motif sequences are often reciprocally presented by regulatory proteins that interact at crucial regulatory steps in the cell life cycle. While biophysical studies of single WW domain-single PPXY motif complexes abound in the literature, the molecular mechanisms of multivalent WW domain-PPXY assemblies are still poorly understood. By way of investigating such assemblies, we characterized the multivalent association of the entire cognate binding domains, two WW sequences and five PPXY motifs respectively, of the Yorkie transcription coactivator and the Warts tumor suppressor. Isothermal titration calorimetry, sedimentation velocity, size-exclusion chromatography coupled to multi-angle light scattering and native-state mass spectrometry of Yorkie WW domains interactions with the full-length Warts PPXY domain, and numerous PPXY motif variants of Warts show that the two proteins assemble via binding of tandem WW domains to adjacent PPXY pairs to produce an ensemble of interconverting complexes of variable stoichiometries, binding energetics and WW domain occupancy. Apparently, the Yorkie tandem WW domains first target the two adjacent PPXY motifs at the C-terminus of the Warts polypeptide and additional WW domains bind unoccupied motifs. Similar ensembles of interconverting conformers may be common in multivalent WW domain-PPXY interactions to promote the adaptability and versatility of WW domain-PPXY mediated cellular processes.
Collapse
Affiliation(s)
- Kasie Baker
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Ethiene Kwok
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Patrick Reardon
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Diego J Rodriguez
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Amber D Rolland
- Department of Chemistry & Biochemistry, University of Oregon, Eugene, OR 97403, USA; Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Jesse W Wilson
- Department of Chemistry & Biochemistry, University of Oregon, Eugene, OR 97403, USA; Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - James S Prell
- Department of Chemistry & Biochemistry, University of Oregon, Eugene, OR 97403, USA; Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Afua Nyarko
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|