1
|
Brown PA. Transcriptomic signatures of atheroresistance in the human atrium and ventricle highlight potential candidates for targeted atherosclerosis therapeutics. Biochem Biophys Rep 2025; 42:102007. [PMID: 40248137 PMCID: PMC12004712 DOI: 10.1016/j.bbrep.2025.102007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Atherosclerosis risk is not uniform throughout the cardiovascular system. This study therefore aimed to compare the transcriptomes of atheroresistant human atrium and ventricle with atheroprone coronary arteries to identify transcriptomic signatures of atheroresistance and potential targets for atherosclerosis therapeutics. Using publicly available gene read counts, differentially expressed genes between the atrium, ventricle, and coronary artery were identified for each contrast and validated against the Swiss Institute of Bioinformatics' Bgee database. Over-representation analysis and active-subnetwork-oriented enrichment assessment then identified enriched terms, which were grouped into endothelial dysfunction-related processes. Potential biological significance was further explored with pathway analysis. Among 21474 features, 12656 differentially expressed genes were identified across the three contrasts and associated with 1215 enriched terms. There were 315 down-regulated and 133 up-regulated genes associated with endothelial dysfunction-related processes across the contrasts, including immune modulators, cell adhesion molecules, and lipid metabolism- and coagulation-related molecules. Differentially expressed genes were associated with six down-regulated Kyoto Encyclopedia of Genes and Genomes pathways, related to immune cell and associated endothelium functions. Review of regulated genes associated with endothelial dysfunction-related processes and included in these pathways, indicate immune cell-associated B cell scaffold protein with ankyrin repeats 1, as well as arterial endothelial cell-associated vascular cell adhesion molecule 1 and cadherin 5, as potential atherosclerosis targets.
Collapse
Affiliation(s)
- Paul A. Brown
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Kingston 7, Jamaica
| |
Collapse
|
2
|
Meng Y, Sui L, Xu T, Zhao H, Yuan Q, Sun L. Research and Application Prospect of Nanomedicine in Kidney Disease: A Bibliometric Analysis From 2003 to 2024. Int J Nanomedicine 2025; 20:3007-3030. [PMID: 40093546 PMCID: PMC11910916 DOI: 10.2147/ijn.s510016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Kidney disease is a major public health concern that has a significant effect on a patient's life span and quality of life. However, effective treatment for most kidney diseases is lacking. Nanotechnology mainly explores the design, characterization, production, and applications of objects in the nanoscale range and has been widely used in the medical field. To date, there has been an increasing amount of research on the application of nanotechnology in kidney disease. However, systematic bibliometric studies remain rare. In this review, data collected from the Web of Science Core Collection database until December 31, 2024, were subjected to a bibliometric analysis. A total of 1179 articles and reviews were included. The publication trends, countries, institutions, authors, co-authorship, co-citations, journals, keywords, and references pertaining to this topic were examined. The results showed that nanotechnology research in kidney disease is increasing. The leading country, organization, and author were China, Sichuan University, and Professor Peng Huang, respectively. ACS APPLIED MATERIALS & INTERFACES was the top journal among the 464 journals in which articles on nanotechnology in kidney disease were published. KIDNEY INTERNATIONAL was the most cited journal in the field. The most significant increases were shown for "acute kidney disease", "drug delivery", "oxidative stress", "diabetic nephropathy", and "chronic kidney disease", indicating the current research hotspots. Furthermore, the development prospects and challenges of nanotechnology in kidney disease were discussed in this review. How to achieve precise drug delivery to render kidney-targeting therapy a reality may be problematic in future studies.
Collapse
Affiliation(s)
- Yilin Meng
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, People’s Republic of China
| | - Lu Sui
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, People’s Republic of China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, People’s Republic of China
| | - Hainan Zhao
- Department of Nephrology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou121001, People’s Republic of China
| | - Quan Yuan
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Li Sun
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, People’s Republic of China
| |
Collapse
|
3
|
Weiss D, Yeung N, Ramachandra AB, Humphrey JD. Transcriptional regulation of postnatal aortic development. Cells Dev 2024; 180:203971. [PMID: 39426523 PMCID: PMC11634634 DOI: 10.1016/j.cdev.2024.203971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/14/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
The aorta exhibits tremendous changes in geometry, composition, and mechanical properties during postnatal development. These changes are necessarily driven by transcriptional changes, both genetically programmed and mechano-responsive, but there has not been a careful comparison of time-course changes in the transcriptional profile and biomechanical phenotype. Here, we show that the greatest period of differential gene expression in the normal postnatal mouse aorta occurs prior to weaning at three weeks of age though with important evolution of many transcripts thereafter. We identify six general temporal patterns, including transcripts that monotonically decrease to lower or increase to higher steady state values as well as those that either peak or dip prior to or near weaning. We show that diverse transcripts within individual groupings correlate well over time, and that sub-sets of these groups correlate well with the developmental progression of different biomechanical metrics that are expected to be involved in mechano-sensing. In particular, expression of genes for elastin and elastin-associated glycoproteins tend to correlate well with the ratio of systolic-to-diastolic stress whereas genes for collagen fibers correlate well with the daily rate of change of systolic stress and genes for mechano-sensing proteins tend to correlate well with the systolic stress itself. We conclude that different groupings of genes having different temporal expression patterns correlate well with different measures of the wall mechanics, hence emphasizing a need for age-dependent, gene-specific computational modeling of postnatal development.
Collapse
Affiliation(s)
- D Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Mechanical & Materials Engineering, University of Denver, Denver, CO, USA
| | - N Yeung
- School of the Biological Sciences, University of Cambridge, Cambridge, UK
| | - A B Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Mechanical Engineering, Iowa State University, Ames, IA, USA
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Pavlović N, Kelam N, Racetin A, Filipović N, Pogorelić Z, Prusac IK, Vukojević K. Expression Profiles of ITGA8 and VANGL2 Are Altered in Congenital Anomalies of the Kidney and Urinary Tract (CAKUT). Molecules 2024; 29:3294. [PMID: 39064873 PMCID: PMC11279313 DOI: 10.3390/molecules29143294] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Kidney failures in infants are mostly caused by congenital anomalies of the kidney and urinary tract (CAKUT), which are among the most common congenital birth disorders worldwide when paired with cardiac abnormalities. People with CAKUT often have severe kidney failure as a result of a wide range of abnormalities that can occur alone or in conjunction with other syndromic disorders. In this study, we aimed to investigate the expression pattern of CAKUT candidate genes alpha-8 integrin (ITGA8) and Van Gogh-like 2 (VANGL2) in fetal tissues of healthy and CAKUT-affected kidneys using immunohistochemistry and immunofluorescence. We found that under CAKUT circumstances, the expressions of ITGA8 and VANGL2 are changed. Additionally, we showed that VANGL2 expression is constant during fetal aging, but ITGA8 expression varies. Moreover, compared to normal healthy kidneys (CTRL), ITGA8 is poorly expressed in duplex kidneys (DKs) and dysplastic kidneys (DYS), whereas VANGL2 is substantially expressed in dysplastic kidneys (DYS) and poorly expressed in hypoplastic kidneys (HYP). These results point to VANGL2 and ITGA8 as potential prognostic indicators for CAKUT malformations. Further research is necessary to explore the molecular mechanisms underlying this differential expression of ITGA8 and VANGL2.
Collapse
Affiliation(s)
- Nikola Pavlović
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, 21000 Split, Croatia; (N.P.); (K.V.)
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, 21000 Split, Croatia; (N.P.); (K.V.)
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, 21000 Split, Croatia; (N.P.); (K.V.)
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, 21000 Split, Croatia; (N.P.); (K.V.)
| | - Zenon Pogorelić
- Department of Pediatric Surgery, University Hospital of Split, 21000 Split, Croatia
- Department of Surgery, School of Medicine, University of Split, 21000 Split, Croatia
| | - Ivana Kuzmić Prusac
- Department of Pathology, University Hospital Centre Split, Spinciceva 1, 21000 Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, 21000 Split, Croatia; (N.P.); (K.V.)
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
5
|
Kwartler CS, Pinelo JEE. Use of iPSC-Derived Smooth Muscle Cells to Model Physiology and Pathology. Arterioscler Thromb Vasc Biol 2024; 44:1523-1536. [PMID: 38695171 PMCID: PMC11209779 DOI: 10.1161/atvbaha.123.319703] [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: 06/28/2024]
Abstract
The implementation of human induced pluripotent stem cell (hiPSC) models has introduced an additional tool for identifying molecular mechanisms of disease that complement animal models. Patient-derived or CRISPR/Cas9-edited induced pluripotent stem cells differentiated into smooth muscle cells (SMCs) have been leveraged to discover novel mechanisms, screen potential therapeutic strategies, and model in vivo development. The field has evolved over almost 15 years of research using hiPSC-SMCs and has made significant strides toward overcoming initial challenges such as the lineage specificity of SMC phenotypes. However, challenges both specific (eg, the lack of specific markers to thoroughly validate hiPSC-SMCs) and general (eg, a lack of transparency and consensus around methodology in the field) remain. In this review, we highlight the recent successes and remaining challenges of the hiPSC-SMC model.
Collapse
Affiliation(s)
- Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jose Emiliano Esparza Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
6
|
Komatsu S, Kato N, Kitai H, Funahashi Y, Noda Y, Tsubota S, Tanaka A, Sato Y, Maeda K, Saito S, Furuhashi K, Ishimoto T, Kosugi T, Maruyama S, Kadomatsu K. Detecting and exploring kidney-derived extracellular vesicles in plasma. Clin Exp Nephrol 2024; 28:617-628. [PMID: 38436899 PMCID: PMC11190017 DOI: 10.1007/s10157-024-02464-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/13/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) have received considerable attention as ideal biomarkers for kidney diseases. Most reports have focused on urinary EVs, that are mainly derived from the cells in the urinary tract. However, the detection and the application of kidney-derived EVs in plasma remains uncertain. METHODS We examined the kidney-derived small EVs (sEVs) in plasma that were supposedly released from renal mesangial and glomerular endothelial cells, using clinical samples from healthy controls and patients with kidney transplants. Plasma from healthy controls underwent ultracentrifugation, followed by on-bead flow cytometry, targeting α8 integrin, an antigen-specific to mesangial cells. To confirm the presence of kidney-derived sEVs in peripheral blood, plasma from ABO-incompatible kidney transplant recipients was ultracentrifuged, followed by western blotting for donor blood type antigens. RESULTS Immunohistochemistry and immunoelectron microscopy confirmed α8 integrin expression in kidney mesangial cells and their sEVs. The CD9-α8 integrin double-positive sEVs were successfully detected using on-bead flow cytometry. Western blot analysis further revealed transplanted kidney-derived sEVs containing blood type B antigens in non-blood type B recipients, who had received kidneys from blood type B donors. Notably, a patient experiencing graft kidney loss exhibited diminished signals of sEVs containing donor blood type antigens. CONCLUSION Our findings demonstrate the potential usefulness of kidney-derived sEVs in plasma in future research for kidney diseases.
Collapse
Affiliation(s)
- Shintaro Komatsu
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Noritoshi Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Hiroki Kitai
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshio Funahashi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuhei Noda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akihito Tanaka
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuka Sato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kayaho Maeda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoji Saito
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuhiro Furuhashi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takuji Ishimoto
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
7
|
Lyu QR, Fu K. Tissue-specific Cre driver mice to study vascular diseases. Vascul Pharmacol 2023; 153:107241. [PMID: 37923099 DOI: 10.1016/j.vph.2023.107241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Vascular diseases, including atherosclerosis and abdominal aneurysms, are the primary cause of mortality and morbidity among the elderly worldwide. The life quality of patients is significantly compromised due to inadequate therapeutic approaches and limited drug targets. To expand our comprehension of vascular diseases, gene knockout (KO) mice, especially conditional knockout (cKO) mice, are widely used for investigating gene function and mechanisms of action. The Cre-loxP system is the most common method for generating cKO mice. Numerous Cre driver mice have been established to study the main cell types that compose blood vessels, including endothelial cells, smooth muscle cells, and fibroblasts. Here, we first discuss the characteristics of each layer of the arterial wall. Next, we provide an overview of the representative Cre driver mice utilized for each of the major cell types in the vessel wall and their most recent applications in vascular biology. We then go over Cre toxicity and discuss the practical methods for minimizing Cre interference in experimental outcomes. Finally, we look into the future of tissue-specific Cre drivers by introducing the revolutionary single-cell RNA sequencing and dual recombinase system.
Collapse
Affiliation(s)
- Qing Rex Lyu
- Medical Research Center, Chongqing General Hospital, Chongqing 401147, China; Chongqing Academy of Medical Sciences, Chongqing 401147, China.
| | - Kailong Fu
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
8
|
Deaton RA, Bulut G, Serbulea V, Salamon A, Shankman LS, Nguyen AT, Owens GK. A New Autosomal Myh11-CreERT2 Smooth Muscle Cell Lineage Tracing and Gene Knockout Mouse Model-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:203-211. [PMID: 36519470 PMCID: PMC9877184 DOI: 10.1161/atvbaha.122.318160] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The Myh11 promoter is extensively used as a smooth muscle cell (SMC) Cre-driver and is regarded as the most restrictive and specific promoter available to study SMCs. Unfortunately, in the existing Myh11-CreERT2 mouse, the transgene was inserted on the Y chromosome precluding the study of female mice. Given the importance of including sex as a biological variable and that numerous SMC-based diseases have a sex-dependent bias, the field has been tremendously limited by the lack of a model to study both sexes. Here, we describe a new autosomal Myh11-CreERT2 mouse (referred to as Myh11-CreERT2-RAD), which allows for SMC-specific lineage tracing and gene knockout studies in vivo using both male and female mice. METHODS A Myh11-CreERT2-RAD transgenic C57BL/6 mouse line was generated using bacterial artificial chromosome clone RP23-151J22 modified to contain a Cre-ERT2 after the Myh11 start codon. Myh11-CreERT2-RAD mice were crossed with 2 different fluorescent reporter mice and tested for SMC-specific labeling by flow cytometric and immunofluorescence analyses. RESULTS Myh11-CreERT2-RAD transgene insertion was determined to be on mouse chromosome 2. Myh11-CreERT2-RAD fluorescent reporter mice showed Cre-dependent, tamoxifen-inducible labeling of SMCs equivalent to the widely used Myh11-CreERT2 mice. Labeling was equivalent in both male and female Cre+ mice and was limited to vascular and visceral SMCs and pericytes in various tissues as assessed by immunofluorescence. CONCLUSIONS We generated and validated the function of an autosomal Myh11-CreERT2-RAD mouse that can be used to assess sex as a biological variable with respect to the normal and pathophysiological functions of SMCs.
Collapse
Affiliation(s)
- Rebecca A. Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Gamze Bulut
- College of William and Mary, Williamsburg, Virginia 23185
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Anita Salamon
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | | | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| |
Collapse
|
9
|
Gómez-Conde S, Dunand O, Hummel A, Morinière V, Gauthier M, Mesnard L, Heidet L. Bi-allelic pathogenic variants in ITGA8 cause slowly progressive renal disease of unknown etiology. Clin Genet 2023; 103:114-118. [PMID: 36089563 DOI: 10.1111/cge.14229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 12/13/2022]
Abstract
Integrin Subunit Alpha 8 gene (ITGA8) encodes an integrin chain that is known to be critical in the early stage of the kidney development. Bi-allelic pathogenic variants in ITGA8 are associated with bilateral renal agenesis, as well as anomalies involving urogenital system. Here, we report two unrelated patients presenting with slowly progressing chronic kidney disease associated with bilateral renal hypodysplasia carrying homozygous loss of function variants in the ITGA8 gene. These results broaden the clinical and genotypic spectrum of ITGA8 defects, revealing the high and unexpected degree of phenotypic heterogeneity of this autosomal recessive disease. Our study emphasizes the usefulness of Next-Generation Sequencing in unraveling the genetic cause of chronic kidney disease of unknown etiology, and raises the question of genetic modifiers involved in the variation of the phenotypes associated with autosomal recessive ITGA8 pathogenic variants.
Collapse
Affiliation(s)
- Sara Gómez-Conde
- APHP-Centre, Service de Néphrologie Pédiatrique, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Universitaire Necker-Enfants malades, Institut Imagine, Université Paris-Cité, Paris, France
| | - Olivier Dunand
- Service de Néphrologie Pédiatrique, Centre Hospitalier Universitaire Felix Guyon, Saint Denis, France
| | - Aurélie Hummel
- APHP-Centre, Service de Néphrologie, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Universitaire Necker-Enfants malades, Paris, France
| | - Vincent Morinière
- APHP-Centre, Fédération de Génétique et Médecine Génomique, Service de Médecine Génomique des Maladies Rares, Hôpital Universitaire Necker-Enfants malades, Paris, France
| | - Marion Gauthier
- Service de Néphrologie et Dialyse, Hôpital André Grégoire, Montreuil, France
| | - Laurent Mesnard
- APHP-Sorbonne Université, Département de Néphrologie, Hôpital Tenon, Service des Soins Intensifs Néphrologiques et Rein Aigu (SINRA), Paris, France
| | - Laurence Heidet
- APHP-Centre, Service de Néphrologie Pédiatrique, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Universitaire Necker-Enfants malades, Institut Imagine, Université Paris-Cité, Paris, France
| |
Collapse
|
10
|
Warthi G, Faulkner JL, Doja J, Ghanam AR, Gao P, Yang AC, Slivano OJ, Barris CT, Kress TC, Zawieja SD, Griffin SH, Xie X, Ashworth A, Christie CK, Bryant WB, Kumar A, Davis MJ, Long X, Gan L, de Chantemèle EJB, Lyu Q, Miano JM. Generation and Comparative Analysis of an Itga8-CreER T2 Mouse with Preferential Activity in Vascular Smooth Muscle Cells. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1084-1100. [PMID: 36424917 PMCID: PMC9681021 DOI: 10.1038/s44161-022-00162-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/29/2022] [Indexed: 11/12/2022]
Abstract
All current smooth muscle cell (SMC) Cre mice similarly recombine floxed alleles in vascular and visceral SMCs. Here, we present an Itga8-CreER T2 knock-in mouse and compare its activity with a Myh11-CreER T2 mouse. Both Cre drivers demonstrate equivalent recombination in vascular SMCs. However, Myh11-CreER T2 mice, but not Itga8-CreER T2 mice, display high activity in visceral SMC-containing tissues such as intestine, show early tamoxifen-independent activity, and produce high levels of CreERT2 protein. Whereas Myh11-CreER T2 -mediated knockout of serum response factor (Srf) causes a lethal intestinal phenotype precluding analysis of the vasculature, loss of Srf with Itga8-CreER T2 (Srf Itga8 ) yields viable mice with no evidence of intestinal pathology. Male and female Srf Itga8 mice exhibit vascular contractile incompetence, and angiotensin II causes elevated blood pressure in wild type, but not Srf Itga8 , male mice. These findings establish the Itga8-CreER T2 mouse as an alternative to existing SMC Cre mice for unfettered phenotyping of vascular SMCs following selective gene loss.
Collapse
Affiliation(s)
- Ganesh Warthi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jessica L. Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jaser Doja
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Amr R. Ghanam
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Pan Gao
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Allison C. Yang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Orazio J. Slivano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Taylor C. Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Scott D. Zawieja
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Susan H. Griffin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Xiaoling Xie
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94158
| | - Christine K. Christie
- Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - William B. Bryant
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Ajay Kumar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Michael J. Davis
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Lin Gan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | | | - Qing Lyu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Biomedical and Health Institute, Chongqing Institute of Green and Intelligence Technology, Chongqing, China 400714
- Chongqing General Hospital, Chongqing, China 401147
| | - Joseph M. Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| |
Collapse
|
11
|
A genotype-first analysis in a cohort of Mullerian anomaly. J Hum Genet 2022; 67:347-352. [PMID: 35022528 DOI: 10.1038/s10038-021-00996-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/08/2022]
Abstract
Müllerian anomaly (M.A.) is a group of congenital anatomic abnormalities caused by aberrations of the development process of the Müllerian duct. M.A. can either be isolated or be involved in Mendelian syndromes, such as Dandy-Walker syndrome, Holt-Oram syndrome and Bardet-Biedl syndrome, which are often associated with both uterus and kidney malformations. In this study, we applied a genotype-first approach to analyze the whole-exome sequencing data of 492 patients with M.A. Six potential pathogenic variants were found in five genes previously related to female urogenital deformities (PKD1, SON, SALL1, BMPR1B, ITGA8), which are partially overlapping with our patients' phenotypes. We further identified eight incidental findings in seven genes related to Mendelian syndromes without known association with reproductive anomalies (TEK, COL11A1, ANKRD11, LEMD3, DLG5, SPTB, BMP2), which represent potential phenotype expansions of these genes.
Collapse
|
12
|
Yokosaki Y, Nishimichi N. New Therapeutic Targets for Hepatic Fibrosis in the Integrin Family, α8β1 and α11β1, Induced Specifically on Activated Stellate Cells. Int J Mol Sci 2021; 22:12794. [PMID: 34884600 PMCID: PMC8657911 DOI: 10.3390/ijms222312794] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
A huge effort has been devoted to developing drugs targeting integrins over 30 years, because of the primary roles of integrins in the cell-matrix milieu. Five αv-containing integrins, in the 24 family members, have been a central target of fibrosis. Currently, a small molecule against αvβ1 is undergoing a clinical trial for NASH-associated fibrosis as a rare agent aiming at fibrogenesis. Latent TGFβ activation, a distinct talent of αv-integrins, has been intriguing as a therapeutic target. None of the αv-integrin inhibitors, however, has been in the clinical market. αv-integrins commonly recognize an Arg-Gly-Asp (RGD) sequence, and thus the pharmacophore of inhibitors for the 5-integrins is based on the same RGD structure. The RGD preference of the integrins, at the same time, dilutes ligand specificity, as the 5-integrins share ligands containing RGD sequence such as fibronectin. With the inherent little specificity in both drugs and targets, "disease specificity" has become less important for the inhibitors than blocking as many αv-integrins. In fact, an almighty inhibitor for αv-integrins, pan-αv, was in a clinical trial. On the contrary, approved integrin inhibitors are all specific to target integrins, which are expressed in a cell-type specific manner: αIIbβ3 on platelets, α4β1, α4β7 and αLβ2 on leukocytes. Herein, "disease specific" integrins would serve as attractive targets. α8β1 and α11β1 are selectively expressed in hepatic stellate cells (HSCs) and distinctively induced upon culture activation. The exceptional specificity to activated HSCs reflects a rather "pathology specific" nature of these new integrins. The monoclonal antibodies against α8β1 and α11β1 in preclinical examinations may illuminate the road to the first medical agents.
Collapse
Affiliation(s)
- Yasuyuki Yokosaki
- Integrin-Matrix Biomedical Science, Translational Research Center, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan;
| | | |
Collapse
|
13
|
Regina C, Hamed E, Andrieux G, Angenendt S, Schneider M, Ku M, Follo M, Wachtel M, Ke E, Kikuchi K, Henssen AG, Schäfer BW, Boerries M, Wagers AJ, Keller C, Hettmer S. Negative correlation of single-cell PAX3:FOXO1 expression with tumorigenicity in rhabdomyosarcoma. Life Sci Alliance 2021; 4:4/9/e202001002. [PMID: 34187933 PMCID: PMC8321661 DOI: 10.26508/lsa.202001002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/11/2022] Open
Abstract
Single-cell PAX3:FOXO1 expression in rhabdomyosarcoma is variable. PAX3:FOXO1 low cell states are characterized by more efficient adhesion, migration and tumor-propagating capacity. Rhabdomyosarcomas (RMS) are phenotypically and functionally heterogeneous. Both primary human RMS cultures and low-passage Myf6Cre,Pax3:Foxo1,p53 mouse RMS cell lines, which express the fusion oncoprotein Pax3:Foxo1 and lack the tumor suppressor Tp53 (Myf6Cre,Pax3:Foxo1,p53), exhibit marked heterogeneity in PAX3:FOXO1 (P3F) expression at the single cell level. In mouse RMS cells, P3F expression is directed by the Pax3 promoter and coupled to eYFP. YFPlow/P3Flow mouse RMS cells included 87% G0/G1 cells and reorganized their actin cytoskeleton to produce a cellular phenotype characterized by more efficient adhesion and migration. This translated into higher tumor-propagating cell frequencies of YFPlow/P3Flow compared with YFPhigh/P3Fhigh cells. Both YFPlow/P3Flow and YFPhigh/P3Fhigh cells gave rise to mixed clones in vitro, consistent with fluctuations in P3F expression over time. Exposure to the anti-tropomyosin compound TR100 disrupted the cytoskeleton and reversed enhanced migration and adhesion of YFPlow/P3Flow RMS cells. Heterogeneous expression of PAX3:FOXO1 at the single cell level may provide a critical advantage during tumor progression.
Collapse
Affiliation(s)
- Carla Regina
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Ebrahem Hamed
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sina Angenendt
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Michaela Schneider
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Manching Ku
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Wachtel
- University Children's Hospital, Children's Research Center and Department of Oncology, Zürich, Switzerland
| | - Eugene Ke
- Department of Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Ken Kikuchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Anton G Henssen
- Experimental and Clinical Research Center of the Max Delbrück Center and Charité Berlin, Berlin, Germany
| | - Beat W Schäfer
- University Children's Hospital, Children's Research Center and Department of Oncology, Zürich, Switzerland
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Centre Freiburg, Medical Center-University of Freiburg, Freiburg, Germany
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, MA, USA.,Joslin Diabetes Center, Boston, MA, USA.,Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, Beaverton, OR, USA
| | - Simone Hettmer
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany .,Comprehensive Cancer Centre Freiburg, Medical Center-University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Freiburg, Germany
| |
Collapse
|
14
|
Wang Y, Wang W, Yang X, Chen W, Yang X, Pan X, Xu P, Zhu W, Han Y, Chen X. ITGA8 positive cells in the conventional outflow tissue exhibit Schlemm's canal endothelial cell properties. Life Sci 2021; 278:119564. [PMID: 33961857 DOI: 10.1016/j.lfs.2021.119564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 11/17/2022]
Abstract
AIMS Elevated intraocular pressure is primarily induced by the increased resistance of conventional outflow of aqueous humor. Dysfunction of the juxtacanalicular region of trabecular meshwork (TM) and Schlemm's canal (SC) endothelium, as the main conventional outflow tissue, have been implicated as the major reasons for the increased resistance. Integrins are widespread in these tissues, especially alpha8 integrin (ITGA8). We aim to investigate the properties of cells expressing ITGA8 in the conventional outflow tissue. MAIN METHODS Fluorescence in situ hybridization (FISH) and immunofluorescence (IF) were performed to detect the mRNA and protein levels of ITGA8 in human conventional outflow tissue. ITGA8-positive cells were isolated from the cultured human TM cells through a magnetic bead-based approach. Flow Cytometry was used to determine the purification efficiency. The expressions of TM and SC biomarkers and dexamethasone-induced myocilin secretion capacity of ITGA8-positive cells was assessed by Real-time PCR, IF and Western blot. A gel contraction assay was performed to evaluate contractility of ITGA8-positive cells after endothelin 1 treatment. KEY FINDINGS ITGA8 was found with robust expression near the inner wall of SC endothelium. After purification, the proportion of ITGA8-positive cells were increased by about 10%. ITGA8-positive cells were identified with the properties as SC endothelial cells, such as more robust expressions of SC biomarkers, less dexamethasone-inducible myocilin expression, and stronger contractility. SIGNIFICANCE This study demonstrated that cells expressing ITGA8 in SC region possess more properties as SC endothelial cells. Our data implicate a crucial role of ITGA8 in aqueous humor (AH) outflow resistance regulation.
Collapse
Affiliation(s)
- Yanan Wang
- School of Basic Medicine, Qingdao University, Qingdao 266021, China; Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Wenyan Wang
- School of Basic Medicine, Qingdao University, Qingdao 266021, China; Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Xuejiao Yang
- Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Wenshi Chen
- Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xian Yang
- Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaojing Pan
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Peilong Xu
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Wei Zhu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang, University & Capital Medical University, Beijing 100730, China
| | - Yantao Han
- School of Basic Medicine, Qingdao University, Qingdao 266021, China.
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
15
|
Nephronectin as a Matrix Effector in Cancer. Cancers (Basel) 2021; 13:cancers13050959. [PMID: 33668838 PMCID: PMC7956348 DOI: 10.3390/cancers13050959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The extracellular matrix provides an important scaffold for cells and tissues of multicellular organisms. The scaffold not only provides a secure anchorage point, but also functions as a reservoir for signalling molecules, sequestered and released when necessary. A dysregulated extracellular matrix may therefore modulate cellular behaviour, as seen during cancer progression. The extracellular matrix protein nephronectin was discovered two decades ago and found to regulate important embryonic developmental processes. Loss of either nephronectin or its receptor, integrin α8β1, leads to underdeveloped kidneys. Recent findings show that nephronectin is also dysregulated in breast cancer and plays a role in promoting metastasis. To enable therapeutic intervention, it is important to fully understand the role of nephronectin and its receptors in cancer progression. In this review, we summarise the literature on nephronectin, analyse the structure and domain-related functions of nephronectin and link these functions to potential roles in cancer progression. Abstract The extracellular matrix protein nephronectin plays an important regulatory role during embryonic development, controlling renal organogenesis through integrin α8β1 association. Nephronectin has three main domains: five N-terminal epidermal growth factor-like domains, a linker region harbouring two integrin-binding motifs (RGD and LFEIFEIER), and a C-terminal MAM domain. In this review, we look into the domain-related functions of nephronectin, and tissue distribution and expression. During the last two decades it has become evident that nephronectin also plays a role during cancer progression and in particular metastasis. Nephronectin is overexpressed in both human and mouse breast cancer compared to normal breast tissue where the protein is absent. Cancer cells expressing elevated levels of nephronectin acquire increased ability to colonise distant organs. In particular, the enhancer-motif (LFEIFEIER) which is specific to the integrin α8β1 association induces viability via p38 MAPK and plays a role in colonization. Integrins have long been desired as therapeutic targets, where low efficiency and receptor redundancy have been major issues. Based on the summarised publications, the enhancer-motif of nephronectin could present a novel therapeutic target.
Collapse
|
16
|
Muhl L, Genové G, Leptidis S, Liu J, He L, Mocci G, Sun Y, Gustafsson S, Buyandelger B, Chivukula IV, Segerstolpe Å, Raschperger E, Hansson EM, Björkegren JLM, Peng XR, Vanlandewijck M, Lendahl U, Betsholtz C. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat Commun 2020; 11:3953. [PMID: 32769974 PMCID: PMC7414220 DOI: 10.1038/s41467-020-17740-1] [Citation(s) in RCA: 366] [Impact Index Per Article: 73.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/10/2020] [Indexed: 12/25/2022] Open
Abstract
Many important cell types in adult vertebrates have a mesenchymal origin, including fibroblasts and vascular mural cells. Although their biological importance is undisputed, the level of mesenchymal cell heterogeneity within and between organs, while appreciated, has not been analyzed in detail. Here, we compare single-cell transcriptional profiles of fibroblasts and vascular mural cells across four murine muscular organs: heart, skeletal muscle, intestine and bladder. We reveal gene expression signatures that demarcate fibroblasts from mural cells and provide molecular signatures for cell subtype identification. We observe striking inter- and intra-organ heterogeneity amongst the fibroblasts, primarily reflecting differences in the expression of extracellular matrix components. Fibroblast subtypes localize to discrete anatomical positions offering novel predictions about physiological function(s) and regulatory signaling circuits. Our data shed new light on the diversity of poorly defined classes of cells and provide a foundation for improved understanding of their roles in physiological and pathological processes. To define and distinguish fibroblasts from vascular mural cells have remained challenging. Here, using single-cell RNA sequencing and tissue imaging, the authors provide a molecular basis for cell type classification and reveal inter- and intra-organ diversity of these cell types.
Collapse
Affiliation(s)
- Lars Muhl
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden. .,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden.
| | - Guillem Genové
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Stefanos Leptidis
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Jianping Liu
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Liqun He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury, Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China.,Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammerskjölds väg 20, SE-75185, Uppsala, Sweden
| | - Giuseppe Mocci
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammerskjölds väg 20, SE-75185, Uppsala, Sweden
| | - Sonja Gustafsson
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Byambajav Buyandelger
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Indira V Chivukula
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Åsa Segerstolpe
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden.,Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elisabeth Raschperger
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Emil M Hansson
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden
| | - Johan L M Björkegren
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden.,Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM) BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Vanlandewijck
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden.,Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammerskjölds väg 20, SE-75185, Uppsala, Sweden
| | - Urban Lendahl
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, SE-17177, Stockholm, Sweden
| | - Christer Betsholtz
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157, Huddinge, Sweden. .,Department of Medicine Huddinge, Karolinska Institutet, SE-14157, Huddinge, Sweden. .,Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammerskjölds väg 20, SE-75185, Uppsala, Sweden.
| |
Collapse
|
17
|
Nephronectin promotes breast cancer brain metastatic colonization via its integrin-binding domains. Sci Rep 2020; 10:12237. [PMID: 32699247 PMCID: PMC7376038 DOI: 10.1038/s41598-020-69242-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
This study demonstrates a role for the extracellular matrix protein nephronectin (NPNT) in promoting experimental breast cancer brain metastasis, possibly through enhanced binding to- and migration through brain endothelial cells. With the introduction of more targeted breast cancer treatments, a prolonged survival has resulted during the last decade. Consequently, an increased number of patients develop metastasis in the brain, a challenging organ to treat. We recently reported that NPNT was highly expressed in primary breast cancer and associated with unfavourable prognosis. The current study addresses our hypothesis that NPNT promotes brain metastases through its integrin-binding motifs. SAGE-sequencing revealed that NPNT was significantly up-regulated in human breast cancer tissue compared to pair-matched normal breast tissue. Human brain metastatic breast cancers expressed both NPNT and its receptor, integrin α8β1. Using an open access repository; BreastMark, we found a correlation between high NPNT mRNA levels and poor prognosis for patients with the luminal B subtype. The 66cl4 mouse cell line was used for expression of wild-type and mutant NPNT, which is unable to bind α8β1. Using an in vivo model of brain metastatic colonization, 66cl4-NPNT cells showed an increased ability to form metastatic lesions compared to cells with mutant NPNT, possibly through reduced endothelial adhesion and transmigration.
Collapse
|
18
|
Tension enhances cell proliferation and collagen synthesis by upregulating expressions of integrin αvβ3 in human keloid-derived mesenchymal stem cells. Life Sci 2018; 219:272-282. [PMID: 30597173 DOI: 10.1016/j.lfs.2018.12.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/19/2018] [Accepted: 12/25/2018] [Indexed: 12/21/2022]
Abstract
AIMS Keloids are a dermal fibrotic disease whose etiology remains totally unknown and for which there is no successful treatment. Mechanical tension, in addition, is closely associated with the germination and development of keloids. In this study, we investigated the influence of human keloid-derived mesenchymal stem cells (KD-MSCs) on cell proliferation, collagen synthesis, and expressions of integrin αvβ3 under tension. MAIN METHODS KD-MSCs and human normal skin-derived mesenchymal stem cells (NS-MSCs) were isolated and cultured in stem cell medium with a gradual increase in the serum concentration. Cell proliferation and collagen synthesis were detected by Cell Counting Kit-8 (CCK-8) assay and hydroxyproline content analysis under tension respectively. We investigated the messenger RNA expressions of nine integrin subunits, including integrin units α2, α3, α5, αv, α8, α10, α11, β1, and β3, in KD-MSCs stimulated with tension. Identification of differentially expressed genes was performed by Western blot analysis and immunocytochemistry staining. KEY FINDINGS We obtained high-purity KD-MSCs and NS-MSCs using the culture method of decreasing serum concentration gradient gradually. Furthermore, we found that tension enhances cell proliferation and collagen synthesis and promotes expressions of integrin αvβ3 in KD-MSCs. In addition, blocking experiments showed that increased integrin αvβ3 expression affects cell proliferation and collagen synthesis of KD-MSCs under tension. SIGNIFICANCE Our results suggest that integrin αvβ3 receptor may be sensitive molecules of mechanical tension and could contribute to the occurrence and development of keloids. It could lead to novel targets for therapeutic intervention, treatment, and prevention of recurrence for keloid disorders.
Collapse
|
19
|
Role of integrin alpha8 in murine model of lung fibrosis. PLoS One 2018; 13:e0197937. [PMID: 29813125 PMCID: PMC5973593 DOI: 10.1371/journal.pone.0197937] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/01/2018] [Indexed: 01/13/2023] Open
Abstract
Background Integrin α8 (ITGA8) heterodimerizes with integrin β1 and is highly expressed in stromal cells of the lung. Platelet-derived growth factor receptor beta (PDGFRβ+) cells constitute a major population of contractile myofibroblasts in the lung following bleomycin-induced fibrosis. Integrin α8β1 is upregulated in fibrotic foci in bleomycin-induced lung injury. However, the functional role of ITGA8 in fibrogenesis has not been characterized. In this study, we examined whether genetic deletion of ITGA8 from PDGFRβ+ cells in the lung altered fibrosis. Methods Pdgfrb-Cre/+;Itga8flox/- or Pdgfrb-Cre/+;Itga8flox/flox (Cre+) and control mice (Cre-) were used for in vitro and in vivo studies. Primary cultures of PDGFRβ+ cells were exposed to TGFβ, followed by RNA isolation for qPCR. For in vivo studies, Cre+ and Cre- mice were characterized at baseline and after bleomycin-induced fibrosis. Results PDGFRβ-selected cells from Cre+ animals showed higher levels of Col1a1 expression after treatment with TGFβ. However, Cre- and Cre+ animals showed no significant difference in measures of acute lung injury or fibrosis following bleomycin challenge. Conclusion While ITGA8 deletion in lung PDGFRβ+ stromal cells showed evidence of greater Col1a1 mRNA expression after TGFβ treatment in vitro, no functional difference was detected in vivo.
Collapse
|
20
|
Khalifeh-Soltani A, Gupta D, Ha A, Podolsky MJ, Datta R, Atabai K. The Mfge8-α8β1-PTEN pathway regulates airway smooth muscle contraction in allergic inflammation. FASEB J 2018; 32:fj201800109R. [PMID: 29763381 DOI: 10.1096/fj.201800109r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Asthma affects ∼300 million people worldwide. Despite multiple treatment options, asthma treatment remains unsatisfactory in a subset of patients. Airway obstruction is a hallmark of allergic asthma and is largely due to airway smooth muscle hypercontractility induced by airway inflammation. Identification of molecular pathways that regulate airway smooth muscle hypercontractility is of considerable therapeutic interest. We previously identified roles for milk fat globule epidermal growth factor-like 8 (Mfge8) in opposing the effects of allergic inflammation on increasing airway smooth muscle contractile force. In this study, we delineate the signaling pathway by which Mfge8 mediates these effects. By using genetic and pharmacologic approaches, we show that the α8β1 integrin and the phosphatase and tensin homolog (PTEN) mediate the effects of Mfge8 on preventing IL-13-induced increases in airway contractility. Tracheal rings from mice with smooth muscle-specific deletion of α8β1 or PTEN have enhanced contraction in response to treatment with IL-13. Enhanced IL-13-induced tracheal ring contraction in Mfge8-/- mice was abolished by treatment with the PI3K inhibitor. Mechanistically, IL-13 induces ubiquitination and degradation of PTEN protein. Our findings identify a role for the Mfge8-α8β1-PTEN pathway in regulating the force of airway smooth muscle contraction in the setting of allergic inflammation.-Khalifeh-Soltani, A., Gupta, D., Ha, A., Podolsky, M. J., Datta, R., Atabai, K. The Mfge8-α8β1-PTEN pathway regulates airway smooth muscle contraction in allergic inflammation.
Collapse
Affiliation(s)
- Amin Khalifeh-Soltani
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Deepti Gupta
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Arnold Ha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Lung Biology Center, University of California, San Francisco, San Francisco, California, USA; and
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Ritwik Datta
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Lung Biology Center, University of California, San Francisco, San Francisco, California, USA; and
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Lacolley P, Regnault V, Segers P, Laurent S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol Rev 2017; 97:1555-1617. [DOI: 10.1152/physrev.00003.2017] [Citation(s) in RCA: 483] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
The cushioning function of large arteries encompasses distension during systole and recoil during diastole which transforms pulsatile flow into a steady flow in the microcirculation. Arterial stiffness, the inverse of distensibility, has been implicated in various etiologies of chronic common and monogenic cardiovascular diseases and is a major cause of morbidity and mortality globally. The first components that contribute to arterial stiffening are extracellular matrix (ECM) proteins that support the mechanical load, while the second important components are vascular smooth muscle cells (VSMCs), which not only regulate actomyosin interactions for contraction but mediate also mechanotransduction in cell-ECM homeostasis. Eventually, VSMC plasticity and signaling in both conductance and resistance arteries are highly relevant to the physiology of normal and early vascular aging. This review summarizes current concepts of central pressure and tensile pulsatile circumferential stress as key mechanical determinants of arterial wall remodeling, cell-ECM interactions depending mainly on the architecture of cytoskeletal proteins and focal adhesion, the large/small arteries cross-talk that gives rise to target organ damage, and inflammatory pathways leading to calcification or atherosclerosis. We further speculate on the contribution of cellular stiffness along the arterial tree to vascular wall stiffness. In addition, this review provides the latest advances in the identification of gene variants affecting arterial stiffening. Now that important hemodynamic and molecular mechanisms of arterial stiffness have been elucidated, and the complex interplay between ECM, cells, and sensors identified, further research should study their potential to halt or to reverse the development of arterial stiffness.
Collapse
Affiliation(s)
- Patrick Lacolley
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Véronique Regnault
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Patrick Segers
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Stéphane Laurent
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| |
Collapse
|
22
|
Herdl S, Huebner H, Volkert G, Marek I, Menendez-Castro C, Noegel SC, Ruebner M, Rascher W, Hartner A, Fahlbusch FB. Integrin α8 Is Abundant in Human, Rat, and Mouse Trophoblasts. Reprod Sci 2017; 24:1426-1437. [PMID: 28136130 DOI: 10.1177/1933719116689597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Integrins exert regulatory functions in placentogenesis. Null mutation of certain integrin α subunits leads to placental defects with subsequent fetal growth restriction or embryonic lethality in mice. So far, the placental role of α8 integrin remains to be determined. METHODS Localization of α8 integrin and its ligands, fibronectin (FN) and osteopontin (OPN), was studied by immunohistochemistry in human, rat, and mouse placenta. The vascularization of the placental labyrinth layer of α8 integrin-deficient mice was determined by CD31 staining. In humans, α8 integrin expression was assessed via real-time polymerase chain reaction in healthy placentas, in the placental pathologies such as intrauterine growth restriction (IUGR), preeclampsia, and HELLP-syndrome (hemolysis, elevated liver enzymes, low platelet count), as well as in primary extravillous trophoblasts (EVT) and villous trophoblasts. RESULTS In humans, α8 integrin was detected in first and third trimester syncytiotrophoblast and EVT. Although OPN showed the same localization, FN was observed in EVT only. No expressional changes in α8 integrin were detected in the placental pathologies studied. Rodent placenta showed α8 integrin expression in giant cells and in the labyrinth layer. The localization of OPN and FN, however, showed species-specific differences. Knockout of α8 integrin in mice did not cause IUGR, despite some reduction in labyrinth layer vascularization. CONCLUSION α8 Integrin is expressed in functional placental compartments among its ligands, OPN and/or FN, across species. Although this may point to a regulatory role in trophoblast function, our data from α8 integrin-deficient mice indicated only mild placental pathology. Thus, the lack of placental α8 integrin seems to be largely compensated for.
Collapse
Affiliation(s)
- Sebastian Herdl
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hanna Huebner
- 2 Department of Gynaecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gudrun Volkert
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ines Marek
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Carlos Menendez-Castro
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie C Noegel
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Ruebner
- 2 Department of Gynaecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Rascher
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Hartner
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian B Fahlbusch
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
23
|
Ran C, Mehdi RN, Fardell C, Xiang F, Nissbrandt H, Sydow O, Wirdefeldt K, Belin AC. No Association Between rs7077361 in ITGA8 and Parkinson's Disease in Sweden. Open Neurol J 2016; 10:25-9. [PMID: 27583043 PMCID: PMC4994103 DOI: 10.2174/1874205x01610010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/10/2016] [Accepted: 05/15/2016] [Indexed: 11/22/2022] Open
Abstract
Background: Integrin alpha 8 (ITGA8) encodes the alpha 8 subunit of the integrin alpha8beta1 protein and has recently been suggested as a new candidate gene for Parkinson’s disease, an age related neurodegenerative disease with unknown etiology. ITGA8 is a transmembrane protein involved in several cellular processes, such as cell adhesion, migration and cytoskeletal rearrangement. Objective: Screen a Swedish case control material for rs7077361, a genetic variant in ITGA8, in order to investigate its possible implication in Parkinson’s disease in Sweden. Method: Rs7077361 was genotyped using TaqMan quantitative Real-time PCR and tested for association using appropriate statistical methods. Results: We have screened 502 Swedish Parkinson patients and 599 healthy control individuals for rs7077361 in ITGA8. This genetic variant was in Hardy Weinberg equilibrium in the Swedish population. Allele and genotype frequencies were highly similar between the patients and controls and statistical testing showed that this genetic maker did not associate with Parkinson’s disease (p=0.67). Conclusion: Our results do not support the hypothesis of ITGA8 as a candidate gene for Parkinson’s disease in Sweden.
Collapse
Affiliation(s)
- Caroline Ran
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Camilla Fardell
- Department of Pharmacology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Fengqing Xiang
- Department of Women´s and Children´s Health, Karolinska Institutet, Stockholm, Sweden
| | - Hans Nissbrandt
- Department of Pharmacology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Olof Sydow
- Department of Neurology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Karin Wirdefeldt
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
24
|
Talbot JC, Nichols JT, Yan YL, Leonard IF, BreMiller RA, Amacher SL, Postlethwait JH, Kimmel CB. Pharyngeal morphogenesis requires fras1-itga8-dependent epithelial-mesenchymal interaction. Dev Biol 2016; 416:136-148. [PMID: 27265864 PMCID: PMC4967372 DOI: 10.1016/j.ydbio.2016.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/31/2016] [Indexed: 01/08/2023]
Abstract
Both Fras1 and Itga8 connect mesenchymal cells to epithelia by way of an extracellular 'Fraser protein complex' that functions in signaling and adhesion; these proteins are vital to the development of several vertebrate organs. We previously found that zebrafish fras1 mutants have craniofacial defects, specifically, shortened symplectic cartilages and cartilage fusions that spare joint elements. During a forward mutagenesis screen, we identified a new zebrafish mutation, b1161, that we show here disrupts itga8, as confirmed using CRISPR-generated itga8 alleles. fras1 and itga8 single mutants and double mutants have similar craniofacial phenotypes, a result expected if loss of either gene disrupts function of the Fraser protein complex. Unlike fras1 mutants or other Fraser-related mutants, itga8 mutants do not show blistered tail fins. Thus, the function of the Fraser complex differs in the craniofacial skeleton and the tail fin. Focusing on the face, we find that itga8 mutants consistently show defective outpocketing of a late-forming portion of the first pharyngeal pouch, and variably express skeletal defects, matching previously characterized fras1 mutant phenotypes. In itga8 and fras1 mutants, skeletal severity varies markedly between sides, indicating that both mutants have increased developmental instability. Whereas fras1 is expressed in epithelia, we show that itga8 is expressed complementarily in facial mesenchyme. Paired with the observed phenotypic similarity, this expression indicates that the genes function in epithelial-mesenchymal interactions. Similar interactions between Fras1 and Itga8 have previously been found in mouse kidney, where these genes both regulate Nephronectin (Npnt) protein abundance. We find that zebrafish facial tissues express both npnt and the Fraser gene fibrillin2b (fbn2b), but their transcript levels do not depend on fras1 or itga8 function. Using a revertible fras1 allele, we find that the critical window for fras1 function in the craniofacial skeleton is between 1.5 and 3 days post fertilization, which coincides with the onset of fras1-dependent and itga8-dependent morphogenesis. We propose a model wherein Fras1 and Itga8 interact during late pharyngeal pouch morphogenesis to sculpt pharyngeal arches through epithelial-mesenchymal interactions, thereby stabilizing the developing craniofacial skeleton.
Collapse
Affiliation(s)
- Jared Coffin Talbot
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA; Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| | - James T Nichols
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Yi-Lin Yan
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Isaac F Leonard
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Ruth A BreMiller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Sharon L Amacher
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | | | - Charles B Kimmel
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
25
|
Khalifeh-Soltani A, Ha A, Podolsky MJ, McCarthy DA, McKleroy W, Azary S, Sakuma S, Tharp KM, Wu N, Yokosaki Y, Hart D, Stahl A, Atabai K. α8β1 integrin regulates nutrient absorption through an Mfge8-PTEN dependent mechanism. eLife 2016; 5. [PMID: 27092791 PMCID: PMC4868538 DOI: 10.7554/elife.13063] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/18/2016] [Indexed: 12/25/2022] Open
Abstract
Coordinated gastrointestinal smooth muscle contraction is critical for proper nutrient absorption and is altered in a number of medical disorders. In this work, we demonstrate a critical role for the RGD-binding integrin α8β1 in promoting nutrient absorption through regulation of gastrointestinal motility. Smooth muscle-specific deletion and antibody blockade of α8 in mice result in enhanced gastric antral smooth muscle contraction, more rapid gastric emptying, and more rapid transit of food through the small intestine leading to malabsorption of dietary fats and carbohydrates as well as protection from weight gain in a diet-induced model of obesity. Mechanistically, ligation of α8β1 by the milk protein Mfge8 reduces antral smooth muscle contractile force by preventing RhoA activation through a PTEN-dependent mechanism. Collectively, our results identify a role for α8β1 in regulating gastrointestinal motility and identify α8 as a potential target for disorders characterized by hypo- or hyper-motility. DOI:http://dx.doi.org/10.7554/eLife.13063.001 Animals absorb nutrients from the food they eat in a complicated process that involves multiple steps. In the mouth, teeth break down the food into smaller chunks. Then the food passes through the stomach and small intestine, where enzymes break it down into individual molecules that are small enough to be absorbed by cells that line the small intestine. These cells then package the molecules and release them into the bloodstream so that they can be distributed to the rest of the body. Muscles in the wall of the small intestine control how quickly food travels through this part of the gut. If food moves too quickly, the cells that line the intestine have less time to absorb the food molecules and may fail to absorb enough nutrients. If the food moves too slowly, an individual may experience nausea or vomiting, or the contents of their stomach may spill into their lungs. In 2014, researchers reported that a protein in breast milk called Mfge8 helps to boost the number of fat molecules absorbed from food. Now, Khalifeh-Soltani et al. – including some of the same researchers involved in the earlier work – show that Mfge8 also slows the rate at which food travels through the small intestine in mice. Mfge8 binds to another protein called integrin α8β1 to control how often the intestine muscles contract. Genetically engineered mice that lacked integrin α8β1 developed diarrhea and food passed through their intestines more quickly than in normal mice. Furthermore, these mice did not gain as much weight as normal mice when they were fed a high-fat diet. Khalifeh-Soltani et al.’s findings show that Mfge8 has a dual role in controlling the absorption of food molecules in the small intestine. The next challenge is to find out whether drugs that alter the activity of integrin α8β1 could be used to help treat patients with diseases in which food moves too quickly, or too slowly, through the gut. DOI:http://dx.doi.org/10.7554/eLife.13063.002
Collapse
Affiliation(s)
- Amin Khalifeh-Soltani
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Arnold Ha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Donald A McCarthy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - William McKleroy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Saeedeh Azary
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Stephen Sakuma
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Kevin M Tharp
- Metabolic Biology, University of California, Berkeley, Berkeley, United States.,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, United States
| | - Nanyan Wu
- Lung Biology Center, University of California, San Francisco, San Francisco, United States
| | - Yasuyuki Yokosaki
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Health Administration Center, Hiroshima University, Hiroshima, Japan
| | - Daniel Hart
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Andreas Stahl
- Metabolic Biology, University of California, Berkeley, Berkeley, United States.,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, United States
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States.,Lung Biology Center, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
26
|
Epitopes in α8β1 and other RGD-binding integrins delineate classes of integrin-blocking antibodies and major binding loops in α subunits. Sci Rep 2015; 5:13756. [PMID: 26349930 PMCID: PMC4563375 DOI: 10.1038/srep13756] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/04/2015] [Indexed: 12/28/2022] Open
Abstract
Identification of epitopes for integrin-blocking monoclonal antibodies (mAbs) has aided our understanding of structure-function relationship of integrins. We mapped epitopes of chicken anti-integrin-α8-subunit-blocking mAbs by mutational analyses, examining regions that harboured all mapped epitopes recognized by mAbs against other α-subunits in the RGD-binding-integrin subfamily. Six mAbs exhibited blocking function, and these mAbs recognized residues on the same W2:41-loop on the top-face of the β-propeller. Loop-tips sufficiently close to W2:41 (<25 Å) contained within a footprint of the mAbs were mutated, and the loop W3:34 on the bottom face was identified as an additional component of the epitope of one antibody, clone YZ5. Binding sequences on the two loops were conserved in virtually all mammals, and that on W3:34 was also conserved in chickens. These indicate 1) YZ5 binds both top and bottom loops, and the binding to W3:34 is by interactions to conserved residues between immunogen and host species, 2) five other blocking mAbs solely bind to W2:41 and 3) the α8 mAbs would cross-react with most mammals. Comparing with the mAbs against the other α-subunits of RGD-integrins, two classes were delineated; those binding to "W3:34 and an top-loop", and "solely W2:41", accounting for 82% of published RGD-integrin-mAbs.
Collapse
|
27
|
Marek I, Volkert G, Hilgers KF, Bieritz B, Rascher W, Reinhardt DP, Hartner A. Fibrillin-1 and alpha8 integrin are co-expressed in the glomerulus and interact to convey adhesion of mesangial cells. Cell Adh Migr 2015; 8:389-95. [PMID: 25482639 DOI: 10.4161/cam.28988] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Fibrillin-1 is a microfibrillar extracellular matrix protein that was described to be a ligand for α8 integrin. α8 integrin is a matrix receptor specifically expressed in mesangial and smooth muscle cells of the kidney. In previous studies we detected glomerular expression of fibrillin-1. Moreover, fibrillin-1 promoted adhesion, migration, and proliferation of mesangial cells. We hypothesized that fibrillin-1 and α8 integrin might interact in the glomerulus, and thus, regulate mesangial cell properties. Our studies showed that fibrillin-1 and α8 integrin colocalize in the glomerular mesangium. Induction of experimental glomerulonephritis led to an increase of both fibrillin-1 and α8 integrin expression. In vitro studies revealed that mesangial cells deficient for α8 integrin adhere weaker to fibrillin-1 and migrate more easily on fibrillin-1 than wild-type mesangial cells. Baseline proliferation on fibrillin-1 is higher in α8 integrin-deficient mesangial cells, but the induction of proliferation is not different in α8 integrin-deficient and wild-type mesangial cells. We conclude that fibrillin-1 and α8 integrin interact, and thus, regulate mesangial cell adhesion and migration. The concomitant induction of both fibrillin-1 and α8 integrin in a self-limited model of glomerular injury points to a protective role of the interaction of fibrillin-1 with α8 integrin in the glomerulus resulting in reduced damage of the glomerular tuft as a consequence of firm adhesion of mesangial cells.
Collapse
Affiliation(s)
- Ines Marek
- a Department for Pediatrics and Adolescent Medicine ; University Hospital of Erlangen ; Erlangen , Germany
| | | | | | | | | | | | | |
Collapse
|
28
|
Menendez-Castro C, Cordasic N, Neureiter D, Amann K, Marek I, Volkert G, Stintzing S, Jahn A, Rascher W, Hilgers KF, Hartner A. Under-expression of α8 integrin aggravates experimental atherosclerosis. J Pathol 2015; 236:5-16. [PMID: 25511181 DOI: 10.1002/path.4501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/28/2014] [Accepted: 12/09/2014] [Indexed: 11/08/2022]
Abstract
Integrins play an important role in vascular biology. The α8 integrin chain attenuates smooth muscle cell migration but its functional role in the development of atherosclerosis is unclear. Therefore, we studied the contribution of α8 integrin to atherosclerosis and vascular remodelling. We hypothesized that α8 integrin expression is reduced in atherosclerotic lesions, and that its under-expression leads to a more severe course of atherosclerosis. α8 Integrin was detected by immunohistochemistry and qPCR and α8 integrin-deficient mice were used to induce two models of atherosclerotic lesions. First, ligation of the carotid artery led to medial thickening and neointima formation, which was quantified in carotid cross-sections. Second, after crossing into ApoE-deficient mice, the formation of advanced vascular lesions with atherosclerotic plaques was quantified in aortic en face preparations stained with Sudan IV. Parameters of renal physiology and histopathology were assessed: α8 integrin was detected in the media of human and murine vascular tissue and was down-regulated in arteries with advanced atherosclerotic lesions. In α8 integrin-deficient mice (α8(-/-) ) as well as α8(+/-) and α8(+/+) littermates, carotid artery ligation increased media:lumen ratios in all genotypes, with higher values in ligated α8(-/-) and α8(+/-) compared to ligated α8(+/+) animals. Carotid artery ligation increased smooth muscle cell number in the media of α8(+/+) mice and, more prominently, of α8(-/-) or α8(+/-) mice. On an ApoE(-/-) background, α8(+/-) and α8(-/-) mice developed more atherosclerotic plaques than α8(+/+) mice. α8 Integrin expression was reduced in α8(+/-) animals. Renal damage with increased serum creatinine and glomerulosclerosis was detected in α8(-/-) mice only. Thus, under-expression of α8 integrin aggravates vascular lesions, while a complete loss of α8 integrin results in reduced renal mass and additional renal disease in the presence of generalized atherosclerosis. Our data support the hypothesis that integrin α8β1 has a protective role in arterial remodelling and atherosclerosis.
Collapse
Affiliation(s)
- Carlos Menendez-Castro
- Department of Paediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Akison LK, Boden MJ, Kennaway DJ, Russell DL, Robker RL. Progesterone receptor-dependent regulation of genes in the oviducts of female mice. Physiol Genomics 2014; 46:583-92. [DOI: 10.1152/physiolgenomics.00044.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Oviducts play a critical role in gamete and embryo transport, as well as supporting early embryo development. Progesterone receptor (PGR) is a transcription factor highly expressed in oviductal cells, while its activating ligand, progesterone, surges to peak levels as ovulation approaches. Progesterone is known to regulate oviduct cilia beating and muscular contractions in vitro, but how PGR may mediate this in vivo is poorly understood. We used PGR null mice to identify genes potentially regulated by PGR in the oviducts during the periovulatory period. Histologically, oviducts from PGR null mice showed no gross structural or morphological defects compared with normal littermates. However, microarray analysis of oviducts at 8 h posthuman chorionic gonadotropin revealed >1,000 PGR-dependent genes. Using reverse-transcription polymerase chain reaction (RT-PCR) we selected 10 genes for validation based on their potential roles in oocyte/embryo transport and support. Eight genes were confirmed to be downregulated ( Adamts1, Itga8, Edn3, Prlr, Ptgfr, Des, Myocd, and Actg2) and one upregulated ( Agtr2) in PGR null oviducts. Expression of these genes was also assessed in oviducts of naturally cycling mice during ovulation and day 1 and day 4 of pregnancy. Adamts1, Itga8, Edn3, Prlr, and Ptgfr were significantly upregulated in oviducts at ovulation/mating. However, most genes showed basal levels of expression at other times. The exceptions were Prlr and Ptgfr, which showed pulsatile increases on day 1 and/or day 4 of pregnancy. This is the first, comprehensive study to elucidate putative PGR-regulated genes in the oviduct and reveals key downstream targets potentially mediating oocyte and embryo transport.
Collapse
Affiliation(s)
- Lisa K. Akison
- The Robinson Research Institute, School of Paediatrics & Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Michael J. Boden
- The Robinson Research Institute, School of Paediatrics & Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| | - David J. Kennaway
- The Robinson Research Institute, School of Paediatrics & Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Darryl L. Russell
- The Robinson Research Institute, School of Paediatrics & Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Rebecca L. Robker
- The Robinson Research Institute, School of Paediatrics & Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
30
|
Volkert G, Jahn A, Dinkel C, Fahlbusch F, Zürn C, Hilgers KF, Rascher W, Hartner A, Marek I. Contribution of the α8 Integrin Chain to the Expression of Extracellular Matrix Components. ACTA ACUST UNITED AC 2014; 21:89-98. [DOI: 10.3109/15419061.2013.876012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Zallocchi M, Johnson BM, Meehan DT, Delimont D, Cosgrove D. α1β1 integrin/Rac1-dependent mesangial invasion of glomerular capillaries in Alport syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1269-1280. [PMID: 23911822 DOI: 10.1016/j.ajpath.2013.06.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 06/11/2013] [Accepted: 06/14/2013] [Indexed: 01/27/2023]
Abstract
Alport syndrome, hereditary glomerulonephritis with hearing loss, results from mutations in type IV collagen COL4A3, COL4A4, or COL4A5 genes. The mechanism for delayed glomerular disease onset is unknown. Comparative analysis of Alport mice and CD151 knockout mice revealed progressive accumulation of laminin 211 in the glomerular basement membrane. We show mesangial processes invading the capillary loops of both models as well as in human Alport glomeruli, as the likely source of this laminin. L-NAME salt-induced hypertension accelerated mesangial cell process invasion. Cultured mesangial cells showed reduced migratory potential when treated with either integrin-linked kinase inhibitor or Rac1 inhibitor, or by deletion of integrin α1. Treatment of Alport mice with Rac1 inhibitor or deletion of integrin α1 reduced mesangial cell process invasion of the glomerular capillary tuft. Laminin α2-deficient Alport mice show reduced mesangial process invasion, and cultured laminin α2-null cells showed reduced migratory potential, indicating a functional role for mesangial laminins in progression of Alport glomerular pathogenesis. Collectively, these findings predict a role for biomechanical insult in the induction of integrin α1β1-dependent Rac1-mediated mesangial cell process invasion of the glomerular capillary tuft as an initiation mechanism of Alport glomerular pathology.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Department of Genetics, Boys Town National Research Hospital, Omaha, Nebraska
| | - Brianna M Johnson
- Department of Genetics, Boys Town National Research Hospital, Omaha, Nebraska
| | - Daniel T Meehan
- Department of Genetics, Boys Town National Research Hospital, Omaha, Nebraska
| | - Duane Delimont
- Department of Genetics, Boys Town National Research Hospital, Omaha, Nebraska
| | - Dominic Cosgrove
- Department of Genetics, Boys Town National Research Hospital, Omaha, Nebraska; Department of Biochemistry, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
32
|
Abstract
Vascular smooth muscle cells have attracted considerable interest as a model for a flexible program of gene expression. This cell type arises throughout the embryo body plan via poorly understood signaling cascades that direct the expression of transcription factors and microRNAs which, in turn, orchestrate the activation of contractile genes collectively defining this cell lineage. The discovery of myocardin and its close association with serum response factor has represented a major break-through for the molecular understanding of vascular smooth muscle cell differentiation. Retinoids have been shown to improve the outcome of vessel wall remodeling following injury and have provided further insights into the molecular circuitry that defines the vascular smooth muscle cell phenotype. This review summarizes the progress to date in each of these areas of vascular smooth muscle cell biology.
Collapse
|
33
|
Hartner A, Menendez-Castro C, Cordasic N, Marek I, Volkert G, Klanke B, Rascher W, Hilgers KF. Tubulointerstitial de novo expression of the α8 integrin chain in a rodent model of renal fibrosis--a potential target for anti-fibrotic therapy? PLoS One 2012; 7:e48362. [PMID: 23144868 PMCID: PMC3493553 DOI: 10.1371/journal.pone.0048362] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/24/2012] [Indexed: 11/24/2022] Open
Abstract
In the normal kidney, the α8 integrin chain is expressed only on mesangial cells and vascular smooth muscle cells. α8 integrin ligates several matrix molecules including fibronectin, osteopontin and fibrillin-1. Recently, we detected de novo expression of α8 integrin on epithelial cells in renal cysts. We hypothesized that the α8 integrin chain is induced in tubular epithelia undergoing dedifferentiation and contributes to the fibrotic response in the tubulointerstitium (TI) after unilateral ureteral obstruction (UUO). After induction of UUO in rats by ligation of the right ureter, increased expression of the α8 integrin chain and its ligands was observed. In the TI, α8 integrin was localized to cytokeratin-positive epithelial cells and to interstitial fibroblasts; and colocalized with its ligands. In mice underexpressing α8 integrin UUO led to collagen deposition and fibroblast activation comparable to wild types. Mice lacking α8 integrin showed even more TI damage, fibroblast activation and collagen deposition after UUO compared to wild type mice. We conclude that the expression of the α8 integrin chain and its ligands is strongly induced in the TI after UUO, but underexpression of α8 integrin does not attenuate TI fibrosis. Mice lacking the α8 integrin chain are even more susceptible to TI damage than wild type mice. Thus, interactions of α8 integrin with its ligands do not seem to contribute to the development or progression of TI fibrosis in UUO. Targeting α8 integrin might not be a useful approach for anti-fibrotic therapy.
Collapse
Affiliation(s)
- Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Expression and promoter analysis of a highly restricted integrin alpha gene in vascular smooth muscle. Gene 2012; 513:82-9. [PMID: 23142384 DOI: 10.1016/j.gene.2012.10.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 09/18/2012] [Accepted: 10/29/2012] [Indexed: 11/19/2022]
Abstract
Full genome annotation requires gene expression analysis and elucidation of promoter activity. Here, we analyzed the expression and promoter of a highly restricted integrin gene, Itga8. RNase protection and quantitative RT-PCR showed Itga8 to be expressed most abundantly in vascular smooth muscle cells (SMC). Transcription start site mapping of Itga8 revealed the immediate 5' promoter region to be poorly conserved with orthologous sequences in the human genome. Further comparative sequence analysis showed a number of conserved non-coding sequence modules around the Itga8 gene. The immediate promoter region and an upstream conserved sequence module were each found to contain a CArG box, which is a binding site for serum response factor (SRF). Luciferase reporter assays revealed activity of several Itga8 promoter constructs with no apparent restricted activity to SMC types. Further, neither SRF nor its coactivator, Myocardin (MYOCD), was able to induce several distinct Itga8 promoter constructs. Transgenic mouse studies failed to reveal Itga8 promoter activity, indicating distal regulatory elements likely control this gene's in vivo expression profile. Interestingly, although the promoter was unresponsive to SRF/MYOCD, the endogenous Itga8 gene showed increases in expression upon ectopic MYOCD expression even though knockdown of SRF both in vitro and in vivo failed to demonstrate a corresponding change in Itga8. Collectively, these data demonstrate that Itga8 expression is CArG-SRF independent, but MYOCD dependent through an as yet unknown sequence module that is distal from the promoter region.
Collapse
|
35
|
RGD-Dependent Epithelial Cell-Matrix Interactions in the Human Intestinal Crypt. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:248759. [PMID: 22988499 PMCID: PMC3440950 DOI: 10.1155/2012/248759] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 07/05/2012] [Accepted: 07/22/2012] [Indexed: 01/09/2023]
Abstract
Interactions between the extracellular matrix (ECM) and integrin receptors trigger structural and functional bonds between the cell microenvironment and the cytoskeleton. Such connections are essential for adhesion structure integrity and are key players in regulating transduction of specific intracellular signals, which in turn regulate the organization of the cell microenvironment and, consequently, cell function. The RGD peptide-dependent integrins represent a key subgroup of ECM receptors involved in the maintenance of epithelial homeostasis. Here we review recent findings on RGD-dependent ECM-integrin interactions and their roles in human intestinal epithelial crypt cells.
Collapse
|
36
|
Sánchez-Cortés J, Mrksich M. Using self-assembled monolayers to understand α8β1-mediated cell adhesion to RGD and FEI motifs in nephronectin. ACS Chem Biol 2011; 6:1078-86. [PMID: 21790180 PMCID: PMC3200005 DOI: 10.1021/cb200186j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nephronectin is an extracellular matrix protein that interacts with the α8β1 integrin receptor and plays a role in tissue and organ development, though the motifs that mediate adhesion to the receptor remain unclear. This paper describes the use of self-assembled monolayers to study the adhesion of α8β1-presenting cells to the RGD and DLFEIFEIER ligands in nephronectin and found that both ligands can independently mediate cell adhesion through nonoverlapping binding sites on the integrin. Peptide truncation experiments showed FEI to be the minimal binding sequence within the DLFEIFEIER sequence, and adhesion experiments with peptides that include both the RGD and FEI sequences demonstrate that the two peptides bind synergistically to the receptor. Finally, a peptide array was used to establish a strict requirement for the glutamate residue of FEI and tolerance of other aromatic and hydrophobic residues in the first and third positions, respectively. This work provides an enhanced understanding of the binding of nephronectin with α8β1 and identifies a peptide ligand that can be used for targeting the α8β1 integrin.
Collapse
Affiliation(s)
- Juan Sánchez-Cortés
- Department of Chemistry and Howard Hughes Medical Institute, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Milan Mrksich
- Department of Chemistry and Howard Hughes Medical Institute, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| |
Collapse
|
37
|
Bagavant H, Kalantarinia K, Scindia Y, Deshmukh U. Novel therapeutic approaches to lupus glomerulonephritis: translating animal models to clinical practice. Am J Kidney Dis 2011; 57:498-507. [PMID: 21239097 PMCID: PMC3042497 DOI: 10.1053/j.ajkd.2010.10.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 10/11/2010] [Indexed: 11/11/2022]
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease frequently affecting the kidney. Renal involvement is characterized by glomerular immune complex deposits and proliferative glomerulonephritis progressing to glomerulosclerosis and kidney failure. The development of systemic lupus erythematosus is regulated genetically, and lupus susceptibility genes have been linked to immune hyper-responsiveness and loss of immune regulation. In addition to the systemic immune defects, recent studies in animal models show that susceptibility to lupus nephritis is influenced by intrinsic renal factors. Thus, renal cell responses to immune-mediated glomerular injury determine disease outcome. This supports the idea that future treatments for lupus nephritis need to focus on regulating end-organ responses. The feasibility of this approach has been shown in animal models of kidney disease. For more than 50 years, the emphasis in management of lupus nephritis has been suppression of autoimmune responses and systemic control of inflammation. This review describes recently developed targeted drug delivery technologies and potential targets that can regulate glomerular cell responses, offering a novel therapeutic approach for lupus nephritis.
Collapse
Affiliation(s)
- Harini Bagavant
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | |
Collapse
|
38
|
Marek I, Volkert G, Jahn A, Fahlbusch F, Zürn C, Ozcan Z, Goppelt-Struebe M, Hilgers KF, Rascher W, Hartner A. Lack of α8 integrin leads to morphological changes in renal mesangial cells, but not in vascular smooth muscle cells. BMC Cell Biol 2010; 11:102. [PMID: 21194485 PMCID: PMC3022721 DOI: 10.1186/1471-2121-11-102] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 12/31/2010] [Indexed: 11/21/2022] Open
Abstract
Background Extracellular matrix receptors of the integrin family are known to regulate cell adhesion, shape and functions. The α8 integrin chain is expressed in glomerular mesangial cells and in vascular smooth muscle cells. Mice deficient for α8 integrin have structural alterations in glomeruli but not in renal arteries. For this reason we hypothesized that mesangial cells and vascular smooth muscle cells differ in their respective capacity to compensate for the lack of α8 integrin. Results Wild type and α8 integrin-deficient mesangial cells varied markedly in cell morphology and expression or localization of cytoskeletal molecules. In α8 integrin-deficient mesangial cells α-smooth muscle actin and CTGF were downregulated. In contrast, there were no comparable differences between α8 integrin-deficient and wild type vascular smooth muscle cells. Expression patterns of integrins were altered in α8 integrin-deficient mesangial cells compared to wild type mesangial cells, displaying a prominent overexpression of α2 and α6 integrins, while expression patterns of the these integrins were not different between wild type and α8 integrin-deficient vascular smooth muscle cells, respectively. Cell proliferation was augmented in α8 integrin-deficient mesangial cells, but not in vascular smooth muscle cells, compared to wild type cells. Conclusions Our findings suggest that α8 integrin deficiency has differential effects in mesangial cells and vascular smooth muscle cells. While the phenotype of vascular smooth muscle cells lacking α8 integrin is not altered, mesangial cells lacking α8 integrin differ considerably from wild type mesangial cells which might be a consequence of compensatory changes in the expression patterns of other integrins. This could result in glomerular changes in α8 integrin-deficient mice, while the vasculature is not affected in these mice.
Collapse
Affiliation(s)
- Ines Marek
- Hospital for Children and Adolescents, Universität Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hartner A, Cordasic N, Menendez-Castro C, Volkert G, Yabu JM, Kupraszewicz-Hutzler M, Rascher W, Hilgers KF. Lack of {alpha}8-integrin aggravates podocyte injury in experimental diabetic nephropathy. Am J Physiol Renal Physiol 2010; 299:F1151-7. [PMID: 20826576 DOI: 10.1152/ajprenal.00058.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Development of diabetic nephropathy is accompanied by changes in integrin-mediated cell-matrix interactions. The α8-integrin chain is specifically expressed in mesangial cells of the glomerulus. During experimental hypertension, α8-integrin plays a protective role in the glomerulus. We hypothesized that α8-integrin is involved in maintaining the integrity of the glomerulus in diabetic nephropathy. Experimental streptozotocin (STZ) diabetes led to an increased expression and glomerular deposition of α8-integrin. To test the functional role of α8-integrin, STZ diabetes was induced in mice with a homozygous (α8-/-) or heterozygous (α8+/-) deletion of the α8-integrin gene and in wild-type litters (α8+/+). Blood glucose and mean arterial blood pressure were not different in α8-/- and α8+/+ mice after 6 wk of diabetes. However, diabetic α8-/- mice developed significantly higher albuminuria and more glomerulosclerosis than diabetic α8+/+ mice. Moreover, in diabetic α8-/- mice, the number of glomerular cells staining positive for the podocyte markers WT-1 and vimentin were reduced more prominently than in diabetic α8+/+. The filtration barrier protein nephrin was downregulated in diabetic glomeruli with the strongest reduction observed in α8-/- mice. Taken together, α8-/- mice developed more severe glomerular lesions and podocyte damage after onset of STZ diabetes than α8+/+ mice, indicating that α8-integrin is protective for the structure and function of the glomerulus and maintains podocyte integrity during the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Andrea Hartner
- Dept. of Pediatrics, Loschgestrasse 15, D-91054 Erlangen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Benjamin JT, Gaston DC, Halloran BA, Schnapp LM, Zent R, Prince LS. The role of integrin alpha8beta1 in fetal lung morphogenesis and injury. Dev Biol 2009; 335:407-17. [PMID: 19769957 DOI: 10.1016/j.ydbio.2009.09.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 09/11/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
Abstract
Prenatal inflammation prevents normal lung morphogenesis and leads to bronchopulmonary dysplasia (BPD), a common complication of preterm birth. We previously demonstrated in a bacterial endotoxin mouse model of BPD that disrupting fibronectin localization in the fetal lung mesenchyme causes arrested saccular airway branching. In this study we show that expression of the fibronectin receptor, integrin alpha8beta1 is decreased in the lung mesenchyme in the same inflammation model suggesting it is required for normal lung development. We verified a role for integrin alpha8beta1 in lung development using integrin alpha8-null mice, which develop fusion of the medial and caudal lobes as well as abnormalities in airway division. We further show in vivo and in vitro that alpha8-null fetal lung mesenchymal cells fail to form stable adhesions and have increased migration. Thus we propose that integrin alpha8beta1 plays a critical role in lung morphogenesis by regulating mesenchymal cell adhesion and migration. Furthermore, our data suggest that disruption of the interactions between extracellular matrix and integrin alpha8beta1 may contribute to the pathogenesis of BPD.
Collapse
Affiliation(s)
- John T Benjamin
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232-0493, USA
| | | | | | | | | | | |
Collapse
|
41
|
Integrin alpha8beta1 regulates adhesion, migration and proliferation of human intestinal crypt cells via a predominant RhoA/ROCK-dependent mechanism. Biol Cell 2009; 101:695-708. [PMID: 19527220 PMCID: PMC2782361 DOI: 10.1042/bc20090060] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background. Integrins are transmembrane αβ heterodimer receptors that function as structural and functional bridges between the cytoskeleton and ECM (extracellular matrix) molecules. The RGD (arginine-glycine-aspartate tripeptide motif)-dependent integrin α8β1 has been shown to be involved in various cell functions in neuronal and mesenchymal-derived cell types. Its role in epithelial cells remains unknown. Results. Integrin α8β1 was found to be expressed in the crypt cell population of the human intestine but was absent from differentiating and mature epithelial cells of the villus. The function of α8β1 in epithelial crypt cells was investigated at the cellular level using normal HIECs (human intestinal epithelial cells). Specific knockdown of α8 subunit expression using an shRNA (small-hairpin RNA) approach showed that α8β1 plays important roles in RGD-dependent cell adhesion, migration and proliferation via a RhoA/ROCK (Rho-associated kinase)-dependent mechanism as demonstrated by active RhoA quantification and pharmacological inhibition of ROCK. Moreover, loss of α8β1, through RhoA/ROCK, impairs FA (focal adhesion) complex integrity as demonstrated by faulty vinculin recruitment. Conclusions. Integrin α8β1 is expressed in epithelial cells. In intestinal crypt cells, α8β1 is closely involved in the regulation of adhesion, migration and cell proliferation via a predominant RhoA/ROCK-dependent mechanism. These results suggest an important role for this integrin in intestinal crypt cell homoeostasis.
Collapse
|
42
|
Hartner A, Cordasic N, Rascher W, Hilgers KF. Deletion of the alpha8 integrin gene does not protect mice from myocardial fibrosis in DOCA hypertension. Am J Hypertens 2009; 22:92-9. [PMID: 19008864 DOI: 10.1038/ajh.2008.309] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND In the heart, the alpha8 integrin chain is expressed in fibroblasts and vascular smooth-muscle cells but its functional role in the myocardium is unknown. Integrins can contribute to tissue fibrosis in several organs. We tested the hypothesis that alpha8 integrin-mediated cell-matrix interactions add to cardiac fibrotic alterations during hypertension. METHODS Desoxycorticosterone-acetate (DOCA)-salt hypertension was induced in mice homozygous for a deletion of the alpha8 integrin chain and wild-type mice. Histological and immunohistochemical evaluations were performed in heart tissue. RESULTS Blood pressure was slightly higher in DOCA-treated alpha8 integrin-deficient mice compared to DOCA-treated wild types. Expression of alpha8 integrin and its ligands fibronectin and osteopontin was increased in the hearts of DOCA-treated wild types compared to salt-loaded controls. However, relative left ventricular weights did not differ between DOCA-treated wild types and alpha8 integrin-deficient mice. Moreover, expansion of collagen I immunoreactivity and cell proliferation was similar in both groups. The number of osteopontin-positive cells was not different in DOCA-treated alpha8 integrin-deficient and DOCA-treated wild-type mice. Despite of a comparable degree of fibrosis in both groups, alpha-smooth-muscle actin and discoidin domain receptor 2 (DDR2)-positive myofibroblasts were only detected in wild-type DOCA-treated mice, not in DOCA-treated alpha8 integrin-deficient mice. CONCLUSIONS The results show that lack of alpha8 integrin does not reduce fibrotic changes in the hearts of DOCA-salt hypertensive mice. Our findings do not argue for a profibrotic effect of an increased alpha8 integrin expression in the myocardium in hypertension.
Collapse
|
43
|
Scindia Y, Deshmukh U, Thimmalapura PR, Bagavant H. Anti-alpha8 integrin immunoliposomes in glomeruli of lupus-susceptible mice: a novel system for delivery of therapeutic agents to the renal glomerulus in systemic lupus erythematosus. ARTHRITIS AND RHEUMATISM 2008; 58:3884-91. [PMID: 19035491 PMCID: PMC2678908 DOI: 10.1002/art.24026] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Glomerular mesangial cells are active participants in the pathogenesis of lupus glomerulonephritis (GN). Thus, targeted delivery of therapeutic agents to mesangial cells would be an attractive approach to treatment. However, lack of known unique mesangial cell surface markers has hampered this process. This study was undertaken in a mouse model of lupus GN to identify mesangial markers and to develop a system for targeted drug delivery to the glomerulus. METHODS Based on previous observations, alpha8 integrin expressed on the surface of glomerular mesangial cells was selected as a target molecule for delivery. Two mouse strains susceptible to lupus GN, NZM2328 and (NZM2328 x NOD)F1, were studied. Glomerular expression of alpha8 integrin in normal and nephritic mice was confirmed by immunofluorescence and quantitative polymerase chain reaction analysis. Liposomes were formulated and conjugated with an anti-alpha8 integrin antibody. These immunoliposomes were loaded with DiI, a red fluorescent dye, to allow tracking in vivo, and injected into the tail vein of female mice at different ages. Specificity of targeting was studied by fluorescence microscopy and flow cytometry. RESULTS Expression of alpha8 integrin was observed in the glomeruli of normal and nephritic mice. Anti-alpha8 integrin immunoliposomes were detected in the glomerulus and glomerular mesangial cells after tail vein injection in normal and nephritic mice. Delivery of DiI by anti-alpha8 integrin immunoliposomes was tissue specific, being observed predominantly in the glomeruli, with some nonspecific uptake by CD11b cells. CONCLUSION These findings are the first demonstration of specific delivery of anti-alpha8 integrin immunoliposomes to the mesangium following tail vein injection in mice. Anti-alpha8 integrin immunoliposomes thus offer a novel approach for targeted drug therapy in lupus and other glomerular diseases.
Collapse
Affiliation(s)
- Yogesh Scindia
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908
| | - Umesh Deshmukh
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908
| | - Puspha-Rekha Thimmalapura
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908
| | - Harini Bagavant
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
44
|
Lu X, Lu D, Scully M, Kakkar V. The Role of Integrins in Cancer and the Development of Anti-Integrin Therapeutic Agents for Cancer Therapy. PERSPECTIVES IN MEDICINAL CHEMISTRY 2008. [DOI: 10.1177/1177391x0800200003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Integrins have been reported to mediate cell survival, proliferation, differentiation, and migration programs. For this reason, the past few years have seen an increased interest in the implications of integrin receptors in cancer biology and tumor cell aggression. This review considers the potential role of integrins in cancer and also addresses why integrins are present attractive targets for drug design. It discusses of the several properties of the integrin-based chemotherapeutic agents currently under consideration clinically and provides an insight into cancer drug development using integrin as a target.
Collapse
Affiliation(s)
- Xinjie Lu
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR U.K
| | - Dong Lu
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, U.K
| | - Mike Scully
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR U.K
| | - Vijay Kakkar
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR U.K
| |
Collapse
|
45
|
Hartner A, Marek I, Cordasic N, Haas C, Schocklmann H, Hulsmann-Volkert G, Plasa I, Rascher W, Hilgers KF, Amann K. Glomerular regeneration is delayed in nephritic alpha 8-integrin-deficient mice: contribution of alpha 8-integrin to the regulation of mesangial cell apoptosis. Am J Nephrol 2007; 28:168-78. [PMID: 17951999 DOI: 10.1159/000110022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Accepted: 08/29/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS alpha(8)beta(1)-Integrin is expressed in mesangial cells. In vitro studies suggest a role for alpha(8)-integrin in the regulation of cell proliferation and apoptosis. We tested the hypothesis that alpha(8)-integrin is essential for the healing process after mesangioproliferative glomerulonephritis. METHODS Mice homozygous for a deletion of the alpha(8)-integrin chain were compared with wild-type mice. To study glomerular healing, we used the habu toxin model of reversible mesangioproliferative glomerulonephritis. Animals received 6 mg/kg habu toxin intravenously; controls received saline only. RESULTS Early mesangiolysis occurred in wild-type and alpha(8)-integrin-deficient mice. However, mesangiolysis was no longer detectable after 7 days in wild types but persisted after 14 days in alpha(8)-integrin-deficient animals. Mesangial activation marker alpha-smooth muscle actin was detectable only at day 7 in wild-type mice but persisted until day 14 in alpha(8)-integrin-deficient mice. In wild types, glomerular cell proliferation and apoptosis peaked at day 7 and decreased thereafter but remained elevated in alpha(8)-integrin-deficient mice until day 28. In cultivated mesangial cells, alpha(8)-integrin expression was associated with increased cell survival. CONCLUSION Interactions between alpha(8)-integrin and the mesangial matrix may contribute to healing of glomerular injury by influencing cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Andrea Hartner
- Hospital for Children and Adolescents, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zargham R, Wamhoff BR, Thibault G. RNA interference targeting alpha8 integrin attenuates smooth muscle cell growth. FEBS Lett 2007; 581:939-43. [PMID: 17300786 DOI: 10.1016/j.febslet.2007.01.069] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 01/26/2007] [Indexed: 01/10/2023]
Abstract
alpha8 integrin gene silencing has been shown to result in the stress fibre disassembly. Stress fibres are required for cell adhesion to promote passage through cell cycle. Thus, we hypothesized that alpha8 integrin gene silencing might affect vascular smooth muscle cell (VSMC) growth. Short interference RNA (siRNA) targeting alpha8 integrin in rat VSMCs resulted in reduced DNA synthesis. Moreover, siRNA-alpha8 integrin prevented thrombin-induced proliferation. RhoA plays a critical role in regulating VSMC growth. alpha8 integrin co-immunoprecipitated with RhoA and siRNA-alpha8 reduced membrane associated RhoA. Our data suggest that alpha8 integrin expression is critical for VSMC growth, which has potential implications in postangioplasty neointimal hyperplasia.
Collapse
Affiliation(s)
- Ramin Zargham
- Institut de Recherches Cliniques de Montréal, Université de Montréal and Experimental Medicine Department, McGill University, 110 Avenue Des Pins Ouest, Montréal, Que., Canada.
| | | | | |
Collapse
|
47
|
Umesh A, Thompson MA, Chini EN, Yip KP, Sham JSK. Integrin ligands mobilize Ca2+ from ryanodine receptor-gated stores and lysosome-related acidic organelles in pulmonary arterial smooth muscle cells. J Biol Chem 2006; 281:34312-23. [PMID: 16963791 DOI: 10.1074/jbc.m606765200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Extracellular matrix (ECM) protein receptors, or integrins, participate in vascular remodeling and the systemic myogenic response. Synthetic ligands and ECM fragments regulate the vascular smooth muscle cell contractile state by altering intracellular Ca2+ levels ([Ca2+]i). Information on the Ca2+ effect of integrins in vascular smooth muscle cells is limited, but nonexistent in pulmonary arterial smooth muscle cells (PASMCs). We therefore characterized integrin expression in endothelium-denuded pulmonary arteries, and explored [Ca2+]i mobilization pathways induced by soluble ligands in rat PASMCs. Reverse transcriptase-PCR showed mRNA expression of integrins alpha1, alpha2, alpha3, alpha4, alpha5, alpha7, alpha8, alpha(v), beta1, beta3, and beta4, and immunoblots of alpha5, alpha(v), beta1, and beta3 confirmed protein expression. Exposure of PASMCs to integrin-binding peptides (0.5 mM) containing the arginine-glycine-aspartate (RGD) motif elicited [Ca2+]i responses with an order of potency of GRGDNP > GRGDSP > GRGDTP = cyclo-RGD. Pharmacological analysis revealed that the GRGDSP-induced Ca2+ response was unrelated to Ca2+ influx and the inositol triphosphate receptor-gated Ca2+ store, but partially blocked by ryanodine or inhibition of lysosome-related acidic organelles with bafilomycin A1. Simultaneous inhibition of both pathways was necessary to abolish the response. GRGDSP treatment increased cyclic ADP-ribose, the endogenous activator of ryanodine receptors, by 70%. GRGDSP also rapidly reduced Lysotracker Red accumulation, confirming direct modulation of acidic organelles. These data are the first demonstration of integrin-mediated Ca2+ regulation in PASMCs. The presence of an array of integrins, and activation of ryanodine-sensitive Ca2+ stores and lysosome-like organelles by GRGDSP suggest important roles for integrin-dependent Ca2+ signaling in regulating PASMC function.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Calcium/metabolism
- Calcium Signaling
- Cells, Cultured
- Fluorescence
- Integrins/metabolism
- Ligands
- Lysosomes/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Oligopeptides/pharmacology
- Organelles/metabolism
- Peptide Fragments/pharmacology
- Protein Binding
- Pulmonary Artery/cytology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction
- Ryanodine/pharmacology
- Ryanodine Receptor Calcium Release Channel/metabolism
Collapse
Affiliation(s)
- Anita Umesh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA
| | | | | | | | | |
Collapse
|
48
|
Cui S, Li C, Ema M, Weinstein J, Quaggin SE. Rapid isolation of glomeruli coupled with gene expression profiling identifies downstream targets in Pod1 knockout mice. J Am Soc Nephrol 2005; 16:3247-55. [PMID: 16207825 DOI: 10.1681/asn.2005030278] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Mouse mutations have provided tremendous insights into the molecular basis of renal and glomerular development. However, genes often play important roles during multiple stages of nephrogenesis, making it difficult to determine the role of a gene in a specific cell lineage such as the podocyte. Conditional gene targeting and chimeric analysis are two possible approaches to dissect the function of genes in specific cell populations. However, these are labor-intensive and costly and require the generation, validation, and analysis of additional transgenic lines. For overcoming these shortcomings and, specifically, for studying the role of gene function in developing glomeruli, a technique to isolate and purify glomeruli from murine embryos was developed. Combined with gene expression profiling, this method was used to identify differentially expressed genes in glomeruli from Pod1 knockout (KO) mice that die in the perinatal period with multiple renal defects. Glomeruli from early developing stages (late S-shape/early capillary loop) onward can be isolated successfully from wild-type and KO kidneys at 18.5 d postcoitus, and RNA can readily be obtained and used for genome-wide microarray analysis. With this approach, 3986 genes that are differently expressed between glomeruli from Pod1 KO and wild-type mice were identified, including a four-fold reduction of alpha 8 integrin mRNA in glomeruli from Pod1 KO mice that was confirmed by immunostaining. This procedure may be adapted to any transgenic strain, providing a rapid and efficient method to dissect the function of specific genes in glomerular development.
Collapse
Affiliation(s)
- Shiying Cui
- The Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
49
|
Ekwa-Ekoka C, Diaz GA, Carlson C, Hasegawa T, Samudrala R, Lim KC, Yabu JM, Levy B, Schnapp LM. Genomic organization and sequence variation of the human integrin subunit alpha8 gene (ITGA8). Matrix Biol 2005; 23:487-96. [PMID: 15579315 DOI: 10.1016/j.matbio.2004.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2004] [Revised: 07/31/2004] [Accepted: 08/09/2004] [Indexed: 11/20/2022]
Abstract
The integrin alpha8 is highly expressed during kidney and lung development. alpha8-deficient mice display abnormal renal development suggesting that alpha8 plays a critical role in organogenesis. Therefore, it would be of considerable interest to understand the genomic structure, localization and sequence variation of the alpha8 gene. Using FISH and genomic database analysis, we show that alpha8 gene maps to chromosome 10p13 and consists of >200 kbp organized into 30 exons. Examination of 47 individuals from two different ethnic groups (European and African descent) identified 286 varying sites. The diversity of alpha8 is comparable to that of other regions within the human genome. Eight of the varying sites were located in the coding regions: six resulted in nonsynonymous substitutions of which two lead to non-conservative changes in protein. None of the sites showed significant deviation from Hardy-Weinberg equilibrium. We mapped the coding region single nucleotide polymorphisms (SNPs) onto a model of the predicted alpha8 structure and found all the SNPs were located in the "calf" of the extracellular domain. In the European population, the linkage disequilibrium statistic D' showed three blocks of relatively non-recombinant regions in the alpha8 gene while the African population showed more evidence of recombination. The observed patterns of the linkage disequilibrium statistic R2 suggest that a large number of sites will need to be genotyped to ensure coverage of the entire gene for genetic association studies. Identification of the sequence variation will allow genetic association studies of alpha8 in kidney and lung disease.
Collapse
MESH Headings
- Base Sequence
- Chromosome Mapping
- Chromosomes, Human, Pair 10
- DNA/metabolism
- DNA, Complementary/metabolism
- Databases, Genetic
- Exons
- Genetic Variation
- Genome
- Genotype
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Integrin alpha Chains/genetics
- Integrin alpha Chains/metabolism
- Kidney/metabolism
- Linkage Disequilibrium
- Lung/metabolism
- Models, Genetic
- Models, Molecular
- Molecular Sequence Data
- Polymorphism, Genetic
- Polymorphism, Single Nucleotide
- Protein Conformation
- Protein Structure, Secondary
- Sequence Homology, Amino Acid
Collapse
|
50
|
Farias E, Lu M, Li X, Schnapp LM. Integrin α8β1–fibronectin interactions promote cell survival via PI3 kinase pathway. Biochem Biophys Res Commun 2005; 329:305-11. [PMID: 15721307 DOI: 10.1016/j.bbrc.2005.01.125] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Indexed: 11/20/2022]
Abstract
Integrin signaling plays a critical role in many aspects of normal growth, differentiation, and injury response. In the adult, alpha8beta1 is expressed in alveolar myofibroblasts and is upregulated in pulmonary fibrosis and other models of organ injury. Following injury, survival of fibronectin-producing myofibroblasts cells is an important determinant of development of fibrosis. Using stable alpha8-transfected cell lines, we show that interactions of alpha8beta1 with its ligand, fibronectin, promote cell survival during serum deprivation. Multiple cell signaling pathways were activated following fibronectin adhesion, including PI3 kinase and MAP kinase. However, the alpha8-mediated cell survival was blocked by LY294002, a PI3 kinase inhibitor, but not by staurosporine, a PKC inhibitor, or PD98059, a MAPK kinase inhibitor. A dominant negative construct of PI3 kinase also inhibited alpha8-mediated cell survival. Therefore, alpha8-mediated survival appears to be mediated by the PI3 kinase pathway. Survival of alpha8-expressing myofibroblasts may contribute to persistent fibrosis following injury.
Collapse
Affiliation(s)
- Eduardo Farias
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | |
Collapse
|