1
|
Yue L, Lu Z, Guo T, Liu J, Yuan C, Yang B. Association of SLIT3 and ZNF280B Gene Polymorphisms with Wool Fiber Diameter. Animals (Basel) 2023; 13:3552. [PMID: 38003169 PMCID: PMC10668676 DOI: 10.3390/ani13223552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/06/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
The SLIT3 gene encodes a secreted protein, and the ZNF280B gene is a member of the transcription factor family. Both genes have multiple biological functions. This study was conducted to investigate the association between SLIT3 and ZNF280B gene polymorphisms and wool fiber diameter and to determine potential molecular marker sites for breeding sheep with fine wool. We used Kompetitive Allele-Specific PCR to type the single nucleotide polymorphism (SNP) loci in the SLIT3 and ZNF280B genes within 1081 Alpine Merino sheep and associated these SNPs with wool fiber diameter. The results revealed one SNP in SLIT3 and ZNF280B, which were each related to sheep fiber diameter. The wool fiber diameters of sheep with the CC genotype in SLIT3 g.478807C>G and AA genotype in ZNF280B g.677G>A were the smallest and differed significantly from the diameters of other genotypes (p < 0.05). These results suggest potential molecular marker sites for fine-wool sheep breeding.
Collapse
Affiliation(s)
- Lin Yue
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (L.Y.); (J.L.)
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Zengkui Lu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (L.Y.); (J.L.)
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Tingting Guo
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (L.Y.); (J.L.)
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Jianbin Liu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (L.Y.); (J.L.)
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Chao Yuan
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (L.Y.); (J.L.)
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Bohui Yang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (L.Y.); (J.L.)
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| |
Collapse
|
2
|
Mascarenhas R, Meirelles PM, Batalha-Filho H. Urbanization drives adaptive evolution in a Neotropical bird. Curr Zool 2023; 69:607-619. [PMID: 37637315 PMCID: PMC10449428 DOI: 10.1093/cz/zoac066] [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] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/16/2022] [Indexed: 08/29/2023] Open
Abstract
Urbanization has dramatic impacts on natural habitats and such changes may potentially drive local adaptation of urban populations. Behavioral change has been specifically shown to facilitate the fast adaptation of birds to changing environments, but few studies have investigated the genetic mechanisms of this process. Such investigations could provide insights into questions about both evolutionary theory and management of urban populations. In this study, we investigated whether local adaptation has occurred in urban populations of a Neotropical bird species, Coereba flaveola, specifically addressing whether observed behavioral adaptations are correlated to genetic signatures of natural selection. To answer this question, we sampled 24 individuals in urban and rural environments, and searched for selected loci through a genome-scan approach based on RADseq genomic data, generated and assembled using a reference genome for the species. We recovered 46 loci as putative selection outliers, and 30 of them were identified as associated with biological processes possibly related to urban adaptation, such as the regulation of energetic metabolism, regulation of genetic expression, and changes in the immunological system. Moreover, genes involved in the development of the nervous system showed signatures of selection, suggesting a link between behavioral and genetic adaptations. Our findings, in conjunction with similar results in previous studies, support the idea that cities provide a similar selective pressure on urban populations and that behavioral plasticity may be enhanced through genetic changes in urban populations.
Collapse
Affiliation(s)
- Rilquer Mascarenhas
- National Institute of Science and Technology in Interdisciplinary and Transdisciplinary Studies in Ecology and Evolution (INCT IN-TREE), Instituto de Biologia, Universidade Federal da Bahia, 40170-115 Salvador, Bahia, Brazil
| | - Pedro Milet Meirelles
- National Institute of Science and Technology in Interdisciplinary and Transdisciplinary Studies in Ecology and Evolution (INCT IN-TREE), Instituto de Biologia, Universidade Federal da Bahia, 40170-115 Salvador, Bahia, Brazil
| | - Henrique Batalha-Filho
- National Institute of Science and Technology in Interdisciplinary and Transdisciplinary Studies in Ecology and Evolution (INCT IN-TREE), Instituto de Biologia, Universidade Federal da Bahia, 40170-115 Salvador, Bahia, Brazil
| |
Collapse
|
3
|
Zhu Q, Zhao X, Zhang D, Xia W, Zhang J. Abnormal expression of SLIT3 induces intravillous vascularization dysplasia in ectopic pregnancy. PeerJ 2023; 11:e14850. [PMID: 36793891 PMCID: PMC9924138 DOI: 10.7717/peerj.14850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 01/12/2023] [Indexed: 02/12/2023] Open
Abstract
Objective To investigate whether the morphology, capillary number, and transcriptome expression profiles of ectopic pregnancy (EP) villi differ from those of normal pregnancy (NP) villi. Methods Hematoxylin-eosin (HE) and immunohistochemistry (IHC) staining for CD31 were conducted to compare differences in morphology and capillary number between EP and NP villi. Differentially expressed (DE) miRNAs and mRNAs were determined from transcriptome sequencing of both types of villi and used to construct a miRNA-mRNA network, from which hub genes were identified. Candidate DE-miRNAs and DE-mRNAs were validated by quantitative reverse transcription (qRT)-PCR. Correlations were identified between the number of capillaries and serum beta human chorionic gonadotropin (β-HCG) levels and between the expression levels of hub genes associated with angiogenesis and β-HCG levels. Results The mean and total cross-sectional areas of placental villi were significantly increased in EP compared with NP villi. Capillary density was greatly reduced in EP villi and was positively correlated with β-HCG levels. A total of 49 DE-miRNAs and 625 DE-mRNAs were identified from the sequencing data. An integrated analysis established a miRNA-mRNA network containing 32 DE-miRNAs and 103 DE-mRNAs. Based on the validation of hub mRNAs and miRNAs in the network, a regulatory pathway involving miR-491-5p-SLIT3 was discovered, which may have a role in the development of villous capillaries. Conclusion Villus morphology, capillary number, and miRNA/mRNA expression profiles in villous tissues were aberrant in EP placentas. Specifically, SLIT3, which is regulated by miR-491-5p, may contribute to the regulation of villous angiogenesis and was established as a putative predictor of chorionic villus development, providing a basis for future research.
Collapse
Affiliation(s)
- Qian Zhu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Xiaoya Zhao
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Duo Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Wei Xia
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Jian Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Municipal Key Clinical Specialty, Shanghai, China
| |
Collapse
|
4
|
Münch J, Engesser M, Schönauer R, Hamm JA, Hartig C, Hantmann E, Akay G, Pehlivan D, Mitani T, Coban Akdemir Z, Tüysüz B, Shirakawa T, Dateki S, Claus LR, van Eerde AM, Smol T, Devisme L, Franquet H, Attié-Bitach T, Wagner T, Bergmann C, Höhn AK, Shril S, Pollack A, Wenger T, Scott AA, Paolucci S, Buchan J, Gabriel GC, Posey JE, Lupski JR, Petit F, McCarthy AA, Pazour GJ, Lo CW, Popp B, Halbritter J. Biallelic pathogenic variants in roundabout guidance receptor 1 associate with syndromic congenital anomalies of the kidney and urinary tract. Kidney Int 2022; 101:1039-1053. [PMID: 35227688 PMCID: PMC10010616 DOI: 10.1016/j.kint.2022.01.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 11/30/2021] [Accepted: 01/11/2022] [Indexed: 11/16/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) represent the most common cause of chronic kidney failure in children. Despite growing knowledge of the genetic causes of CAKUT, the majority of cases remain etiologically unsolved. Genetic alterations in roundabout guidance receptor 1 (ROBO1) have been associated with neuronal and cardiac developmental defects in living individuals. Although Slit-Robo signaling is pivotal for kidney development, diagnostic ROBO1 variants have not been reported in viable CAKUT to date. By next-generation-sequencing methods, we identified six unrelated individuals and two non-viable fetuses with biallelic truncating or combined missense and truncating variants in ROBO1. Kidney and genitourinary manifestation included unilateral or bilateral kidney agenesis, vesicoureteral junction obstruction, vesicoureteral reflux, posterior urethral valve, genital malformation, and increased kidney echogenicity. Further clinical characteristics were remarkably heterogeneous, including neurodevelopmental defects, intellectual impairment, cerebral malformations, eye anomalies, and cardiac defects. By in silico analysis, we determined the functional significance of identified missense variants and observed absence of kidney ROBO1 expression in both human and murine mutant tissues. While its expression in multiple tissues may explain heterogeneous organ involvement, variability of the kidney disease suggests gene dosage effects due to a combination of null alleles with mild hypomorphic alleles. Thus, comprehensive genetic analysis in CAKUT should include ROBO1 as a new cause of recessively inherited disease. Hence, in patients with already established ROBO1-associated cardiac or neuronal disorders, screening for kidney involvement is indicated.
Collapse
Affiliation(s)
- Johannes Münch
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany; Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Marie Engesser
- Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Ria Schönauer
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany; Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - J Austin Hamm
- East Tennessee Children's Hospital, Genetic Center, Knoxville, Tennessee, USA
| | - Christin Hartig
- Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Elena Hantmann
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany; Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Gulsen Akay
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Department of Pediatrics, University of Utah, Salt Lake, Utah, USA
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; Texas Children's Hospital, Houston, Texas, USA
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Zeynep Coban Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Beyhan Tüysüz
- Department of Pediatric Genetics, Istanbul University Cerrahpasa Medical Faculty, Istanbul, Turkey
| | | | - Sumito Dateki
- Department of Pediatrics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Laura R Claus
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Thomas Smol
- Centre Hospitalier Universitaire de Lille, Institut de Génétique Médicale, Lille, France
| | - Louise Devisme
- Centre Hospitalier Universitaire de Lille, Institut de Pathologie, Lille, France
| | - Hélène Franquet
- Centre Hospitalier Universitaire de Lille, Institut de Pathologie, Lille, France
| | - Tania Attié-Bitach
- Laboratoire de biologie médicale multisites SeqOIA, Paris, France; Service de Médecine Génomique des Maladies Rares, APHP.Centre, Université de Paris, Paris, France
| | - Timo Wagner
- Medizinische Genetik Mainz, Limbach Genetics, Mainz, Germany
| | - Carsten Bergmann
- Medizinische Genetik Mainz, Limbach Genetics, Mainz, Germany; Department of Medicine, Nephrology, University Hospital Freiburg, Freiburg, Germany
| | - Anne Kathrin Höhn
- Division of Pathology, University of Leipzig Medical Center, Leipzig, Germany
| | - Shirlee Shril
- Division of Nephrology, Boston Children's Hospital, Boston, USA
| | - Ari Pollack
- Division of Genetic Medicine, University of Washington, Seattle, Washington, USA
| | - Tara Wenger
- Division of Genetic Medicine, University of Washington, Seattle, Washington, USA
| | - Abbey A Scott
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, Washington, USA
| | - Sarah Paolucci
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jillian Buchan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Texas Children's Hospital, Houston, Texas, USA; Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Florence Petit
- Centre Hospitalier Universitaire de Lille, Clinique de Génétique Guy Fontaine, Lille, France
| | | | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Bernt Popp
- Institute for Human Genetics, University of Leipzig Medical Center, Leipzig, Germany.
| | - Jan Halbritter
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany; Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
5
|
Honeycutt SE, N'Guetta PEY, O'Brien LL. Innervation in organogenesis. Curr Top Dev Biol 2022; 148:195-235. [PMID: 35461566 PMCID: PMC10636594 DOI: 10.1016/bs.ctdb.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Proper innervation of peripheral organs helps to maintain physiological homeostasis and elicit responses to external stimuli. Disruptions to normal function can result in pathophysiological consequences. The establishment of connections and communication between the central nervous system and the peripheral organs is accomplished through the peripheral nervous system. Neuronal connections with target tissues arise from ganglia partitioned throughout the body. Organ innervation is initiated during development with stimuli being conducted through several types of neurons including sympathetic, parasympathetic, and sensory. While the physiological modulation of mature organs by these nerves is largely understood, their role in mammalian development is only beginning to be uncovered. Interactions with cells in target tissues can affect the development and eventual function of several organs, highlighting their significance. This chapter will cover the origin of peripheral neurons, factors mediating organ innervation, and the composition and function of organ-specific nerves during development. This emerging field aims to identify the functional contribution of innervation to development which will inform future investigations of normal and abnormal mammalian organogenesis, as well as contribute to regenerative and organ replacement efforts where nerve-derived signals may have significant implications for the advancement of such studies.
Collapse
Affiliation(s)
- Samuel E Honeycutt
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Pierre-Emmanuel Y N'Guetta
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lori L O'Brien
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
6
|
Zhu H, Wang J, Nie W, Armando I, Han F. ADAMs family in kidney physiology and pathology. EBioMedicine 2021; 72:103628. [PMID: 34653870 PMCID: PMC8517843 DOI: 10.1016/j.ebiom.2021.103628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 11/21/2022] Open
Abstract
A disintegrin and metalloproteinases (ADAMs) family are proteolytic transmembrane proteases that modulate diverse cell functions and coordinate intercellular communication. ADAMs are responsible for regulating cell proliferation, differentiation, migration, and organ morphogenesis in kidney development. Abnormally activated ADAMs drive inflammation and fibrosis in response to kidney diseases such as acute kidney injury, diabetic kidney disease, polycystic kidney disease, and chronic allograft nephropathy. ADAM10 and ADAM17, known as the most characterized members of ADAMs, are extensively investigated in kidney diseases. Notably, ADAM proteases have the potential to be targets for developing novel treatment approaches in kidney diseases.
Collapse
Affiliation(s)
- Huanhuan Zhu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Junni Wang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wanyun Nie
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ines Armando
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Chan K, Li X. Current Epigenetic Insights in Kidney Development. Genes (Basel) 2021; 12:genes12081281. [PMID: 34440455 PMCID: PMC8391601 DOI: 10.3390/genes12081281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/07/2021] [Accepted: 08/19/2021] [Indexed: 12/31/2022] Open
Abstract
The kidney is among the best characterized developing tissues, with the genes and signaling pathways that regulate embryonic and adult kidney patterning and development having been extensively identified. It is now widely understood that DNA methylation and histone modification patterns are imprinted during embryonic development and must be maintained in adult cells for appropriate gene transcription and phenotypic stability. A compelling question then is how these epigenetic mechanisms play a role in kidney development. In this review, we describe the major genes and pathways that have been linked to epigenetic mechanisms in kidney development. We also discuss recent applications of single-cell RNA sequencing (scRNA-seq) techniques in the study of kidney development. Additionally, we summarize the techniques of single-cell epigenomics, which can potentially be used to characterize epigenomes at single-cell resolution in embryonic and adult kidneys. The combination of scRNA-seq and single-cell epigenomics will help facilitate the further understanding of early cell lineage specification at the level of epigenetic modifications in embryonic and adult kidney development, which may also be used to investigate epigenetic mechanisms in kidney diseases.
Collapse
Affiliation(s)
- Katrina Chan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
| | - Xiaogang Li
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: ; Tel.: +1-507-266-0110
| |
Collapse
|
8
|
Kaya TB, Aydemir O, Ceylaner S, Ceylaner G, Tekin AN. Isolated congenital diaphragm hernia associated with homozygous SLIT3 gene variant in dizygous twins. Eur J Med Genet 2021; 64:104215. [PMID: 33933663 DOI: 10.1016/j.ejmg.2021.104215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 11/16/2022]
Abstract
Congenital diaphragmatic hernia (CDH) is a serious life-threatening birth defect characterized by abnormal development in the muscular or tendinous portion of the diaphragm during embryogenesis. Despite its high incidence, the etiology of CDH hasn't been fully understood. Genetic factors are important in pathogenesis; however, few single genes have been definitively implicated in human CDH. SLIT1, SLIT2, and SLIT3 (slit guidance ligand) are three human homologs of the drosophila Slit gene. They interact with roundabout (Robo) homolog receptors to affect cell migration, adhesion, cell motility, and angiogenesis and play important roles in cell signaling pathways including the guidance of axons. In this report, we presented dizygous twin babies with CDH related to the SLIT3 gene variant. Previous studies showed that Slit3 null mice had congenital diaphragmatic hernias on or near the ventral midline portion of the central tendon. This is the first report of homozygous SLIT3 variant associated with CDH in humans.
Collapse
Affiliation(s)
- Tugba Barsan Kaya
- Eskişehir Osmangazi University Faculty of Medicine, Department of Neonatology, Eskişehir, Turkey.
| | - Ozge Aydemir
- Eskişehir Osmangazi University Faculty of Medicine, Department of Neonatology, Eskişehir, Turkey
| | | | | | - Ayse Neslihan Tekin
- Eskişehir Osmangazi University Faculty of Medicine, Department of Neonatology, Eskişehir, Turkey
| |
Collapse
|
9
|
Szemes M, Melegh Z, Bellamy J, Park JH, Chen B, Greenhough A, Catchpoole D, Malik K. Transcriptomic Analyses of MYCN-Regulated Genes in Anaplastic Wilms' Tumour Cell Lines Reveals Oncogenic Pathways and Potential Therapeutic Vulnerabilities. Cancers (Basel) 2021; 13:656. [PMID: 33562123 PMCID: PMC7915280 DOI: 10.3390/cancers13040656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
The MYCN proto-oncogene is deregulated in many cancers, most notably in neuroblastoma, where MYCN gene amplification identifies a clinical subset with very poor prognosis. Gene expression and DNA analyses have also demonstrated overexpression of MYCN mRNA, as well as focal amplifications, copy number gains and presumptive change of function mutations of MYCN in Wilms' tumours with poorer outcomes, including tumours with diffuse anaplasia. Surprisingly, however, the expression and functions of the MYCN protein in Wilms' tumours still remain obscure. In this study, we assessed MYCN protein expression in primary Wilms' tumours using immunohistochemistry of tissue microarrays. We found MYCN protein to be expressed in tumour blastemal cells, and absent in stromal and epithelial components. For functional studies, we used two anaplastic Wilms' tumour cell-lines, WiT49 and 17.94, to study the biological and transcriptomic effects of MYCN depletion. We found that MYCN knockdown consistently led to growth suppression but not cell death. RNA sequencing identified 561 MYCN-regulated genes shared by WiT49 and 17.94 cell-lines. As expected, numerous cellular processes were downstream of MYCN. MYCN positively regulated the miRNA regulator and known Wilms' tumour oncogene LIN28B, the genes encoding methylosome proteins PRMT1, PRMT5 and WDR77, and the mitochondrial translocase genes TOMM20 and TIMM50. MYCN repressed genes including the developmental signalling receptor ROBO1 and the stromal marker COL1A1. Importantly, we found that MYCN also repressed the presumptive Wilms' tumour suppressor gene REST, with MYCN knockdown resulting in increased REST protein and concomitant repression of RE1-Silencing Transcription factor (REST) target genes. Together, our study identifies regulatory axes that interact with MYCN, providing novel pathways for potential targeted therapeutics for poor-prognosis Wilms' tumour.
Collapse
Affiliation(s)
- Marianna Szemes
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; (J.B.); (J.H.P.); (B.C.); (A.G.)
| | - Zsombor Melegh
- Department of Cellular Pathology, Southmead Hospital, Bristol BS10 5NB, UK;
| | - Jacob Bellamy
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; (J.B.); (J.H.P.); (B.C.); (A.G.)
| | - Ji Hyun Park
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; (J.B.); (J.H.P.); (B.C.); (A.G.)
| | - Biyao Chen
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; (J.B.); (J.H.P.); (B.C.); (A.G.)
| | - Alexander Greenhough
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; (J.B.); (J.H.P.); (B.C.); (A.G.)
- Department of Applied Sciences, University of the West of England, Bristol BS16 1QY, UK
| | - Daniel Catchpoole
- The Kids Research Institute, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
| | - Karim Malik
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; (J.B.); (J.H.P.); (B.C.); (A.G.)
| |
Collapse
|
10
|
Rafipay A, Dun X, Parkinson DB, Erskine L, Vargesson N. Knockdown of slit signaling during limb development leads to a reduction in humerus length. Dev Dyn 2021; 250:1340-1357. [DOI: 10.1002/dvdy.284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Alexandra Rafipay
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Xin‐Peng Dun
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - Lynda Erskine
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Neil Vargesson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| |
Collapse
|
11
|
Zhang Y, Hu L, Li X, Chen L, Yang X. Slit2 is a potential biomarker for renal impairment in systemic lupus erythematosus. Clin Exp Med 2020; 21:63-71. [PMID: 33079290 DOI: 10.1007/s10238-020-00664-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/14/2020] [Indexed: 12/29/2022]
Abstract
Slit2 glycoprotein has been described to regulate the inflammatory response and be involved in autoimmune diseases. Here, we investigated the expression of Slit2 and its potential significance in systemic lupus erythematosus (SLE). A total of 103 patients with SLE participated in our study. The levels of serum Slit2 were measured by enzyme-linked immunosorbent assay, and the expression of Slit2 in renal tissue was detected by immunohistochemistry. Patients with active disease had higher levels of serum Slit2 than patients with inactive disease and controls. Patients with sole skin impairment or sole renal impairment or both skin and renal impairment had higher levels of serum Slit2 than patients with neither skin nor renal impairment. Patients with chronic kidney disease (CKD) had higher levels of serum Slit2 than patients with no CKD. Levels of serum Slit2 in patients with active disease were positively correlated with the SLE Disease Activity Index, complement C4, and anti-dsDNA antibody. Levels of serum Slit2 in patients with CKD were positively correlated with serum creatinine, urine protein, and glomerular filtration rate. The expression of Slit2 and its receptor Roundabout1 (Robo1) in the renal tissue of patients with lupus nephritis were higher than controls. Moreover, renal Slit2 was positively correlated with renal chronic index. Our data indicated that Slit2 may contribute to renal impairment and this may be a potential biomarker for SLE.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China.,Department of Medicine, Hangzhou Dingqiao Hospital, Hangzhou, 310021, People's Republic of China
| | - Lingzhen Hu
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
| | - Xiang Li
- Clinical Laboratory, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
| | - Liheng Chen
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
| | - Xuyan Yang
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China.
| |
Collapse
|
12
|
Gong L, Wang S, Shen L, Liu C, Shenouda M, Li B, Liu X, Shaw JA, Wineman AL, Yang Y, Xiong D, Eichmann A, Evans SM, Weiss SJ, Si MS. SLIT3 deficiency attenuates pressure overload-induced cardiac fibrosis and remodeling. JCI Insight 2020; 5:136852. [PMID: 32644051 PMCID: PMC7406261 DOI: 10.1172/jci.insight.136852] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/06/2020] [Indexed: 01/28/2023] Open
Abstract
In pulmonary hypertension and certain forms of congenital heart disease, ventricular pressure overload manifests at birth and is an obligate hemodynamic abnormality that stimulates myocardial fibrosis, which leads to ventricular dysfunction and poor clinical outcomes. Thus, an attractive strategy is to attenuate the myocardial fibrosis to help preserve ventricular function. Here, by analyzing RNA-sequencing databases and comparing the transcript and protein levels of fibrillar collagen in WT and global-knockout mice, we found that slit guidance ligand 3 (SLIT3) was present predominantly in fibrillar collagen-producing cells and that SLIT3 deficiency attenuated collagen production in the heart and other nonneuronal tissues. We then performed transverse aortic constriction or pulmonary artery banding to induce left and right ventricular pressure overload, respectively, in WT and knockout mice. We discovered that SLIT3 deficiency abrogated fibrotic and hypertrophic changes and promoted long-term ventricular function and overall survival in both left and right ventricular pressure overload. Furthermore, we found that SLIT3 stimulated fibroblast activity and fibrillar collagen production, which coincided with the transcription and nuclear localization of the mechanotransducer yes-associated protein 1. These results indicate that SLIT3 is important for regulating fibroblast activity and fibrillar collagen synthesis in an autocrine manner, making it a potential therapeutic target for fibrotic diseases, especially myocardial fibrosis and adverse remodeling induced by persistent afterload elevation.
Collapse
Affiliation(s)
- Lianghui Gong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuyun Wang
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Li Shen
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Catherine Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mena Shenouda
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Baolei Li
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaoxiao Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Alan L. Wineman
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dingding Xiong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Paris Cardiovascular Research Center, INSERM U970, Paris, France.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences,,Department of Medicine, and,Department of Pharmacology, UCSD, La Jolla, California, USA
| | - Stephen J. Weiss
- Division of Genetic Medicine,,Department of Internal Medicine,,Life Sciences Institute,,Cellular and Molecular Biology Graduate Program, and,Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ming-Sing Si
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Wu MF, Chuang CY, Lin P, Chen WT, Su SE, Liao CY, Jan MS, Chang JT. Lung Tumorigenesis Alters the Expression of Slit2-exon15 Splicing Variants in Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11020166. [PMID: 30717252 PMCID: PMC6406468 DOI: 10.3390/cancers11020166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/22/2019] [Accepted: 01/29/2019] [Indexed: 12/02/2022] Open
Abstract
Slit2 expression is downregulated in various cancers, including lung cancer. We identified two Slit2 splicing variants at exon15—Slit2-WT and Slit2-ΔE15. In the RT-PCR analyses, the Slit2-WT isoform was predominantly expressed in all the lung cancer specimens and in their normal lung counterparts, whereas Slit2-ΔE15 was equivalently or predominantly expressed in 41% of the pneumothorax specimens. A kRasG12D transgenic mice system was used to study the effects of tumorigenesis on the expressions of the Slit2-exon15 isoforms. The results revealed that a kRasG12D-induced lung tumor increased the Slit2-WT/Slit2-ΔE15 ratio and total Slit2 expression level. However, the lung tumors generated via a tail vein injection of lung cancer cells decreased the Slit2-WT/Slit2-ΔE15 ratio and total Slit2 expression level. Interestingly, the lipopolysaccharide (LPS)-induced lung inflammation also decreased the Slit2-WT/Slit2-ΔE15 ratio. Since Slit2 functions as an anti-inflammatory factor, the expression of Slit2 increases in kRasG12D lungs, which indicates that Slit2 suppresses immunity during tumorigenesis. However, an injection of lung cancer cells via the tail vein and the LPS-induced lung inflammation both decreased the Slit2 expression. The increased Slit2 in the tumor microenvironment was mostly Slit2-WT, which lacks growth inhibitory activity. Thus, the results of our study suggested that the upregulation of Slit2-WT, but not Slit2-ΔE15, in a cancer microenvironment is an important factor in suppressing immunity while not interfering with cancer growth.
Collapse
Affiliation(s)
- Ming-Fang Wu
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Cheng-Yen Chuang
- Division of Thoracic Surgery, Taichung Veterans General Hospital, Taichung 40705 Taiwan.
| | - Pinpin Lin
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Wei-Ting Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Shang-Er Su
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Chen-Yi Liao
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Ming-Shiou Jan
- Department of Microbiology and Immunology, Chung Shan Medical University, Taichung 40201, Taiwan.
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Jinghua Tsai Chang
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| |
Collapse
|
14
|
Yoshimura Y, Taguchi A, Tanigawa S, Yatsuda J, Kamba T, Takahashi S, Kurihara H, Mukoyama M, Nishinakamura R. Manipulation of Nephron-Patterning Signals Enables Selective Induction of Podocytes from Human Pluripotent Stem Cells. J Am Soc Nephrol 2019; 30:304-321. [PMID: 30635375 DOI: 10.1681/asn.2018070747] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/03/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Previous research has elucidated the signals required to induce nephron progenitor cells (NPCs) from pluripotent stem cells (PSCs), enabling the generation of kidney organoids. However, selectively controlling differentiation of NPCs to podocytes has been a challenge. METHODS We investigated the effects of various growth factors in cultured mouse embryonic NPCs during three distinct steps of nephron patterning: from NPC to pretubular aggregate, from the latter to epithelial renal vesicle (RV), and from RV to podocyte. We then applied the findings to human PSC-derived NPCs to establish a method for selective induction of human podocytes. RESULTS Mouse NPC differentiation experiments revealed that phase-specific manipulation of Wnt and Tgf-β signaling is critical for podocyte differentiation. First, optimal timing and intensity of Wnt signaling were essential for mesenchymal-to-epithelial transition and podocyte differentiation. Then, inhibition of Tgf-β signaling supported domination of the RV proximal domain. Inhibition of Tgf-β signaling in the third phase enriched the podocyte fraction by suppressing development of other nephron lineages. The resultant protocol enabled successful induction of human podocytes from PSCs with >90% purity. The induced podocytes exhibited global gene expression signatures comparable to those of adult human podocytes, had podocyte morphologic features (including foot process-like and slit diaphragm-like structures), and showed functional responsiveness to drug-induced injury. CONCLUSIONS Elucidation of signals that induce podocytes during the nephron-patterning process enabled us to establish a highly efficient method for selective induction of human podocytes from PSCs. These PSC-derived podocytes show molecular, morphologic, and functional characteristics of podocytes, and offer a new resource for disease modeling and nephrotoxicity testing.
Collapse
Affiliation(s)
- Yasuhiro Yoshimura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and.,Departments of Nephrology and
| | - Atsuhiro Taguchi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and .,Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and
| | - Junji Yatsuda
- Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomomi Kamba
- Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan; and
| | - Hidetake Kurihara
- Department of Anatomy and Life Structure, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and
| |
Collapse
|
15
|
Liu J, Hou W, Guan T, Tang L, Zhu X, Li Y, Hou S, Zhang J, Chen H, Huang Y. Slit2/Robo1 signaling is involved in angiogenesis of glomerular endothelial cells exposed to a diabetic-like environment. Angiogenesis 2018; 21:237-249. [PMID: 29299781 DOI: 10.1007/s10456-017-9592-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/26/2017] [Indexed: 01/08/2023]
Abstract
Abnormal angiogenesis plays a pathological role in diabetic nephropathy (DN), contributing to glomerular hypertrophy and microalbuminuria. Slit2/Robo1 signaling participates in angiogenesis in some pathological contexts, but whether it is involved in glomerular abnormal angiogenesis of early DN is unclear. The present study evaluated the effects of Slit2/Robo1 signaling pathway on angiogenesis of human renal glomerular endothelial cells (HRGECs) exposed to a diabetic-like environment or recombinant Slit2-N. To remove the effect of Slit2 derived from mesangial cells, human renal mesangial cells (HRMCs) grown in high glucose (HG) medium (33 mM) were transfected with Slit2 siRNA and then the HG-HRMCs-CM with Slit2 depletion was collected after 48 h. HRGECs were cultured in the HG-HRMCs-CM or recombinant Slit2-N for 0, 6, 12, 24, or 48 h. The mRNA and protein expressions of Slit2/Robo1, PI3K/Akt and HIF-1α/VEGF signaling pathways were detected by quantitative real-time PCR, western blotting, and ELISA, respectively. The CCK-8 cell proliferation assay, flow cytometry and the scratch wound-healing assay were used to assess cell proliferation, cycles, and migration, respectively. Matrigel was used to perform a tubule formation assay. Our results showed that the HG-HRMCs-CM with Slit2 depletion enhanced the activation of Slit2/Robo1, PI3K/Akt, and HIF-1α/VEGF signaling in HRGECs in time-dependent manner (0-24 h post-treatment). In addition, the HG-HRMCs-CM with Slit2 depletion significantly promoted HRGECs proliferation, migration, and tube formation. Pretreatment of HRGECs with Robo1 siRNA suppressed the activation of PI3K/Akt and HIF-1α/VEGF signaling and inhibited angiogenesis, whereas PI3K inhibitor suppressed HIF-1α/VEGF signaling, without influencing Robo1 expression. In the HRGECs treated with Slit2-N, Slit2-N time-dependently enhanced the activation of Robo1/PI3K/Akt/VEGF pathway but not HIF-1α activity, and promoted HRGECs proliferation, migration, and tube formation. The effects induced by Slit2 were also abolished by Robo1 siRNA and PI3K inhibitor. Taken together, our findings indicate that in a diabetic-like environment, in addition to mesangial cells, autocrine activation of Slit2/Robo1 signaling of HRGECs may contribute to angiogenesis of HRGECs through PI3K/Akt/VEGF pathway; therefore, Slit2/Robo1 signaling may be a potent therapeutic target for the treatment of abnormal angiogenesis in early DN and may have broad implications for the treatment of other diseases dependent on pathologic angiogenesis.
Collapse
Affiliation(s)
- Junhui Liu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Weiping Hou
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Tao Guan
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Luyao Tang
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xufei Zhu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yi Li
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Shihui Hou
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jun Zhang
- Department of Pathology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Hua Chen
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yunjian Huang
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
16
|
Zhao J, Mommersteeg MTM. Slit-Robo signalling in heart development. Cardiovasc Res 2018; 114:794-804. [PMID: 29538649 PMCID: PMC5909645 DOI: 10.1093/cvr/cvy061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 01/16/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023] Open
Abstract
The Slit ligands and their Robo receptors are well-known for their roles during axon guidance in the central nervous system but are still relatively unknown in the cardiac field. However, data from different animal models suggest a broad involvement of the pathway in many aspects of heart development, from cardiac cell migration and alignment, lumen formation, chamber formation, to the formation of the ventricular septum, semilunar and atrioventricular valves, caval veins, and pericardium. Absence of one or more of the genes in the pathway results in defects ranging from bicuspid aortic valves to ventricular septal defects and abnormal venous connections to the heart. Congenital heart defects are the most common congenital malformations found in life new-born babies and progress in methods for large scale human genetic testing has significantly enhanced the identification of new causative genes involved in human congenital heart disease. Recently, loss of function variants in ROBO1 have also been linked to ventricular septal defects and tetralogy of Fallot in patients. Here, we will give an overview of the role of the Slit-Robo signalling pathway in Drosophila, zebrafish, and mouse heart development. The extent of these data warrant further attention on the SLIT-ROBO signalling pathway as a candidate for an array of human congenital heart defects.
Collapse
Affiliation(s)
- Juanjuan Zhao
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Mathilda T M Mommersteeg
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
17
|
Rasmussen M, Sunde L, Nielsen ML, Ramsing M, Petersen A, Hjortshøj TD, Olsen TE, Tabor A, Hertz JM, Johnsen I, Sperling L, Petersen OB, Jensen UB, Møller FG, Petersen MB, Lildballe DL. Targeted gene sequencing and whole-exome sequencing in autopsied fetuses with prenatally diagnosed kidney anomalies. Clin Genet 2018; 93:860-869. [PMID: 29194579 DOI: 10.1111/cge.13185] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 01/16/2023]
Abstract
Identification of fetal kidney anomalies invites questions about underlying causes and recurrence risk in future pregnancies. We therefore investigated the diagnostic yield of next-generation sequencing in fetuses with bilateral kidney anomalies and the correlation between disrupted genes and fetal phenotypes. Fetuses with bilateral kidney anomalies were screened using an in-house-designed kidney-gene panel. In families where candidate variants were not identified, whole-exome sequencing was performed. Genes uncovered by this analysis were added to our kidney panel. We identified likely deleterious variants in 11 of 56 (20%) families. The kidney-gene analysis revealed likely deleterious variants in known kidney developmental genes in 6 fetuses and TMEM67 variants in 2 unrelated fetuses. Kidney histology was similar in the latter 2 fetuses-presenting a distinct prenatal form of nephronophthisis. Exome sequencing identified ROBO1 variants in one family and a GREB1L variant in another family. GREB1L and ROBO1 were added to our kidney-gene panel and additional variants were identified. Next-generation sequencing substantially contributes to identifying causes of fetal kidney anomalies. Genetic causes may be supported by histological examination of the kidneys. This is the first time that SLIT-ROBO signaling is implicated in human bilateral kidney agenesis.
Collapse
Affiliation(s)
- M Rasmussen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - L Sunde
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - M L Nielsen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - M Ramsing
- Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - A Petersen
- Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - T D Hjortshøj
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - T E Olsen
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - A Tabor
- Department of Obstetrics, Center of Fetal Medicine, Rigshospitalet, Copenhagen, Denmark
| | - J M Hertz
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - I Johnsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - L Sperling
- Department of Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
| | - O B Petersen
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | - U B Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - F G Møller
- Department of Pediatrics, Herning Regional Hospital, Herning, Denmark
| | - M B Petersen
- Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - D L Lildballe
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
18
|
Tumelty KE, Higginson-Scott N, Fan X, Bajaj P, Knowlton KM, Shamashkin M, Coyle AJ, Lu W, Berasi SP. Identification of direct negative cross-talk between the SLIT2 and bone morphogenetic protein-Gremlin signaling pathways. J Biol Chem 2018; 293:3039-3055. [PMID: 29317497 DOI: 10.1074/jbc.m117.804021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/06/2017] [Indexed: 12/28/2022] Open
Abstract
Slit guidance ligand 2 (SLIT2) is a large, secreted protein that binds roundabout (ROBO) receptors on multiple cell types, including neurons and kidney podocytes. SLIT2-ROBO-mediated signaling regulates neuronal migration and ureteric bud (UB) outgrowth during kidney development as well as glomerular filtration in adult kidneys. Additionally, SLIT2 binds Gremlin, an antagonist of bone morphogenetic proteins (BMPs), and BMP-Gremlin signaling also regulates UB formation. However, direct cross-talk between the ROBO2-SLIT2 and BMP-Gremlin signaling pathways has not been established. Here, we report the discovery of negative feedback between the SLIT2 and BMP-Gremlin signaling pathways. We found that the SLIT2-Gremlin interaction inhibited both SLIT2-ROBO2 signaling in neurons and Gremlin antagonism of BMP activity in myoblasts and fibroblasts. Furthermore, BMP2 down-regulated SLIT2 expression and promoter activity through canonical BMP signaling. Gremlin treatment, BMP receptor inhibition, and SMAD family member 4 (SMAD4) knockdown rescued BMP-mediated repression of SLIT2. BMP2 treatment of nephron progenitor cells derived from human embryonic stem cells decreased SLIT2 expression, further suggesting an interaction between the BMP2-Gremlin and SLIT2 pathways in human kidney cells. In conclusion, our study has revealed direct negative cross-talk between two pathways, previously thought to be unassociated, that may regulate both kidney development and adult tissue maintenance.
Collapse
Affiliation(s)
- Kathleen E Tumelty
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Nathan Higginson-Scott
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Xueping Fan
- the Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts 02118, and
| | - Piyush Bajaj
- the Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340
| | - Kelly M Knowlton
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Michael Shamashkin
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Anthony J Coyle
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Weining Lu
- the Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts 02118, and
| | - Stephen P Berasi
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139,
| |
Collapse
|
19
|
BMP7 plays a critical role in TMEM100-inhibited cell proliferation and apoptosis in mouse metanephric mesenchymal cells in vitro. In Vitro Cell Dev Biol Anim 2017; 54:111-119. [DOI: 10.1007/s11626-017-0211-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 10/17/2017] [Indexed: 12/18/2022]
|
20
|
Li Y, Zhang XT, Wang XY, Wang G, Chuai M, Münsterberg A, Yang X. Robo signaling regulates the production of cranial neural crest cells. Exp Cell Res 2017; 361:73-84. [PMID: 28987541 DOI: 10.1016/j.yexcr.2017.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/08/2017] [Accepted: 10/03/2017] [Indexed: 11/27/2022]
Abstract
Slit/Robo signaling plays an important role in the guidance of developing neurons in developing embryos. However, it remains obscure whether and how Slit/Robo signaling is involved in the production of cranial neural crest cells. In this study, we examined Robo1 deficient mice to reveal developmental defects of mouse cranial frontal and parietal bones, which are derivatives of cranial neural crest cells. Therefore, we determined the production of HNK1+ cranial neural crest cells in early chick embryo development after knock-down (KD) of Robo1 expression. Detection of markers for pre-migratory and migratory neural crest cells, PAX7 and AP-2α, showed that production of both was affected by Robo1 KD. In addition, we found that the transcription factor slug is responsible for the aberrant delamination/EMT of cranial neural crest cells induced by Robo1 KD, which also led to elevated expression of E- and N-Cadherin. N-Cadherin expression was enhanced when blocking FGF signaling with dominant-negative FGFR1 in half of the neural tube. Taken together, we show that Slit/Robo signaling influences the delamination/EMT of cranial neural crest cells, which is required for cranial bone development.
Collapse
Affiliation(s)
- Yan Li
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China; The key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Tan Zhang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Xiao-Yu Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Guang Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee DD1 5EH, UK
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Xuesong Yang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
21
|
Wu MF, Liao CY, Wang LY, Chang JT. The role of Slit-Robo signaling in the regulation of tissue barriers. Tissue Barriers 2017; 5:e1331155. [PMID: 28598714 PMCID: PMC5501134 DOI: 10.1080/21688370.2017.1331155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 01/12/2023] Open
Abstract
The role of Slit/Robo signaling has extended from initial axon repulsion in the developing nervous system to organ morphogenesis, cancer development and angiogenesis. Slit/Robo signaling regulates similar pathways within these processes. Slit/Robo ensures the homeostasis of the dynamic interaction between cell-cell and cell-matrix interactions. The dysregulation of Slit/Robo signaling damages the tissue barrier, resulting in developmental abnormalities or disease. Here, we summarize how Slit/Robo controls kidney morphogenesis and describe the dual roles of Slit/Robo signaling in the regulation of tumorigenesis and angiogenesis.
Collapse
Affiliation(s)
- Ming-Fang Wu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C.
| | - Chen-Yi Liao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
| | - Ling-Yi Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
| | - Jinghua Tsai Chang
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C.
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
| |
Collapse
|
22
|
Li GJ, Yang Y, Yang GK, Wan J, Cui DL, Ma ZH, Du LJ, Zhang GM. Slit2 suppresses endothelial cell proliferation and migration by inhibiting the VEGF-Notch signaling pathway. Mol Med Rep 2017; 15:1981-1988. [PMID: 28260032 PMCID: PMC5364956 DOI: 10.3892/mmr.2017.6240] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 11/10/2016] [Indexed: 11/11/2022] Open
Abstract
Slit homolog 2 (Slit2) is distributed in various tissues and participates in numerous cellular processes; however, the role of Slit2 in the regulation of angiogenesis remains controversial, since it has previously been reported to exert proangiogenic and antiangiogenic activities. The present study aimed to investigate the effects of Slit2 on vascular endothelial cell proliferation and migration in vitro, and to reveal the possible underlying signaling pathway. Aortic endothelial cells were isolated from Sprague Dawley rats and cultured. Cell proliferation assay, cell migration assay, immunocytochemistry and small interfering RNA transfection were subsequently performed. The results demonstrated that exogenous Slit2 administration markedly suppressed TNF-α-induced endothelial cell proliferation and migration in vitro. In addition, TNF-α application upregulated the protein expression levels of vascular endothelial growth factor (VEGF) and Notch in RAECs, whereas Slit2 administration downregulated VEGF and Notch expression in RAECs cultured in TNF-α conditioned medium. Further studies indicated that knockdown of VEGF suppressed the effects of TNF-α on the induction of RAEC proliferation and migration. VEGF knockdown-induced inhibition of RAEC proliferation and migration in TNF-α conditioned medium was also achieved without Slit2 administration. Furthermore, VEGF knockdown markedly decreased Notch1 and Notch2 expression. These results indicated that Slit2 suppresses TNF-α-induced vascular endothelial cell proliferation and migration in vitro by inhibiting the VEGF-Notch signaling pathway. Therefore, Slit2 may inhibit the proliferation and migration of endothelial cells during vascular development.
Collapse
Affiliation(s)
- Guo-Jian Li
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Yong Yang
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Guo-Kai Yang
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Jia Wan
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Dao-Lei Cui
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Zhen-Huan Ma
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Ling-Juan Du
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Gui-Min Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
23
|
The Caenorhabditis elegans NF2/Merlin Molecule NFM-1 Nonautonomously Regulates Neuroblast Migration and Interacts Genetically with the Guidance Cue SLT-1/Slit. Genetics 2016; 205:737-748. [PMID: 27913619 DOI: 10.1534/genetics.116.191957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/23/2016] [Indexed: 11/18/2022] Open
Abstract
During nervous system development, neurons and their progenitors migrate to their final destinations. In Caenorhabditis elegans, the bilateral Q neuroblasts and their descendants migrate long distances in opposite directions, despite being born in the same posterior region. QR on the right migrates anteriorly and generates the AQR neuron positioned near the head, and QL on the left migrates posteriorly, giving rise to the PQR neuron positioned near the tail. In a screen for genes required for AQR and PQR migration, we identified an allele of nfm-1, which encodes a molecule similar to vertebrate NF2/Merlin, an important tumor suppressor in humans. Mutations in NF2 lead to neurofibromatosis type II, characterized by benign tumors of glial tissues. Here we demonstrate that in C. elegans, nfm-1 is required for the ability of Q cells and their descendants to extend protrusions and to migrate, but is not required for direction of migration. Using a combination of mosaic analysis and cell-specific expression, we show that NFM-1 is required nonautonomously, possibly in muscles, to promote Q lineage migrations. We also show a genetic interaction between nfm-1 and the C. elegans Slit homolog slt-1, which encodes a conserved secreted guidance cue. Our results suggest that NFM-1 might be involved in the generation of an extracellular cue that promotes Q neuroblast protrusion and migration that acts with or in parallel to SLT-1 In vertebrates, NF2 and Slit2 interact in axon pathfinding, suggesting a conserved interaction of NF2 and Slit2 in regulating migratory events.
Collapse
|
24
|
Abstract
Slits are secreted proteins that bind to Roundabout (Robo) receptors. Slit-Robo signaling is best known for mediating axon repulsion in the developing nervous system. However, in recent years the functional repertoire of Slits and Robo has expanded tremendously and Slit-Robo signaling has been linked to roles in neurogenesis, angiogenesis and cancer progression among other processes. Likewise, our mechanistic understanding of Slit-Robo signaling has progressed enormously. Here, we summarize new insights into Slit-Robo evolutionary and system-dependent diversity, receptor-ligand interactions, signaling crosstalk and receptor activation.
Collapse
Affiliation(s)
- Heike Blockus
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, Paris 75012, France Ecole des Neurosciences de Paris, Paris F-75005, France
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, Paris 75012, France
| |
Collapse
|
25
|
Yuan M, Guo H, Li J, Sui C, Qin Y, Wang J, Khan YH, Ye L, Xie F, Wang H, Yuan L, Ye J. Slit2 and Robo1 induce opposing effects on metastasis of hepatocellular carcinoma Sk-hep-1 cells. Int J Oncol 2016; 49:305-15. [PMID: 27176045 DOI: 10.3892/ijo.2016.3506] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/28/2016] [Indexed: 11/05/2022] Open
Abstract
The neural guidance molecular, Slit2, and its cognate receptor, Robo1, play critical roles in the development of the nervous system, nevertheless, their functions are not limited to this system. Numerous studies have shown decreased Slit2 expression in a wide variety of cancers, highlighting its potential as a tumor suppressor. However, the Slit2/Robo1 signaling axis was reported to induce either suppressive or stimulatory effects on tumor growth and metastasis, depending on cellular context. There is a paucity of information on the effects of the Slit2/Robo1 signaling axis on the growth and metastasis of human hepatocellular carcinoma (HCC). Large-scale data mining of the Oncomine database has revealed heterogeneous expression of Slit2 in HCC. We screened the Sk-hep-1, a cell line showing a relatively high level of Slit2, and low level of Robo1 expression. After Slit2 knockdown and Robo1 overexpression in these cells, we found Slit2 and Robo1 exerted opposing effects on tumor growth and metastasis both in in vitro and in vivo models. Slit2 knockdown and Robo1 overexpression in Sk-hep-1 cells promoted tumor growth and metastasis, suggesting a negative and positive role for Slit2 and Robo1, respectively, in tumor progression. Robo1 overexpression upregulated matrix metalloproteinase (MMP)2, -9 and membrane-type1 MMP (MT1-MMP) expression, stimulated MMP2, but not MMP9 activation, and downregulated expression of TIMP1 and 2. The PI3K/Akt signaling pathway is of importance in regulating MMP2 expression in Sk-hep-1 cells, since Robo1 overexpression stimulated phosphorylation of Akt while the PI3K inhibitor LY294002, significantly inhibited the upregulation of MMP2 and also the enhanced cell invasion induced by Robo1 overexpression. We postulate that Robo1 promotes tumor invasion partly by the upregulation of MMP2 after activation of PI3K/Akt signaling pathway. Notably, Slit2 knockdown caused the upregulation of Robo1 expression both at the mRNA and protein levels. Thus, the stimulatory effects of Slit2 knockdown on tumor progression can be ascribed, at least in part, to the upregulation of Robo1 and its positive role in tumor progression.
Collapse
Affiliation(s)
- Mingjing Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Hui Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jing Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Chengzhi Sui
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, P.R. China
| | - Ying Qin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jingjing Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Yasir Hayat Khan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Liying Ye
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Fuan Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Heng Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Li Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jun Ye
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| |
Collapse
|
26
|
Li XT, Yu Q, Zhou QS, Zhao X, Liu ZY, Cui WZ, Liu QX. BmRobo1a and BmRobo1b control axon repulsion in the silkworm Bombyx mori. Gene 2016; 577:215-20. [PMID: 26642898 DOI: 10.1016/j.gene.2015.11.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 11/24/2022]
Abstract
The development of the nervous system is based on the growth and connection of axons, and axon guidance molecules are the dominant regulators during this course. Robo, as the receptor of axon guidance molecule Slit, plays a key role as a conserved repellent cue for axon guidance during the development of the central nervous system. However, the function of Robo in the silkworm Bombyx mori is unknown. In this study, we cloned two novel robo genes in B. mori (Bmrobo1a and Bmrobo1b). BmRobo1a and BmRobo1b lack an Ig and a FNIII domain in the extracellular region and the CC0 and CC2 motifs in the intracellular region. BmRobo1a and BmRobo1b were colocalized with BmSlit in the neuropil. Knock-down of Bmrobo1a and Bmrobo1b by RNA interference (RNAi) resulted in abnormal development of axons. Our results suggest that BmRobo1a and BmRobo1b have repulsive function in axon guidance, even though their structures are different from Robo1 of other species.
Collapse
Affiliation(s)
- Xiao-Tong Li
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China
| | - Qi Yu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China
| | - Qi-Sheng Zhou
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China
| | - Xiao Zhao
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China
| | - Zhao-Yang Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China
| | - Wei-Zheng Cui
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China.
| | - Qing-Xin Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, China.
| |
Collapse
|
27
|
Qin N, Fan XC, Zhang YY, Xu XX, Tyasi TL, Jing Y, Mu F, Wei ML, Xu RF. New insights into implication of the SLIT/ROBO pathway in the prehierarchical follicle development of hen ovary. Poult Sci 2015; 94:2235-46. [PMID: 26188027 DOI: 10.3382/ps/pev185] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 05/19/2015] [Indexed: 01/28/2023] Open
Abstract
The SLIT/Roundabout (ROBO) pathway is involved in follicle development of mammalian ovary, and 2 secreted hormones activin A and inhibin A have potential roles in modulation of the SLIT/ROBO system, but the related actions remain poorly understood in bird. The aims of the present study were to examine the spatial and temporal expression of the SLIT ligand genes (SLIT1, SLIT2, and SLIT3) and their receptor ROBO1, ROBO2, ROBO3, and ROBO4 genes in various-sized prehierarchical follicles during hen ovary development and the effects of activin A and inhibin A on the expression of these genes in the cultured hen follicles. Our result demonstrated that the transcripts of the 3 SLIT genes were highly expressed in the developing follicles and expression patterns of the SLIT transcripts were different from those of ROBO genes detected by real-time quantitative reverse transcriptase PCR. Both SLIT and ROBO transcripts were predominantly expressed in oocytes and granulosa cells from the prehierarchichal follicles examined by in situ hybridization. The localization for SLIT and ROBO proteins was revealed by immunohistochemistry similar to the spatial distribution of their transcript. In cultured follicles (4 to 8 mm in diameter), the expression levels of SLIT and ROBO members are hormonally regulated by activin A (10 ng/mL) and/or inhibin A (20 ng/mL) after treatment for 24 h. However, the expression of only SLIT2, SLIT3, and ROBO3 mRNA presented a directly opposite response to activin A and inhibin A hormones. These results indicate that SLIT/ROBO pathway is implicated in the prehierarchical follicular development of the hen ovary by an intrafollicular autocrine and/or paracrine action, and is influenced by activin A and inhibin A hormones.
Collapse
Affiliation(s)
- N Qin
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - X C Fan
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Y Y Zhang
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - X X Xu
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - T L Tyasi
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Y Jing
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - F Mu
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - M L Wei
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - R F Xu
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| |
Collapse
|
28
|
Hwang DY, Kohl S, Fan X, Vivante A, Chan S, Dworschak GC, Schulz J, van Eerde AM, Hilger AC, Gee HY, Pennimpede T, Herrmann BG, van de Hoek G, Renkema KY, Schell C, Huber TB, Reutter HM, Soliman NA, Stajic N, Bogdanovic R, Kehinde EO, Lifton RP, Tasic V, Lu W, Hildebrandt F. Mutations of the SLIT2-ROBO2 pathway genes SLIT2 and SRGAP1 confer risk for congenital anomalies of the kidney and urinary tract. Hum Genet 2015; 134:905-16. [PMID: 26026792 PMCID: PMC4497857 DOI: 10.1007/s00439-015-1570-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/18/2015] [Indexed: 12/26/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) account for 40-50% of chronic kidney disease that manifests in the first two decades of life. Thus far, 31 monogenic causes of isolated CAKUT have been described, explaining ~12% of cases. To identify additional CAKUT-causing genes, we performed whole-exome sequencing followed by a genetic burden analysis in 26 genetically unsolved families with CAKUT. We identified two heterozygous mutations in SRGAP1 in 2 unrelated families. SRGAP1 is a small GTPase-activating protein in the SLIT2-ROBO2 signaling pathway, which is essential for development of the metanephric kidney. We then examined the pathway-derived candidate gene SLIT2 for mutations in cohort of 749 individuals with CAKUT and we identified 3 unrelated individuals with heterozygous mutations. The clinical phenotypes of individuals with mutations in SLIT2 or SRGAP1 were cystic dysplastic kidneys, unilateral renal agenesis, and duplicated collecting system. We show that SRGAP1 is expressed in early mouse nephrogenic mesenchyme and that it is coexpressed with ROBO2 in SIX2-positive nephron progenitor cells of the cap mesenchyme in developing rat kidney. We demonstrate that the newly identified mutations in SRGAP1 lead to an augmented inhibition of RAC1 in cultured human embryonic kidney cells and that the SLIT2 mutations compromise the ability of the SLIT2 ligand to inhibit cell migration. Thus, we report on two novel candidate genes for causing monogenic isolated CAKUT in humans.
Collapse
Affiliation(s)
- Daw-Yang Hwang
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Stefan Kohl
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xueping Fan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Asaf Vivante
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefanie Chan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Gabriel C Dworschak
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Julian Schulz
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Albertien M van Eerde
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alina C Hilger
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Heon Yung Gee
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tracie Pennimpede
- Max Planck Institute for Molecular Genetics, Developmental Genetics Department, Berlin, Germany
| | - Bernhard G Herrmann
- Max Planck Institute for Molecular Genetics, Developmental Genetics Department, Berlin, Germany
| | - Glenn van de Hoek
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kirsten Y Renkema
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christoph Schell
- Renal Division, University Hospital Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Tobias B Huber
- Renal Division, University Hospital Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Germany
| | - Heiko M Reutter
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Neonatology, Children’s Hospital, University of Bonn, Bonn, Germany
| | - Neveen A Soliman
- Department of Pediatrics, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
- Egyptian Group for Orphan Renal Diseases (EGORD), Cairo, Egypt
| | - Natasa Stajic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Institute of Mother and Child Healthcare of Serbia, Belgrade, Serbia
| | - Radovan Bogdanovic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Institute of Mother and Child Healthcare of Serbia, Belgrade, Serbia
| | | | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Mendelian Genomics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Velibor Tasic
- Department of Pediatric Nephrology, University Children’s Hospital, Skopje, Macedonia
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
29
|
Wainwright EN, Wilhelm D, Combes AN, Little MH, Koopman P. ROBO2 restricts the nephrogenic field and regulates Wolffian duct-nephrogenic cord separation. Dev Biol 2015; 404:88-102. [PMID: 26116176 DOI: 10.1016/j.ydbio.2015.05.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/28/2015] [Accepted: 05/30/2015] [Indexed: 01/03/2023]
Abstract
ROBO2 plays a key role in regulating ureteric bud (UB) formation in the embryo, with mutations in humans and mice leading to supernumerary kidneys. Previous studies have established that the number and position of UB outgrowths is determined by the domain of metanephric mesenchymal Gdnf expression, which is expanded anteriorly in Robo2 mouse mutants. To clarify how this phenotype arises, we used high-resolution 3D imaging to reveal an increase in the number of nephrogenic cord cells, leading to extension of the metanephric mesenchyme field in Robo2-null mouse embryos. Ex vivo experiments suggested a dependence of this effect on proliferative signals from the Wolffian duct. Loss of Robo2 resulted in a failure of the normal separation of the mesenchyme from the Wolffian duct/ureteric epithelium, suggesting that aberrant juxtaposition of these two compartments in Robo2-null mice exposes the mesenchyme to abnormally high levels of proliferative stimuli. Our data suggest a new model in which SLIT-ROBO signalling acts not by attenuating Gdnf expression or activity, but instead by limiting epithelial/mesenchymal interactions in the nascent metanephros and restricting the extent of the nephrogenic field. These insights illuminate the aetiology of multiplex kidney formation in human individuals with ROBO2 mutations.
Collapse
Affiliation(s)
- Elanor N Wainwright
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dagmar Wilhelm
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alexander N Combes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Melissa H Little
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
30
|
Li J, Ye Y, Zhang R, Zhang L, Hu X, Han D, Chen J, He X, Wang G, Yang X, Wang L. Robo1/2 regulate follicle atresia through manipulating granulosa cell apoptosis in mice. Sci Rep 2015; 5:9720. [PMID: 25988316 PMCID: PMC4437031 DOI: 10.1038/srep09720] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/17/2015] [Indexed: 12/12/2022] Open
Abstract
Secreted Slit proteins and their Roundabout (Robo) receptors act as a repulsive cue to prevent axons from migrating to inappropriate locations during the development of the nervous system. Slit/Robo has also been implicated in reproductive system development, but the molecular mechanism of the Slit/Robo pathway in the reproductive system remains poorly understood. Using a transgenic mouse model, we investigated the function of the Slit/Robo pathway on ovarian follicle development and atresia. We first demonstrated that more offspring were born to mice with a partial knockout of the Robo1/2 genes in mice. We next showed that Robo1 and Robo2 are strongly expressed in ovarian granulosa cells. Apoptosis in granulosa cells was reduced when Robo1/2 were partially knocked out, and this observation was further verified by in vitro Robo1/2 knockout experiments in mouse and human granulosa cells. We also found that ovarian angiogenesis was enhanced by a partial lack of Robo1/2 genes. In summary, our data suggest that the Slit/Robo pathway can impact follicle development and atresia by influencing granulosa cell apoptosis.
Collapse
Affiliation(s)
- Jiangchao Li
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuxiang Ye
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Renli Zhang
- Reproductive Medicine Center, Guangdong General Hospital, Guangzhou 515006, China
| | - Lili Zhang
- Reproductive Medicine Center, Guangdong General Hospital, Guangzhou 515006, China
- Southern Medical University, Guangzhou 510515, China
| | - Xiwen Hu
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dong Han
- Reproductive Medicine Center, Guangdong General Hospital, Guangzhou 515006, China
| | - Jiayuan Chen
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaodong He
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Guang Wang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Lijing Wang
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
31
|
Slit2 promotes tumor growth and invasion in chemically induced skin carcinogenesis. J Transl Med 2014; 94:766-76. [PMID: 24840330 DOI: 10.1038/labinvest.2014.70] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 11/09/2022] Open
Abstract
Slit, a neuronal guidance cue, binds to Roundabout (Robo) receptors to modulate neuronal, leukocytic, and endothelial migration. Slit has been reported to have an important effect on tumor growth and metastasis. In the current study, we evaluated the role of Slit2 in skin tumor growth and invasion in mice using a two-step chemical carcinogenesis protocol. We found that Slit2 expression correlated with the loss of basement membrane in the samples of human skin squamous cell carcinoma at different stages of disease progression. Slit2-Tg mice developed significantly more skin tumors than wild-type mice. Furthermore, the skin tumors that occurred in Slit2-Tg mice were significantly larger than those in the wild-type mice 10 weeks after 7,12-dimethylbenz[a]anthracene initiation until the end of the experiment. We also found that pathological development of the wild-type mice was delayed compared with that of Slit2-Tg mice. To further investigate the mechanism of increasing tumors in Slit2-Tg mice, we analyzed the expression of 5-bromo-2'-deoxyuridine (BrdU) in mouse skin lesions and found that the number of BrdU-positive cells and microvessel density in skin lesions were significantly higher in Slit2-Tg mice than in wild-type mice. Histological staining of PAS and type IV collagen and the colocalization of Slit2 and type IV collagen demonstrated varying degrees of loss of the basement membrane in the skin lesions from Slit2-Tg mice that were at the stage of carcinoma in situ. However, the basement membrane was well defined in the wild-type mice. In addition, MMP2, but not MMP9, was upregulated in the skin tissue of Slit2-Tg mice. Interruption of Slit2-Robo1 signaling by the antibody R5 significantly repressed the invasive capability of the squamous cell carcinoma cell line A431. Taken together, our findings reveal that Slit2 promotes DMBA/TPA-induced skin tumorigenesis by increasing cell proliferation, microvessel density, and invasive behavior of cutaneous squamous cell carcinoma, along with loss of basement membrane, by upregulation of MMP2 expression.
Collapse
|
32
|
Fabbro A, Sucapane A, Toma FM, Calura E, Rizzetto L, Carrieri C, Roncaglia P, Martinelli V, Scaini D, Masten L, Turco A, Gustincich S, Prato M, Ballerini L. Adhesion to carbon nanotube conductive scaffolds forces action-potential appearance in immature rat spinal neurons. PLoS One 2013; 8:e73621. [PMID: 23951361 PMCID: PMC3741175 DOI: 10.1371/journal.pone.0073621] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 07/29/2013] [Indexed: 12/19/2022] Open
Abstract
In the last decade, carbon nanotube growth substrates have been used to investigate neurons and neuronal networks formation in vitro when guided by artificial nano-scaled cues. Besides, nanotube-based interfaces are being developed, such as prosthesis for monitoring brain activity. We recently described how carbon nanotube substrates alter the electrophysiological and synaptic responses of hippocampal neurons in culture. This observation highlighted the exceptional ability of this material in interfering with nerve tissue growth. Here we test the hypothesis that carbon nanotube scaffolds promote the development of immature neurons isolated from the neonatal rat spinal cord, and maintained in vitro. To address this issue we performed electrophysiological studies associated to gene expression analysis. Our results indicate that spinal neurons plated on electro-conductive carbon nanotubes show a facilitated development. Spinal neurons anticipate the expression of functional markers of maturation, such as the generation of voltage dependent currents or action potentials. These changes are accompanied by a selective modulation of gene expression, involving neuronal and non-neuronal components. Our microarray experiments suggest that carbon nanotube platforms trigger reparative activities involving microglia, in the absence of reactive gliosis. Hence, future tissue scaffolds blended with conductive nanotubes may be exploited to promote cell differentiation and reparative pathways in neural regeneration strategies.
Collapse
Affiliation(s)
| | | | - Francesca Maria Toma
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Enrica Calura
- Department of Biology, University of Padua, Padova, Italy
| | - Lisa Rizzetto
- Department of Neuroscience, Psychology, Drug Research and Child's Health, University of Florence, Florence, Italy
- Innovation and Research Center, Fondazione Edmund Mach, San Michele all’Adige, Trento, Italy
| | - Claudia Carrieri
- European Molecular Biology Laboratory, Mouse Biology Unit, Monterotondo (Rome), Italy
| | - Paola Roncaglia
- International School for Advanced Studies (SISSA), Trieste, Italy
- European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Valentina Martinelli
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Denis Scaini
- Life Science Department, University of Trieste, Trieste, Italy
- SENIL, ELETTRA Synchrotron Light Source, Trieste, Italy
| | - Lara Masten
- Life Science Department, University of Trieste, Trieste, Italy
| | - Antonio Turco
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | | | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Laura Ballerini
- Life Science Department, University of Trieste, Trieste, Italy
| |
Collapse
|
33
|
Beck TF, Shchelochkov OA, Yu Z, Kim BJ, Hernández-García A, Zaveri HP, Bishop C, Overbeek PA, Stockton DW, Justice MJ, Scott DA. Novel frem1-related mouse phenotypes and evidence of genetic interactions with gata4 and slit3. PLoS One 2013; 8:e58830. [PMID: 23536828 PMCID: PMC3594180 DOI: 10.1371/journal.pone.0058830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/07/2013] [Indexed: 11/27/2022] Open
Abstract
The FRAS1-related extracellular matrix 1 (FREM1) gene encodes an extracellular matrix protein that plays a critical role in the development of multiple organ systems. In humans, recessive mutations in FREM1 cause eye defects, congenital diaphragmatic hernia, renal anomalies and anorectal malformations including anteriorly placed anus. A similar constellation of findings-microphthalmia, cryptophthalmos, congenital diaphragmatic hernia, renal agenesis and rectal prolapse-have been described in FREM1-deficient mice. In this paper, we identify a homozygous Frem1 missense mutation (c.1687A>T, p.Ile563Phe) in an N-ethyl-N-nitrosourea (ENU)-derived mouse strain, crf11, with microphthalmia, cryptophthalmos, renal agenesis and rectal prolapse. This mutation affects a highly conserved residue in FREM1's third CSPG domain. The p.Ile563Phe change is predicted to be deleterious and to cause decreased FREM1 protein stability. The crf11 allele also fails to complement the previously described eyes2 allele of Frem1 (p.Lys826*) providing further evidence that the crf11 phenotype is due to changes affecting Frem1 function. We then use mice bearing the crf11 and eyes2 alleles to identify lung lobulation defects and decreased anogenital distance in males as novel phenotypes associated with FREM1 deficiency in mice. Due to phenotypic overlaps between FREM1-deficient mice and mice that are deficient for the retinoic acid-responsive transcription factor GATA4 and the extracellular matrix protein SLIT3, we also perform experiments to look for in vivo genetic interactions between the genes that encode these proteins. These experiments reveal that Frem1 interacts genetically with Gata4 in the development of lung lobulation defects and with Slit3 in the development of renal agenesis. These results demonstrate that FREM1-deficient mice faithfully recapitulate many of the phenotypes seen in individuals with FREM1 deficiency and that variations in GATA4 and SLIT3 expression modulate some FREM1-related phenotypes in mice.
Collapse
Affiliation(s)
- Tyler F. Beck
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Oleg A. Shchelochkov
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Zhiyin Yu
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bum Jun Kim
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Andrés Hernández-García
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hitisha P. Zaveri
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Colin Bishop
- The Wake Forest Institute for Regenerative Medicine, Winston Salem, North Carolina, United States of America
| | - Paul A. Overbeek
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David W. Stockton
- Departments of Pediatrics and Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Monica J. Justice
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
34
|
Podocytes: A new player for glutamate signaling. Int J Biochem Cell Biol 2012; 44:2272-7. [DOI: 10.1016/j.biocel.2012.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 07/20/2012] [Accepted: 09/16/2012] [Indexed: 11/24/2022]
|
35
|
Fan X, Li Q, Pisarek-Horowitz A, Rasouly HM, Wang X, Bonegio RG, Wang H, McLaughlin M, Mangos S, Kalluri R, Holzman LB, Drummond IA, Brown D, Salant DJ, Lu W. Inhibitory effects of Robo2 on nephrin: a crosstalk between positive and negative signals regulating podocyte structure. Cell Rep 2012; 2:52-61. [PMID: 22840396 DOI: 10.1016/j.celrep.2012.06.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/05/2012] [Accepted: 06/05/2012] [Indexed: 11/20/2022] Open
Abstract
Robo2 is the cell surface receptor for the repulsive guidance cue Slit and is involved in axon guidance and neuronal migration. Nephrin is a podocyte slit-diaphragm protein that functions in the kidney glomerular filtration barrier. Here, we report that Robo2 is expressed at the basal surface of mouse podocytes and colocalizes with nephrin. Biochemical studies indicate that Robo2 forms a complex with nephrin in the kidney through adaptor protein Nck. In contrast to the role of nephrin that promotes actin polymerization, Slit2-Robo2 signaling inhibits nephrin-induced actin polymerization. In addition, the amount of F-actin associated with nephrin is increased in Robo2 knockout mice that develop an altered podocyte foot process structure. Genetic interaction study further reveals that loss of Robo2 alleviates the abnormal podocyte structural phenotype in nephrin null mice. These results suggest that Robo2 signaling acts as a negative regulator on nephrin to influence podocyte foot process architecture.
Collapse
Affiliation(s)
- Xueping Fan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Liao WX, Laurent LC, Agent S, Hodges J, Chen DB. Human placental expression of SLIT/ROBO signaling cues: effects of preeclampsia and hypoxia. Biol Reprod 2012; 86:111. [PMID: 22262697 DOI: 10.1095/biolreprod.110.088138] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Preeclampsia is characterized by dysfunctional endothelium and impaired angiogenesis. Recent studies suggest that the neuronal guidance SLIT/ROBO system regulates tumor angiogenesis. This study investigated if SLIT and ROBO are differentially expressed in healthy term and preeclamptic placentas and if hypoxia regulates SLIT and ROBO expression in placental trophoblast and endothelial cells. Total RNA and protein were extracted from placental tissues of healthy term (n = 5) and preeclamptic (n = 6) pregnancies and used for SLIT/ROBO expression analyses with reverse transcription-polymerase chain reaction (RT-PCR), real-time quantitative-PCR, and immunoblotting. Paraffin-embedded tissues were processed to localize SLIT/ROBO proteins in placental villi by immunohistochemistry. BeWo choriocarcinoma cells and human umbilical vein endothelial cells (HUVEC) were treated with 2% or 10% oxygen or the hypoxia mimetic deferoxamine mesylate (100 μM) to test if hypoxia regulates SLIT/ROBO expression. SLIT2, SLIT3, ROBO1, and ROBO4 mRNA and proteins were detected in the placenta. SLIT2 and ROBO1 proteins localized in the syncytiotrophoblast, and SLIT3, ROBO1, and ROBO4 in capillary endothelium of the placental villi. Levels of ROBO1 and ROBO4 as well as sFLT1 (soluble fms-like tyrosine kinase-1) proteins were significantly greater in preeclamptic placentas compared to normal controls. Hypoxia significantly increased both mRNA and protein levels of SLIT2 in BeWo cells and of SLIT3, ROBO1, and ROBB4 in HUVEC. Thus, trophoblast and endothelial coexpression of SLIT/ROBO suggests an autocrine/paracrine regulatory system for regulating placental function. Differential expression of SLITs and ROBOs in healthy term and preeclamptic placentas and hypoxia regulation of their expressions in placental cells implicate a potential pathophysiological role for this system in preeclampsia.
Collapse
Affiliation(s)
- Wu-Xiang Liao
- Department of Obstetrics & Gynecology, University of California Irvine, Irvine, California 92697, USA
| | | | | | | | | |
Collapse
|
37
|
Abstract
The Slit family of secreted proteins and their transmembrane receptor, Robo, were originally identified in the nervous system where they function as axon guidance cues and branching factors during development. Since their discovery, a great number of additional roles have been attributed to Slit/Robo signaling, including regulating the critical processes of cell proliferation and cell motility in a variety of cell and tissue types. These processes are often deregulated during cancer progression, allowing tumor cells to bypass safeguarding mechanisms in the cell and the environment in order to grow and escape to new tissues. In the past decade, it has been shown that the expression of Slit and Robo is altered in a wide variety of cancer types, identifying them as potential therapeutic targets. Further, studies have demonstrated dual roles for Slits and Robos in cancer, acting as both oncogenes and tumor suppressors. This bifunctionality is also observed in their roles as axon guidance cues in the developing nervous system, where they both attract and repel neuronal migration. The fact that this signaling axis can have opposite functions depending on the cellular circumstance make its actions challenging to define. Here, we summarize our current understanding of the dual roles that Slit/Robo signaling play in development, epithelial tumor progression, and tumor angiogenesis.
Collapse
Affiliation(s)
- Mimmi S. Ballard
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz CA 95064
| | - Lindsay Hinck
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz CA 95064
| |
Collapse
|
38
|
Duncan WC, McDonald SE, Dickinson RE, Shaw JLV, Lourenco PC, Wheelhouse N, Lee KF, Critchley HOD, Horne AW. Expression of the repulsive SLIT/ROBO pathway in the human endometrium and Fallopian tube. Mol Hum Reprod 2010; 16:950-9. [PMID: 20651036 PMCID: PMC2992050 DOI: 10.1093/molehr/gaq055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We investigated whether the repulsive SLIT/ROBO pathway is expressed in the endometrium and is negatively regulated during implantation. We also examined whether deficient expression in the Fallopian tube (FT) may predispose to ectopic pregnancy (EP). Endometrium (n = 21) and FT (n = 17) were collected across the menstrual cycle from fertile women with regular cycles. Decidualized endometrium (n = 6) was obtained from women undergoing termination, and FT (n = 6) was obtained from women with EP. SLIT/ROBO expression was quantified by reverse transcription-PCR and protein localized by immunohistochemistry. The regulation of SLIT/ROBO expression in vitro, by sex steroids and hCG, was assessed in endometrial (hTERT-EEpC) epithelial cells, and the effects of Chlamydia trachomatis infection and smoking were studied in oviductal (OE-E6/E7) epithelial cells. Endometrial SLIT3 was highest in the mid-secretory phase (P = 0.0003) and SLIT1,2 and ROBO1 showed a similar trend. ROBO2 was highest in proliferative phase (P = 0.027) and ROBO3,4 showed a similar trend. SLIT2,3 and ROBO1, 4 were lower in decidua compared with mid-secretory endometrium (P < 0.05). SLITs and ROBOs, excepting ROBO2, were expressed in FT but there were no differences across the cycle or in EP. SLIT/ROBO proteins were localized to endometrial and FT epithelium. Treatment of hTERT-EEpC with a combination of estradiol and medroxyprogesterone acetate inhibited ROBO1 expression (P < 0.01) but hCG had no effect. Acute treatment of OE-E6/E7 with smoking metabolite, cotinine, and C. trachomatis had no effect. These findings imply a regulated role for the endometrial SLIT/ROBO interaction during normal development and pregnancy but that it may not be important in the aetiology of EP.
Collapse
Affiliation(s)
- W C Duncan
- Centre for Reproductive Biology, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SB, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lindenmeyer MT, Eichinger F, Sen K, Anders HJ, Edenhofer I, Mattinzoli D, Kretzler M, Rastaldi MP, Cohen CD. Systematic analysis of a novel human renal glomerulus-enriched gene expression dataset. PLoS One 2010; 5:e11545. [PMID: 20634963 PMCID: PMC2902524 DOI: 10.1371/journal.pone.0011545] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 06/16/2010] [Indexed: 02/04/2023] Open
Abstract
Glomerular diseases account for the majority of cases with chronic renal failure. Several genes have been identified with key relevance for glomerular function. Quite a few of these genes show a specific or preferential mRNA expression in the renal glomerulus. To identify additional candidate genes involved in glomerular function in humans we generated a human renal glomerulus-enriched gene expression dataset (REGGED) by comparing gene expression profiles from human glomeruli and tubulointerstitium obtained from six transplant living donors using Affymetrix HG-U133A arrays. This analysis resulted in 677 genes with prominent overrepresentation in the glomerulus. Genes with 'a priori' known prominent glomerular expression served for validation and were all found in the novel dataset (e.g. CDKN1, DAG1, DDN, EHD3, MYH9, NES, NPHS1, NPHS2, PDPN, PLA2R1, PLCE1, PODXL, PTPRO, SYNPO, TCF21, TJP1, WT1). The mRNA expression of several novel glomerulus-enriched genes in REGGED was validated by qRT-PCR. Gene ontology and pathway analysis identified biological processes previously not reported to be of relevance in glomeruli of healthy human adult kidneys including among others axon guidance. This finding was further validated by assessing the expression of the axon guidance molecules neuritin (NRN1) and roundabout receptor ROBO1 and -2. In diabetic nephropathy, a prevalent glomerulopathy, differential regulation of glomerular ROBO2 mRNA was found.In summary, novel transcripts with predominant expression in the human glomerulus could be identified using a comparative strategy on microdissected nephrons. A systematic analysis of this glomerulus-specific gene expression dataset allows the detection of target molecules and biological processes involved in glomerular biology and renal disease.
Collapse
Affiliation(s)
- Maja T. Lindenmeyer
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
- Institute of Physiology with Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Felix Eichinger
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kontheari Sen
- Institute of Physiology with Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | | | - Ilka Edenhofer
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deborah Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy
| | - Matthias Kretzler
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Maria P. Rastaldi
- Renal Research Laboratory, Fondazione IRCCS Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy
| | - Clemens D. Cohen
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
- Institute of Physiology with Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Han X, Zhang MC. Potential anti-angiogenic role of Slit2 in corneal neovascularization. Exp Eye Res 2010; 90:742-9. [PMID: 20298689 DOI: 10.1016/j.exer.2010.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 02/08/2010] [Accepted: 03/11/2010] [Indexed: 11/23/2022]
Abstract
Slits are large secreted proteins critical for axon guidance and neuronal precursor cell migration in nervous system. Evidence suggests that classical neuronal guidance cues also regulate vascular development. Our objective was to investigate whether neuronal guidance cue Slit2 and Roundabout (Robo) receptors are involved in corneal neovascularization (NV). Corneal NV model in rats was induced by implantation of agarose-coated gelfoam pellets containing basic fibroblast growth factor (bFGF) into corneal stroma. Differential expression of Slit2 and Robo1-4 between normal and neovascularized cornea was detected by real-time RT-PCR and visualized by immunohistochemistry and in situ hybridization. Primary human umbilical vein endothelial cells (HUVECs) were harvested and their expression of Robo1-4 was detected by RT-PCR. Recombinant human Slit2 protein was prepared and the effect of it on the migration of vascular endothelial cells was examined using cell migration assay. Agarose-coated gelfoam pellets were able to induce well-localized and reproducible corneal NV model. A significant down-regulation of Slit2 and a strong up-regulation of Robo1 and Robo4 were seen in neovascularized cornea when compared with normal cornea (P < 0.05). Slit2, Robo1 and Robo4 were throughout the epithelium in normal cornea and markedly weak or absent in epithelium in neovascularized cornea, with Robo1 and Robo4 being prominent in vascular endothelial cells invading the stroma. Primary HUVECs were confirmed to express both Robo1 and Robo4 receptors and their migration was inhibited by Slit2 (P < 0.05). This is the first study to assess the association between Slit2 and corneal NV. Our findings suggest that the interaction of Slit2 with Robo1 and Robo4 receptors plays an essential role in inhibiting pathological neovascular processes of the cornea and may represent a new therapeutic target for corneal NV.
Collapse
Affiliation(s)
- Xi Han
- Department of Ophthalmology, Wuhan Union Hospital, Tongji Medical College of Huazhong University of Science & Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | | |
Collapse
|
41
|
Dickinson RE, Hryhorskyj L, Tremewan H, Hogg K, Thomson AA, McNeilly AS, Duncan WC. Involvement of the SLIT/ROBO pathway in follicle development in the fetal ovary. Reproduction 2010; 139:395-407. [PMID: 19900988 PMCID: PMC2971460 DOI: 10.1530/rep-09-0182] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In humans and domestic mammals, pivotal processes in ovary development, including primordial follicle assembly, occur prenatally. These events are essential for determining fertility in adult life; however, they remain poorly understood at the mechanistic level. In mammals, the SLITs (SLIT1, SLIT2 and SLIT3) and their ROBO (ROBO1, ROBO2, ROBO3/RIG-1 and ROBO4/MAGIC ROBO) receptors regulate neural, leukocyte, vascular smooth muscle cell and endothelial cell migration. In addition, the SLIT/ROBO pathway has functional roles in embryonic development and in the adult ovary by inhibiting cell migration and promoting apoptosis. We therefore characterised follicle formation and investigated the expression and localisation of the ROBO/SLIT pathway in the ovine fetal ovary. Using RT-PCR, we identified SLIT2, SLIT3, ROBO1, ROBO2 and ROBO4 in sheep ovaries harvested across gestation. The real-time quantitative PCR results implied that ROBO2 expression and ROBO4 expression were elevated during the early stages of follicle formation and stayed abundant during primordial follicle maturation (P<0.05). Immunohistochemistry examination demonstrated that ROBO1 was localised to the pre-granulosa cells, while ROBO2, ROBO4 and SLIT2 were expressed in the oocytes of the developing primordial follicle. This indicates that in the fetal ovary, SLIT-ROBO signalling may require an autocrine and paracrine interaction. Furthermore, at the time of increased SLIT-ROBO expression, there was a significant reduction in the number of proliferating oocytes in the developing ovary (P<0.0001). Overall, these results suggest, for the first time, that the SLIT-ROBO pathway is expressed at the time of follicle formation during fetal ovary development.
Collapse
Affiliation(s)
- Rachel E Dickinson
- MRC Human Reproductive Sciences Unit Division of Reproductive and Developmental Sciences, The Queen's Medical Research Institute, Centre for Reproductive Biology, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK.
| | | | | | | | | | | | | |
Collapse
|
42
|
Multiple non-cell-autonomous defects underlie neocortical callosal dysgenesis in Nfib-deficient mice. Neural Dev 2009; 4:43. [PMID: 19961580 PMCID: PMC2802587 DOI: 10.1186/1749-8104-4-43] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 12/04/2009] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Agenesis of the corpus callosum is associated with many human developmental syndromes. Key mechanisms regulating callosal formation include the guidance of axons arising from pioneering neurons in the cingulate cortex and the development of cortical midline glial populations, but their molecular regulation remains poorly characterised. Recent data have shown that mice lacking the transcription factor Nfib exhibit callosal agenesis, yet neocortical callosal neurons express only low levels of Nfib. Therefore, we investigate here how Nfib functions to regulate non-cell-autonomous mechanisms of callosal formation. RESULTS Our investigations confirmed a reduction in glial cells at the midline in Nfib-/- mice. To determine how this occurs, we examined radial progenitors at the cortical midline and found that they were specified correctly in Nfib mutant mice, but did not differentiate into mature glia. Cellular proliferation and apoptosis occurred normally at the midline of Nfib mutant mice, indicating that the decrease in midline glia observed was due to deficits in differentiation rather than proliferation or apoptosis. Next we investigated the development of callosal pioneering axons in Nfib-/- mice. Using retrograde tracer labelling, we found that Nfib is expressed in cingulate neurons and hence may regulate their development. In Nfib-/- mice, neuropilin 1-positive axons fail to cross the midline and expression of neuropilin 1 is diminished. Tract tracing and immunohistochemistry further revealed that, in late gestation, a minor population of neocortical axons does cross the midline in Nfib mutants on a C57Bl/6J background, forming a rudimentary corpus callosum. Finally, the development of other forebrain commissures in Nfib-deficient mice is also aberrant. CONCLUSION The formation of the corpus callosum is severely delayed in the absence of Nfib, despite Nfib not being highly expressed in neocortical callosal neurons. Our results indicate that in addition to regulating the development of midline glial populations, Nfib also regulates the expression of neuropilin 1 within the cingulate cortex. Collectively, these data indicate that defects in midline glia and cingulate cortex neurons are associated with the callosal dysgenesis seen in Nfib-deficient mice, and provide insight into how the development of these cellular populations is controlled at a molecular level.
Collapse
|
43
|
An integrated genome screen identifies the Wnt signaling pathway as a major target of WT1. Proc Natl Acad Sci U S A 2009; 106:11154-9. [PMID: 19549856 DOI: 10.1073/pnas.0901591106] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
WT1, a critical regulator of kidney development, is a tumor suppressor for nephroblastoma but in some contexts functions as an oncogene. A limited number of direct transcriptional targets of WT1 have been identified to explain its complex roles in tumorigenesis and organogenesis. In this study we performed genome-wide screening for direct WT1 targets, using a combination of ChIP-ChIP and expression arrays. Promoter regions bound by WT1 were highly G-rich and resembled the sites for a number of other widely expressed transcription factors such as SP1, MAZ, and ZNF219. Genes directly regulated by WT1 were implicated in MAPK signaling, axon guidance, and Wnt pathways. Among directly bound and regulated genes by WT1, nine were identified in the Wnt signaling pathway, suggesting that WT1 modulates a subset of Wnt components and responsive genes by direct binding. To prove the biological importance of the interplay between WT1 and Wnt signaling, we showed that WT1 blocked the ability of Wnt8 to induce a secondary body axis during Xenopus embryonic development. WT1 inhibited TCF-mediated transcription activated by Wnt ligand, wild type and mutant, stabilized beta-catenin by preventing TCF4 loading onto a promoter. This was neither due to direct binding of WT1 to the TCF binding site nor to interaction between WT1 and TCF4, but by competition of WT1 and TCF4 for CBP. WT1 interference with Wnt signaling represents an important mode of its action relevant to the suppression of tumor growth and guidance of development.
Collapse
|
44
|
Piper M, Plachez C, Zalucki O, Fothergill T, Goudreau G, Erzurumlu R, Gu C, Richards LJ. Neuropilin 1-Sema signaling regulates crossing of cingulate pioneering axons during development of the corpus callosum. ACTA ACUST UNITED AC 2009; 19 Suppl 1:i11-21. [PMID: 19357391 DOI: 10.1093/cercor/bhp027] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pioneer axons from the cingulate cortex initiate corpus callosum (CC) development, yet nothing is known about the molecules that regulate their guidance. We demonstrate that neuropilin 1 (Npn1) plays an integral role in the development of the CC. Npn1 is localized to axons of cingulate neurons as they cross the midline, and multiple class 3 semaphorins (Semas) are expressed around the developing CC, implicating these guidance molecules in the regulation of Npn1-expressing axons emanating from the cingulate cortex. Furthermore, axons from the cingulate cortex display guidance errors in Npn1(Sema-) mice, a knockin mouse line in which Npn1 is unable to bind Semas. Analysis of mice deficient in the transcription factor Emx2 demonstrated that the cingulate cortex of these mice was significantly reduced in comparison to wild-type controls at E17 and that the CC was absent in rostral sections. Expression of Npn1 was absent in rostral sections of Emx2 mutants, suggesting that Npn1-expressing cingulate pioneers are required for CC formation. These data highlight a central role for Npn1 in the development of projections from the cingulate cortex and further illustrate the importance of these pioneer axons in the formation of the CC.
Collapse
Affiliation(s)
- Michael Piper
- Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Anitha A, Nakamura K, Yamada K, Suda S, Thanseem I, Tsujii M, Iwayama Y, Hattori E, Toyota T, Miyachi T, Iwata Y, Suzuki K, Matsuzaki H, Kawai M, Sekine Y, Tsuchiya K, Sugihara GI, Ouchi Y, Sugiyama T, Koizumi K, Higashida H, Takei N, Yoshikawa T, Mori N. Genetic analyses of roundabout (ROBO) axon guidance receptors in autism. Am J Med Genet B Neuropsychiatr Genet 2008; 147B:1019-27. [PMID: 18270976 DOI: 10.1002/ajmg.b.30697] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Autism is a pervasive developmental disorder diagnosed in early childhood. Abnormalities of serotonergic neurotransmission have been reported in autism. Serotonin transporter (SERT) modulates serotonin levels, and is a major therapeutic target in autism. Factors that regulate SERT expression might be implicated in the pathophysiology of autism. One candidate SERT regulatory protein is the roundabout axon guidance molecule, ROBO. SerT expression in Drosophila is regulated by robo; it plays a vital role in mammalian neurodevelopment also. Here, we examined the associations of ROBO3 and ROBO4 with autism, in a trio association study using DNA from 252 families recruited to AGRE. Four SNPs of ROBO3 (rs3923890, P = 0.023; rs7925879, P = 0.017; rs4606490, P = 0.033; and rs3802905, P = 0.049) and a single SNP of ROBO4 (rs6590109, P = 0.009) showed associations with autism; the A/A genotype of rs3923890 showed lower ADI-R_A scores, which reflect social interaction. Significant haplotype associations were also observed for ROBO3 and ROBO4. We further compared the mRNA expressions of ROBO1, ROBO2, ROBO3, and ROBO4 in the lymphocytes of 19 drug-naïve autistic patients and 20 age- and sex-matched controls. Expressions of ROBO1 (P = 0.018) and ROBO2 (P = 0.023) were significantly reduced in the autistic group; the possibility of using the altered expressions of ROBO as peripheral markers for autism, may be explored. In conclusion, we suggest a possible role of ROBO in the pathogenesis of autism. Abnormalities of ROBO may lead to autism either by interfering with serotonergic system, or by disrupting neurodevelopment. To the best of our knowledge, this is the first report relating ROBO with autism.
Collapse
Affiliation(s)
- A Anitha
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dickinson RE, Myers M, Duncan WC. Novel regulated expression of the SLIT/ROBO pathway in the ovary: possible role during luteolysis in women. Endocrinology 2008; 149:5024-34. [PMID: 18566128 DOI: 10.1210/en.2008-0204] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The human corpus luteum (CL) undergoes luteolysis, associated with marked tissue and vascular remodeling, unless conception occurs and the gland is rescued by human chorionic gonadotropin (hCG). In Drosophila the Slit gene product, a secreted glycoprotein, acts as a ligand for the roundabout (robo) transmembrane receptor. Together they influence the guidance and migration of neuronal and nonneuronal cells. In vertebrates three Slit (Slit1, Slit2, Slit3) and four Robo (Robo1, Robo2, Robo3/Rig-1, Robo4/Magic Robo) genes have been identified. ROBO1, SLIT2, and SLIT3 are also inactivated in human cancers and may regulate apoptosis and metastasis. Because processes such as apoptosis and tissue remodeling occur during the regression of the CL, the aim of this study was to investigate the expression, regulation, and effects of the SLIT and ROBO genes in human luteal cells. Immunohistochemistry and RT-PCR revealed that SLIT2, SLIT3, ROBO1, and ROBO2 are expressed in luteal steroidogenic cells and fibroblast-like cells of the human CL. Furthermore, using real-time quantitative PCR, expression of SLIT2, SLIT3, and ROBO2 was maximal in the late-luteal phase and significantly reduced after luteal rescue in vivo with exogenous hCG (P<0.05). Additionally, hCG significantly inhibited SLIT2, SLIT3, and ROBO2 expression in cultured luteinized granulosa cells (P<0.05). Blocking SLIT-ROBO activity increased migration and significantly decreased levels of apoptosis in primary cultures of luteal cells (P<0.05). Overall, these results suggest the SLIT/ROBO pathway could play an important role in luteolysis in women.
Collapse
Affiliation(s)
- Rachel E Dickinson
- Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, Department of Reproductive and Developmental Sciences, The Queen's Medical Research Institute, Edinburgh, Scotland, United Kingdom.
| | | | | |
Collapse
|
47
|
Prasad A, Paruchuri V, Preet A, Latif F, Ganju RK. Slit-2 induces a tumor-suppressive effect by regulating beta-catenin in breast cancer cells. J Biol Chem 2008; 283:26624-33. [PMID: 18611862 DOI: 10.1074/jbc.m800679200] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SLIT-2 is considered as a candidate tumor suppressor gene, because it is frequently inactivated in various cancers due to hypermethylation of its promoter region and allelic loss. However, the exact mechanism of its tumor-suppressive effect has not been elucidated. Here, we observed that Slit-2-overexpressing breast cancer cells exhibited decreased proliferation and migration capabilities compared with control cells under in vitro conditions. These results were confirmed in vivo in mouse model systems. Mice injected with MCF-7/Slit-2 cells showed a 60-70% reduction in tumor size compared with mice injected with MCF-7/VC cells both in the absence and presence of estrogen. Upon further elucidation, we observed that Slit-2 mediates the tumor-suppressive effect via a coordinated regulation of the beta-catenin and PI3K signaling pathways and by enhancing beta-catenin/E-cadherin-mediated cell-cell adhesion. Our study for the first time reveals that Slit-2-overexpressing breast cancer cells exhibit tumor suppressor capabilities through the novel mechanism of beta-catenin modulation.
Collapse
Affiliation(s)
- Anil Prasad
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|
48
|
Abstract
The mammalian kidney consists of highly specialised cells that function in an integrated manner to maintain homeostasis of body fluids, electrolytes and nutrients. Formation of multicellular structures and differentiation of kidney cells are tightly regulated processes that begin during the fifth week and end by approximately 34 weeks of human gestation. This review focuses on the morphological, cellular and molecular steps required for kidney formation. Although some of this information is derived from studies of the human kidney, much has arisen from the study of genetic models of mammalian kidney morphogenesis. These models reveal mechanisms by which cell lineages are established in the embryonic kidney and the genetic pathways that are involved in their establishment and maintenance.
Collapse
Affiliation(s)
- Norman D Rosenblum
- The Hospital for Sick Children, Division of Nephrology and Program in Developmental and Stem Cell Biology, 555 University Avenue, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Shiau CE, Lwigale PY, Das RM, Wilson SA, Bronner-Fraser M. Robo2-Slit1 dependent cell-cell interactions mediate assembly of the trigeminal ganglion. Nat Neurosci 2008; 11:269-76. [PMID: 18278043 DOI: 10.1038/nn2051] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 01/18/2008] [Indexed: 11/08/2022]
Abstract
Vertebrate cranial sensory ganglia, responsible for sensation of touch, taste and pain in the face and viscera, are composed of both ectodermal placode and neural crest cells. The cellular and molecular interactions allowing generation of complex ganglia remain unknown. Here, we show that proper formation of the trigeminal ganglion, the largest of the cranial ganglia, relies on reciprocal interactions between placode and neural crest cells in chick, as removal of either population resulted in severe defects. We demonstrate that ingressing placode cells express the Robo2 receptor and early migrating cranial neural crest cells express its cognate ligand Slit1. Perturbation of this receptor-ligand interaction by blocking Robo2 function or depleting either Robo2 or Slit1 using RNA interference disrupted proper ganglion formation. The resultant disorganization mimics the effects of neural crest ablation. Thus, our data reveal a novel and essential role for Robo2-Slit1 signaling in mediating neural crest-placode interactions during trigeminal gangliogenesis.
Collapse
Affiliation(s)
- Celia E Shiau
- Division of Biology 139-74, California Institute of Technology, Pasadena, California 91125, USA
| | | | | | | | | |
Collapse
|
50
|
Wang LJ, Zhao Y, Han B, Ma YG, Zhang J, Yang DM, Mao JW, Tang FT, Li WD, Yang Y, Wang R, Geng JG. Targeting Slit-Roundabout signaling inhibits tumor angiogenesis in chemical-induced squamous cell carcinogenesis. Cancer Sci 2008; 99:510-7. [PMID: 18201275 PMCID: PMC11159389 DOI: 10.1111/j.1349-7006.2007.00721.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Slit is a secreted protein known to function through the Roundabout (Robo) receptor as a repellent for axon guidance and neuronal migration, and as an inhibitor in leukocyte chemotaxis. We have previously shown that Slit2 is also secreted by a variety of human cancer cells whereby it acts as a chemoattractant to vascular endothelial cells for tumor angiogenesis. We used a blocking antibody to investigate the role of Slit-Robo signaling in tumor angiogenesis during oral carcinogenesis. In this report we undertook a multistage model of 7,12-dimethyl-1,2-benzanthracene-induced squamous cell carcinoma in the hamster buccal pouch. R5, a monoclonal antibody against the first immunoglobulin domain of Robo1, was used to study whether R5 blocks the Slit-Robo interaction and furthermore inhibits tumor angiogenesis and growth in our model. In addition, the expression of Slit2, von Willebrand factor, and vascular endothelial growth factor were examined using human tissue of oral cheek mucosa with oral squamous cell carcinoma. Our data showed that Slit2 was expressed minimally in normal and hyperplastic mucosa, moderately in dysplastic mucosa, and highly in neoplastic mucosa obtained from hamster buccal pouch. We also found that increased Slit2 expression was associated with higher tumor angiogenesis, as reflected by increased vascular endothelial growth factor expression and microvessel density. A similar Slit2 expression profile was found in human tissue. Importantly, interruption of the Slit2-Robo interaction using R5 inhibited tumor angiogenesis and growth in our in vivo model, which indicates that Slit2-mediated tumor angiogenesis is a critical process underlying the carcinogenesis of chemical-induced squamous cell carcinoma. Therefore, targeting Slit-Robo signaling may offer a novel antiangiogenesis approach for oral cancer therapy.
Collapse
Affiliation(s)
- Li-Jing Wang
- Department of Biochemistry and Molecular Biology, Schools of Life Sciences, Lanzhou University, Lanzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|