1
|
Torcq L, Majello S, Vivier C, Schmidt AA. Tuning apicobasal polarity and junctional recycling in the hemogenic endothelium orchestrates the morphodynamic complexity of emerging pre-hematopoietic stem cells. eLife 2024; 12:RP91429. [PMID: 38809590 PMCID: PMC11136496 DOI: 10.7554/elife.91429] [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: 05/30/2024] Open
Abstract
Hematopoietic stem cells emerge in the embryo from an aortic-derived tissue called the hemogenic endothelium (HE). The HE appears to give birth to cells of different nature and fate but the molecular principles underlying this complexity are largely unknown. Here we show, in the zebrafish embryo, that two cell types emerge from the aortic floor with radically different morphodynamics. With the support of live imaging, we bring evidence suggesting that the mechanics underlying the two emergence types rely, or not, on apicobasal polarity establishment. While the first type is characterized by reinforcement of apicobasal polarity and maintenance of the apical/luminal membrane until release, the second type emerges via a dynamic process reminiscent of trans-endothelial migration. Interfering with Runx1 function suggests that the balance between the two emergence types depends on tuning apicobasal polarity at the level of the HE. In support of this and unexpectedly, we show that Pard3ba - one of the four Pard3 proteins expressed in the zebrafish - is sensitive to interference with Runx1 activity, in aortic endothelial cells. This supports the idea of a signaling cross talk controlling cell polarity and its associated features, between aortic and hemogenic cells. In addition, using new transgenic fish lines that express Junctional Adhesion Molecules and functional interference, we bring evidence for the essential role of ArhGEF11/PDZ-RhoGEF in controlling the HE-endothelial cell dynamic interface, including cell-cell intercalation, which is ultimately required for emergence completion. Overall, we highlight critical cellular and dynamic events of the endothelial-to-hematopoietic transition that support emergence complexity, with a potential impact on cell fate.
Collapse
Affiliation(s)
- Léa Torcq
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
- Sorbonne UniversitéParisFrance
| | - Sara Majello
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
| | - Catherine Vivier
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
| | - Anne A Schmidt
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
| |
Collapse
|
2
|
Lin PK, Sun Z, Davis GE. Defining the Functional Influence of Endothelial Cell-Expressed Oncogenic Activating Mutations on Vascular Morphogenesis and Capillary Assembly. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:574-598. [PMID: 37838010 PMCID: PMC10988768 DOI: 10.1016/j.ajpath.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 10/16/2023]
Abstract
This study sought to define key molecules and signals controlling major steps in vascular morphogenesis, and how these signals regulate pericyte recruitment and pericyte-induced basement membrane deposition. The morphogenic impact of endothelial cell (EC) expression of activating mutants of Kirsten rat sarcoma virus (kRas), mitogen-activated protein kinase 1 (Mek1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), Akt serine/threonine kinase 1 (Akt1), Ras homolog enriched in brain (Rheb) Janus kinase 2 (Jak2), or signal transducer and activator of transcription 3 (Stat3) expression versus controls was evaluated, along with EC signaling events, pharmacologic inhibitor assays, and siRNA suppression experiments. Primary stimulators of EC lumen formation included kRas, Akt1, and Mek1, whereas PIK3CA and Akt1 stimulated a specialized type of cystic lumen formation. In contrast, the key drivers of EC sprouting behavior were Jak2, Stat3, Mek1, PIK3CA, and mammalian target of rapamycin (mTor). These conclusions are further supported by pharmacologic inhibitor and siRNA suppression experiments. EC expression of active Akt1, kRas, and PIK3CA led to markedly dysregulated lumen formation coupled to strongly inhibited pericyte recruitment and basement membrane deposition. For example, activated Akt1 expression in ECs excessively stimulated lumen formation, decreased EC sprouting behavior, and showed minimal pericyte recruitment with reduced mRNA expression of platelet-derived growth factor-BB, platelet-derived growth factor-DD, and endothelin-1, critical EC-derived factors known to stimulate pericyte invasion. The study identified key signals controlling fundamental steps in capillary morphogenesis and maturation and provided mechanistic details on why EC activating mutations induced a capillary deficiency state with abnormal lumens, impaired pericyte recruitment, and basement deposition: predisposing stimuli for the development of vascular malformations.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
3
|
Schumacher JA, Wright ZA, Rufin Florat D, Anand SK, Dasyani M, Batta SPR, Laverde V, Ferrari K, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E and ABL signaling regulate blood vessel size. PLoS Genet 2024; 20:e1010851. [PMID: 38190417 PMCID: PMC10798624 DOI: 10.1371/journal.pgen.1010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/19/2024] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Blood vessels in different vascular beds vary in size, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vessel size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow, eventually leading to the DA collapse. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA size in she mutants correlated with an increased endothelial expression of claudin 5a (cldn5a), which encodes a protein enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates vessel and lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surya Prakash Rao Batta
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Valentina Laverde
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Kaitlin Ferrari
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| |
Collapse
|
4
|
Lin PK, Koller GM, Davis GE. Elucidating the Morphogenic and Signaling Roles of Defined Growth Factors Controlling Human Endothelial Cell Lumen Formation Versus Sprouting Behavior. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2203-2217. [PMID: 37689384 PMCID: PMC10699133 DOI: 10.1016/j.ajpath.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/25/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
Five growth factors [ie, insulin, fibroblast growth factor-2 (FGF-2), stem cell factor, IL-3, and stromal-derived factor 1α] in combination are necessary for human endothelial cells (ECs) to undergo tube morphogenesis, a process requiring both lumen formation and sprouting behavior. This study investigated why these factors are required by subdividing the factors into 4 separate groups: insulin-only, insulin and FGF-2, no FGF-2 (all factors but without FGF-2), and all factors. The study found that the insulin-only condition failed to support EC morphogenesis or survival, the insulin and FGF-2 condition supported primarily EC lumen formation, and the no FGF-2 condition supported EC sprouting behavior. By comparison, the all-factors condition more strongly stimulated both EC lumen formation and sprouting behavior, and signaling analysis revealed prolonged stimulation of multiple promorphogenic signals coupled with inhibition of proregressive signals. Pharmacologic inhibition of Jak kinases more selectively blocked EC sprouting behavior, whereas inhibition of Raf, phosphatidylinositol 3-kinase, and Akt kinases showed selective blockade of lumen formation. Inhibition of Src family kinases and Notch led to increased sprouting coupled to decreased lumen formation, whereas inhibition of Pak, Mek, and mammalian target of rapamycin kinases blocked both sprouting and lumen formation. These findings reveal novel downstream biological and signaling activities of defined factors that are required for the assembly of human EC-lined capillary tube networks.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
5
|
Schumacher JA, Wright ZA, Florat DR, Anand SK, Dasyani M, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E (SHE) and ABL signaling regulate blood vessel size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547455. [PMID: 37461480 PMCID: PMC10349984 DOI: 10.1101/2023.07.03.547455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Blood vessels in different vascular beds vary in lumen diameter, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vascular lumen size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA lumen, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA lumen in she mutants correlated with an increased endothelial expression of claudin 5a and 5b (cldn5a / cldn5b), which encode proteins enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA lumen size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| |
Collapse
|
6
|
Davis GE, Kemp SS. Extracellular Matrix Regulation of Vascular Morphogenesis, Maturation, and Stabilization. Cold Spring Harb Perspect Med 2023; 13:a041156. [PMID: 35817544 PMCID: PMC10578078 DOI: 10.1101/cshperspect.a041156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The extracellular matrix represents a critical regulator of tissue vascularization during embryonic development and postnatal life. In this perspective, we present key information and concepts that focus on how the extracellular matrix controls capillary assembly, maturation, and stabilization, and, in addition, contributes to tissue stability and health. In particular, we present and discuss mechanistic details underlying (1) the role of the extracellular matrix in controlling different steps of vascular morphogenesis, (2) the ability of endothelial cells (ECs) and pericytes to coassemble into elongated and narrow capillary EC-lined tubes with associated pericytes and basement membrane matrices, and (3) the identification of specific growth factor combinations ("factors") and peptides as well as coordinated "factor" and extracellular matrix receptor signaling pathways that are required to form stabilized capillary networks.
Collapse
Affiliation(s)
- George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida 33612, USA
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida 33612, USA
| |
Collapse
|
7
|
Pillay LM, Yano JJ, Davis AE, Butler MG, Ezeude MO, Park JS, Barnes KA, Reyes VL, Castranova D, Gore AV, Swift MR, Iben JR, Kenton MI, Stratman AN, Weinstein BM. In vivo dissection of Rhoa function in vascular development using zebrafish. Angiogenesis 2022; 25:411-434. [PMID: 35320450 DOI: 10.1007/s10456-022-09834-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 02/22/2022] [Indexed: 12/27/2022]
Abstract
The small monomeric GTPase RHOA acts as a master regulator of signal transduction cascades by activating effectors of cellular signaling, including the Rho-associated protein kinases ROCK1/2. Previous in vitro cell culture studies suggest that RHOA can regulate many critical aspects of vascular endothelial cell (EC) biology, including focal adhesion, stress fiber formation, and angiogenesis. However, the specific in vivo roles of RHOA during vascular development and homeostasis are still not well understood. In this study, we examine the in vivo functions of RHOA in regulating vascular development and integrity in zebrafish. We use zebrafish RHOA-ortholog (rhoaa) mutants, transgenic embryos expressing wild type, dominant negative, or constitutively active forms of rhoaa in ECs, pharmacological inhibitors of RHOA and ROCK1/2, and Rock1 and Rock2a/b dgRNP-injected zebrafish embryos to study the in vivo consequences of RHOA gain- and loss-of-function in the vascular endothelium. Our findings document roles for RHOA in vascular integrity, developmental angiogenesis, and vascular morphogenesis in vivo, showing that either too much or too little RHOA activity leads to vascular dysfunction.
Collapse
Affiliation(s)
- Laura M Pillay
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Joseph J Yano
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
- Department of Cell and Molecular Biology, University of Pennsylvania, 440 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Andrew E Davis
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Matthew G Butler
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Megan O Ezeude
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Jong S Park
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Keith A Barnes
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Vanessa L Reyes
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Daniel Castranova
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Aniket V Gore
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Matthew R Swift
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - James R Iben
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Madeleine I Kenton
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
| | - Amber N Stratman
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brant M Weinstein
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Dr. Bethesda, Bethesda, MD, 20892, USA.
| |
Collapse
|
8
|
Han R, Gong R, Liu W, Xu G. Proteome changes associated with the VEGFR pathway and immune system in diabetic macular edema patients at different diabetic retinopathy stages. Curr Eye Res 2022; 47:1050-1060. [PMID: 35435079 DOI: 10.1080/02713683.2022.2068181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE Diabetic macular edema (DME) is a major cause of vision loss in all stages of diabetic retinopathy (DR). However, there is limited recognition of aqueous humor (AH) proteome profiles of DME patients at different DR stages. In this study, we aimed to investigate the AH proteome changes between DME patients at the nonproliferative diabetic retinopathy (NPDR) stage and those at the proliferative diabetic retinopathy (PDR) stage. METHODS A label-free data-independent acquisition based liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis was performed to profile the abundances of AH proteins in 73 eyes from DME patients at different DR stages. Enzyme-linked immunosorbent assay (ELISA) was used to confirm the proteomics results with AH samples from non-diabetic patients and DME patients at the NPDR or PDR stage. RESULTS LC-MS/MS results showed significantly changed expression of 308 proteins between DME patients in the NPDR and PDR groups. Compared to the NPDR group, the proteins relatively up-regulated in the PDR group are involved in the immune system and/or negative regulation of the cell cycle, while proteins relatively down-regulated in the PDR group are associated with the vascular endothelial growth factor receptor (VEGFR) pathway and/or metabolism. ELISA results further verified the proteomic result of down-regulated expression of the immune-associated protein cystatin C (CST3) in the PDR group compared to that in the NPDR and non-diabetic groups. CONCLUSIONS In this study, we reported for the first time the decreased abundances of AH proteins associated with the VEGFR pathway and both down- and up-regulated expression of AH proteins associated with the immune system in the PDR group compared to that in the NPDR group. Furthermore, we found negative correlations of immune-associated protein, CST3 concentration in AH with DR severity and central retinal thickness, suggesting CST3 as a promising target independent of the VEGFR pathway in DME-involved DR treatment.
Collapse
Affiliation(s)
- Ruyi Han
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, 200030, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200030,China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, 200030, China
| | - Ruowen Gong
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, 200030, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200030,China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, 200030, China
| | - Wei Liu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, 200030, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200030,China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, 200030, China
| | - Gezhi Xu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, 200030, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200030,China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, 200030, China
| |
Collapse
|
9
|
Sun Z, Kemp SS, Lin PK, Aguera KN, Davis GE. Endothelial k-RasV12 Expression Induces Capillary Deficiency Attributable to Marked Tube Network Expansion Coupled to Reduced Pericytes and Basement Membranes. Arterioscler Thromb Vasc Biol 2022; 42:205-222. [PMID: 34879709 PMCID: PMC8792373 DOI: 10.1161/atvbaha.121.316798] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We sought to determine how endothelial cell (EC) expression of the activating k-Ras (kirsten rat sarcoma 2 viral oncogene homolog) mutation, k-RasV12, affects their ability to form lumens and tubes and interact with pericytes during capillary assembly Approach and Results: Using defined bioassays where human ECs undergo observable tubulogenesis, sprouting behavior, pericyte recruitment to EC-lined tubes, and pericyte-induced EC basement membrane deposition, we assessed the impact of EC k-RasV12 expression on these critical processes that are necessary for proper capillary network formation. This mutation, which is frequently seen in human ECs within brain arteriovenous malformations, was found to markedly accentuate EC lumen formation mechanisms, with strongly accelerated intracellular vacuole formation, vacuole fusion, and lumen expansion and with reduced sprouting behavior, leading to excessively widened tube networks compared with control ECs. These abnormal tubes demonstrate strong reductions in pericyte recruitment and pericyte-induced EC basement membranes compared with controls, with deficiencies in fibronectin, collagen type IV, and perlecan deposition. Analyses of signaling during tube formation from these k-RasV12 ECs reveals strong enhancement of Src (Src proto-oncogene, non-receptor tyrosine kinase), Pak2 (P21 [RAC1 (Rac family small GTPase 1)] activated kinase 2), b-Raf (v-raf murine sarcoma viral oncogene homolog B1), Erk (extracellular signal-related kinase), and Akt (AK strain transforming) activation and increased expression of PKCε (protein kinase C epsilon), MT1-MMP (membrane-type 1 matrix metalloproteinase), acetylated tubulin and CDCP1 (CUB domain-containing protein 1; most are known EC lumen regulators). Pharmacological blockade of MT1-MMP, Src, Pak, Raf, Mek (mitogen-activated protein kinase) kinases, Cdc42 (cell division cycle 42)/Rac1, and Notch markedly interferes with lumen and tube formation from these ECs. CONCLUSIONS Overall, this novel work demonstrates that EC expression of k-RasV12 disrupts capillary assembly due to markedly excessive lumen formation coupled with strongly reduced pericyte recruitment and basement membrane deposition, which are critical pathogenic features predisposing the vasculature to develop arteriovenous malformations.
Collapse
Affiliation(s)
- Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - Scott S. Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - Kalia N. Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
10
|
Kemp SS, Lin PK, Sun Z, Castaño MA, Yrigoin K, Penn MR, Davis GE. Molecular basis for pericyte-induced capillary tube network assembly and maturation. Front Cell Dev Biol 2022; 10:943533. [PMID: 36072343 PMCID: PMC9441561 DOI: 10.3389/fcell.2022.943533] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Here we address the functional importance and role of pericytes in capillary tube network assembly, an essential process that is required for vascularized tissue development, maintenance, and health. Healthy capillaries may be directly capable of suppressing human disease. Considerable advances have occurred in our understanding of the molecular and signaling requirements controlling EC lumen and tube formation in 3D extracellular matrices. A combination of SCF, IL-3, SDF-1α, FGF-2 and insulin ("Factors") in conjunction with integrin- and MT1-MMP-induced signaling are required for EC sprouting behavior and tube formation under serum-free defined conditions. Pericyte recruitment to the abluminal EC tube surface results in elongated and narrow tube diameters and deposition of the vascular basement membrane. In contrast, EC tubes in the absence of pericytes continue to widen and shorten over time and fail to deposit basement membranes. Pericyte invasion, recruitment and proliferation in 3D matrices requires the presence of ECs. A detailed analysis identified that EC-derived PDGF-BB, PDGF-DD, ET-1, HB-EGF, and TGFβ1 are necessary for pericyte recruitment, proliferation, and basement membrane deposition. Blockade of these individual factors causes significant pericyte inhibition, but combined blockade profoundly interferes with these events, resulting in markedly widened EC tubes without basement membranes, like when pericytes are absent.
Collapse
Affiliation(s)
- Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Maria A Castaño
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Ksenia Yrigoin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Marlena R Penn
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| |
Collapse
|
11
|
Hwang SH, Lee YM, Choi Y, Son HE, Ryu JY, Na KY, Chin HJ, Jeon NL, Kim S. Role of Human Primary Renal Fibroblast in TGF-β1-Mediated Fibrosis-Mimicking Devices. Int J Mol Sci 2021; 22:ijms221910758. [PMID: 34639099 PMCID: PMC8509581 DOI: 10.3390/ijms221910758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 01/21/2023] Open
Abstract
Renal fibrosis is a progressive chronic kidney disease that ultimately leads to end-stage renal failure. Despite several approaches to combat renal fibrosis, an experimental model to evaluate currently available drugs is not ideal. We developed fibrosis-mimicking models using three-dimensional (3D) co-culture devices designed with three separate layers of tubule interstitium, namely, epithelial, fibroblastic, and endothelial layers. We introduced human renal proximal tubular epithelial cells (HK-2), human umbilical-vein endothelial cells, and patient-derived renal fibroblasts, and evaluated the effects of transforming growth factor-β (TGF-β) and TGF-β inhibitor treatment on this renal fibrosis model. The expression of the fibrosis marker alpha smooth muscle actin upon TGF-β1 treatment was augmented in monolayer-cultured HK-2 cells in a 3D disease model. In the vascular compartment of renal fibrosis models, the density of vessels was increased and decreased in the TGF-β-treated group and TGF-β-inhibitor treatment group, respectively. Multiplex ELISA using supernatants in the TGF-β-stimulating 3D models showed that pro-inflammatory cytokine and growth factor levels including interleukin-1 beta, tumor necrosis factor alpha, basic fibroblast growth factor, and TGF-β1, TGF-β2, and TGF-β3 were increased, which mimicked the fibrotic microenvironments of human kidneys. This study may enable the construction of a human renal fibrosis-mimicking device model beyond traditional culture experiments.
Collapse
Affiliation(s)
- Seong-Hye Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
| | - Yun-Mi Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
| | - Yunyeong Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
| | - Hyung Eun Son
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ji Young Ryu
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ki Young Na
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ho Jun Chin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Noo Li Jeon
- Program for Bioengineering, School of Engineering, Seoul National University, Seoul 08826, Korea
- Correspondence: (N.L.J.); (S.K.); Tel.: +82-31-787-7051 (S.K.)
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: (N.L.J.); (S.K.); Tel.: +82-31-787-7051 (S.K.)
| |
Collapse
|
12
|
Lin PK, Salvador J, Xie J, Aguera KN, Koller GM, Kemp SS, Griffin CT, Davis GE. Selective and Marked Blockade of Endothelial Sprouting Behavior Using Paclitaxel and Related Pharmacologic Agents. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2245-2264. [PMID: 34563512 DOI: 10.1016/j.ajpath.2021.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jocelynda Salvador
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jun Xie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kalia N Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
13
|
Castro M, Laviña B, Ando K, Álvarez-Aznar A, Abu Taha A, Brakebusch C, Dejana E, Betsholtz C, Gaengel K. CDC42 Deletion Elicits Cerebral Vascular Malformations via Increased MEKK3-Dependent KLF4 Expression. Circ Res 2020; 124:1240-1252. [PMID: 30732528 DOI: 10.1161/circresaha.118.314300] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE Aberrant formation of blood vessels precedes a broad spectrum of vascular complications; however, the cellular and molecular events governing vascular malformations are not yet fully understood. OBJECTIVE Here, we investigated the role of CDC42 (cell division cycle 42) during vascular morphogenesis and its relative importance for the development of cerebrovascular malformations. METHODS AND RESULTS To avoid secondary systemic effects often associated with embryonic gene deletion, we generated an endothelial-specific and inducible knockout approach to study postnatal vascularization of the mouse brain. Postnatal endothelial-specific deletion of Cdc42 elicits cerebrovascular malformations reminiscent of cerebral cavernous malformations (CCMs). At the cellular level, loss of CDC42 function in brain endothelial cells (ECs) impairs their sprouting, branching morphogenesis, axial polarity, and normal dispersion within the brain tissue. Disruption of CDC42 does not alter EC proliferation, but malformations occur where EC proliferation is the most pronounced during brain development-the postnatal cerebellum-indicating that a high, naturally occurring EC proliferation provides a permissive state for the appearance of these malformations. Mechanistically, CDC42 depletion in ECs elicited increased MEKK3 (mitogen-activated protein kinase kinase kinase 3)-MEK5 (mitogen-activated protein kinase kinase 5)-ERK5 (extracellular signal-regulated kinase 5) signaling and consequent detrimental overexpression of KLF (Kruppel-like factor) 2 and KLF4, recapitulating the hallmark mechanism for CCM pathogenesis. Through genetic approaches, we demonstrate that the coinactivation of Klf4 reduces the severity of vascular malformations in Cdc42 mutant mice. Moreover, we show that CDC42 interacts with CCMs and that CCM3 promotes CDC42 activity in ECs. CONCLUSIONS We show that endothelial-specific deletion of Cdc42 elicits CCM-like cerebrovascular malformations and that CDC42 is engaged in the CCM signaling network to restrain the MEKK3-MEK5-ERK5-KLF2/4 pathway.
Collapse
Affiliation(s)
- Marco Castro
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| | - Bàrbara Laviña
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| | - Koji Ando
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| | - Alberto Álvarez-Aznar
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| | - Abdallah Abu Taha
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| | - Cord Brakebusch
- Biotech Research and Innovation Center, University of Copenhagen, Denmark (C. Brakebusch).,ICMC (Integrated Cardio Metabolic Centre), Karolinska Institutet/AstraZeneca/Integrated Cardio Metabolic Centre, Huddinge, Stockholm, Sweden (C. Betsholtz)
| | - Elisabetta Dejana
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.).,FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology Foundation, Milan, Italy (E.D.)
| | - Christer Betsholtz
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| | - Konstantin Gaengel
- From the Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.C., B.L., K.A., A.Á.-A., A.A.T., E.D., C. Betsholtz, K.G.)
| |
Collapse
|
14
|
Barlow HR, Cleaver O. Building Blood Vessels-One Rho GTPase at a Time. Cells 2019; 8:cells8060545. [PMID: 31174284 PMCID: PMC6627795 DOI: 10.3390/cells8060545] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 01/09/2023] Open
Abstract
Blood vessels are required for the survival of any organism larger than the oxygen diffusion limit. Blood vessel formation is a tightly regulated event and vessel growth or changes in permeability are linked to a number of diseases. Elucidating the cell biology of endothelial cells (ECs), which are the building blocks of blood vessels, is thus critical to our understanding of vascular biology and to the development of vascular-targeted disease treatments. Small GTPases of the Rho GTPase family are known to regulate several processes critical for EC growth and maintenance. In fact, many of the 21 Rho GTPases in mammals are known to regulate EC junctional remodeling, cell shape changes, and other processes. Rho GTPases are thus an attractive target for disease treatments, as they often have unique functions in specific vascular cell types. In fact, some Rho GTPases are even expressed with relative specificity in diseased vessels. Interestingly, many Rho GTPases are understudied in ECs, despite their known expression in either developing or mature vessels, suggesting an even greater wealth of knowledge yet to be gleaned from these complex signaling pathways. This review aims to provide an overview of Rho GTPase signaling contributions to EC vasculogenesis, angiogenesis, and mature vessel barrier function. A particular emphasis is placed on so-called "alternative" Rho GTPases, as they are largely understudied despite their likely important contributions to EC biology.
Collapse
Affiliation(s)
- Haley Rose Barlow
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Ondine Cleaver
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
The precise molecular signals that control endothelial cell-cell adhesion within the vessel wall. Biochem Soc Trans 2018; 46:1673-1680. [PMID: 30514769 PMCID: PMC6299237 DOI: 10.1042/bst20180377] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 12/23/2022]
Abstract
Endothelial cell–cell adhesion within the wall of the vasculature controls a range of physiological processes, such as growth, integrity and barrier function. The adhesive properties of endothelial cells are tightly controlled by a complex cascade of signals transmitted from the surrounding environment or from within the cells themselves, with the dynamic nature of cellular adhesion and the regulating signalling networks now beginning to be appreciated. Here, we summarise the current knowledge of the mechanisms controlling endothelial cell–cell adhesion in the developing and mature blood vasculature.
Collapse
|
16
|
Pulmonary Vascular Platform Models the Effects of Flow and Pressure on Endothelial Dysfunction in BMPR2 Associated Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19092561. [PMID: 30158434 PMCID: PMC6164056 DOI: 10.3390/ijms19092561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Endothelial dysfunction is a known consequence of bone morphogenetic protein type II receptor (BMPR2) mutations seen in pulmonary arterial hypertension (PAH). However, standard 2D cell culture models fail to mimic the mechanical environment seen in the pulmonary vasculature. Hydrogels have emerged as promising platforms for 3D disease modeling due to their tunable physical and biochemical properties. In order to recreate the mechanical stimuli seen in the pulmonary vasculature, we have created a novel 3D hydrogel-based pulmonary vasculature model (“artificial arteriole”) that reproduces the pulsatile flow rates and pressures seen in the human lung. Using this platform, we studied both Bmpr2R899X and WT endothelial cells to better understand how the addition of oscillatory flow and physiological pressure influenced gene expression, cell morphology, and cell permeability. The addition of oscillatory flow and pressure resulted in several gene expression changes in both WT and Bmpr2R899X cells. However, for many pathways with relevance to PAH etiology, Bmpr2R899X cells responded differently when compared to the WT cells. Bmpr2R899X cells were also found not to elongate in the direction of flow, and instead remained stagnant in morphology despite mechanical stimuli. The increased permeability of the Bmpr2R899X layer was successfully reproduced in our artificial arteriole, with the addition of flow and pressure not leading to significant changes in permeability. Our artificial arteriole is the first to model many mechanical properties seen in the lung. Its tunability enables several new opportunities to study the endothelium in pulmonary vascular disease with increased control over environmental parameters.
Collapse
|
17
|
Laviña B, Castro M, Niaudet C, Cruys B, Álvarez-Aznar A, Carmeliet P, Bentley K, Brakebusch C, Betsholtz C, Gaengel K. Defective endothelial cell migration in the absence of Cdc42 leads to capillary-venous malformations. Development 2018; 145:dev.161182. [PMID: 29853619 DOI: 10.1242/dev.161182] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/24/2018] [Indexed: 12/26/2022]
Abstract
Formation and homeostasis of the vascular system requires several coordinated cellular functions, but their precise interplay during development and their relative importance for vascular pathologies remain poorly understood. Here, we investigated the endothelial functions regulated by Cdc42 and their in vivo relevance during angiogenic sprouting and vascular morphogenesis in the postnatal mouse retina. We found that Cdc42 is required for endothelial tip cell selection, directed cell migration and filopodia formation, but dispensable for cell proliferation or apoptosis. Although the loss of Cdc42 seems generally compatible with apical-basal polarization and lumen formation in retinal blood vessels, it leads to defective endothelial axial polarization and to the formation of severe vascular malformations in capillaries and veins. Tracking of Cdc42-depleted endothelial cells in mosaic retinas suggests that these capillary-venous malformations arise as a consequence of defective cell migration, when endothelial cells that proliferate at normal rates are unable to re-distribute within the vascular network.
Collapse
Affiliation(s)
- Bàrbara Laviña
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden
| | - Marco Castro
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden
| | - Colin Niaudet
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden
| | - Bert Cruys
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, Belgium
| | - Alberto Álvarez-Aznar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, Belgium
| | - Katie Bentley
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden.,Computational Biology Laboratory, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Cord Brakebusch
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden .,Integrated Cardio Metabolic Centre (ICMC), Department of Medicine Huddinge, Karolinska Institute, Novum, SE-141 57 Huddinge, Stockholm, Sweden
| | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden
| |
Collapse
|
18
|
Blache U, Ehrbar M. Inspired by Nature: Hydrogels as Versatile Tools for Vascular Engineering. Adv Wound Care (New Rochelle) 2018; 7:232-246. [PMID: 29984113 PMCID: PMC6032659 DOI: 10.1089/wound.2017.0760] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/21/2022] Open
Abstract
Significance: Diseases related to vascular malfunction, hyper-vascularization, or lack of vascularization are among the leading causes of morbidity and mortality. Engineered, vascularized tissues as well as angiogenic growth factor-releasing hydrogels could replace defective tissues. Further, treatments and testing of novel vascular therapeutics will benefit significantly from models that allow for the study of vascularized tissues under physiological relevant in vitro conditions. Recent Advances: Inspired by fibrin, the provisional matrix during wound healing, naturally derived and synthetic hydrogel scaffolds have been developed for vascular engineering. Today, engineers and biologists use commercially available hydrogels to pre-vascularize tissues, to control the delivery of angiogenic growth factors, and to establish vascular diseases models. Critical Issue: For clinical translation, pre-vascularized tissue constructs must be sufficiently large and stable to substitute function-relevant tissue defects and integrate with host vascular perfusion. Moreover, the continuous integration of knowhow from basic vascular biology with innovative, tailorable materials and advanced manufacturing technologies is key to achieving near-physiological tissue models and new treatments to control vascularization. Future Directions: For transplantation, engineered tissues must comprise hierarchically organized vascular trees of different caliber and function. The development of novel vascularization-promoting or -inhibiting therapeutics will benefit from physiologically relevant vessel models. In addition, tissue models representing treatment-relevant vascular tissue functions will increase the capacity to screen for therapeutic compounds and will significantly reduce the need for animals for their validation.
Collapse
Affiliation(s)
- Ulrich Blache
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Nguyen DHT, Gao L, Wong A, Chen CS. Cdc42 regulates branching in angiogenic sprouting in vitro. Microcirculation 2018; 24. [PMID: 28376260 DOI: 10.1111/micc.12372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/27/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES The morphogenetic events that occur during angiogenic sprouting involve several members of the Rho family of GTPases, including Cdc42. However, the precise roles of Cdc42 in angiogenic sprouting have been difficult to elucidate owing to the lack of models to study these events in vitro. Here, we aim to identify the roles of Cdc42 in branching morphogenesis in angiogenesis. METHODS Using a 3D biomimetic model of angiogenesis in vitro, where endothelial cells were seeded inside a cylindrical channel within collagen gel and sprouted from the channel in response to a defined biochemical gradient of angiogenic factors, we inhibited Cdc42 activity with a small molecule inhibitor ML141 and examined the effects of Cdc42 on the morphogenetic processes of angiogenic sprouting. RESULTS We find that partial inhibition of Cdc42 had minimal effects on directional migration of endothelial cells, but led to fewer branching events without affecting the length of these branches. We also observed that antagonizing Cdc42 reduced collective migration in favor of single cell migration. Additionally, Cdc42 also regulated the initiation of filopodial extensions in endothelial tip cells. CONCLUSIONS Our findings suggest that Cdc42 can affect multiple morphogenetic processes during angiogenic sprouting and ultimately impact the architecture of the vasculature.
Collapse
Affiliation(s)
- Duc-Huy T Nguyen
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Gao
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Alec Wong
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Christopher S Chen
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA.,Department of Biomedical Engineering, Boston University, Boston, MA, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| |
Collapse
|
20
|
|
21
|
Shubin AV, Demidyuk IV, Komissarov AA, Rafieva LM, Kostrov SV. Cytoplasmic vacuolization in cell death and survival. Oncotarget 2018; 7:55863-55889. [PMID: 27331412 PMCID: PMC5342458 DOI: 10.18632/oncotarget.10150] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/06/2016] [Indexed: 12/15/2022] Open
Abstract
Cytoplasmic vacuolization (also called cytoplasmic vacuolation) is a well-known morphological phenomenon observed in mammalian cells after exposure to bacterial or viral pathogens as well as to various natural and artificial low-molecular-weight compounds. Vacuolization often accompanies cell death; however, its role in cell death processes remains unclear. This can be attributed to studying vacuolization at the level of morphology for many years. At the same time, new data on the molecular mechanisms of the vacuole formation and structure have become available. In addition, numerous examples of the association between vacuolization and previously unknown cell death types have been reported. Here, we review these data to make a deeper insight into the role of cytoplasmic vacuolization in cell death and survival.
Collapse
Affiliation(s)
- Andrey V Shubin
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia.,Laboratory of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.,Laboratory of Biologically Active Nanostructures, N.F. Gamaleya Institute of Epidemiology and Microbiology, Moscow, Russia
| | - Ilya V Demidyuk
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| | - Alexey A Komissarov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| | - Lola M Rafieva
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| | - Sergey V Kostrov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| |
Collapse
|
22
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
23
|
Gopal SK, Greening DW, Hanssen EG, Zhu HJ, Simpson RJ, Mathias RA. Oncogenic epithelial cell-derived exosomes containing Rac1 and PAK2 induce angiogenesis in recipient endothelial cells. Oncotarget 2017; 7:19709-22. [PMID: 26919098 PMCID: PMC4991413 DOI: 10.18632/oncotarget.7573] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/06/2016] [Indexed: 01/06/2023] Open
Abstract
The metastatic cascade describes the escape of primary tumour cells to distant secondary sites. Cells at the leading tumour edge are thought to undergo epithelial-mesenchymal transition (EMT), to enhance their motility and invasion for spreading. Whether EMT cells directly promote tumour angiogenesis, and the role of exosomes (30-150 nm extracellular vesicles) remains largely unknown. We examined the functional effects of exosomes from MDCK cells, MDCK cells stably expressing YBX1 (MDCKYBX1, intermediate EMT), and Ras-transformed MDCK cells (21D1 cells, complete EMT). 2F-2B cell motility and tube formation (length and branching) was significantly increased following supplementation with MDCKYBX1 or 21D1 exosomes, but not MDCK exosomes. Next, Matrigel™ plugs containing exosome-supplemented 2F-2B cells were subcutaneously injected into mice. Systemic perfusion was only observed for plugs supplemented with MDCKYBX1 or 21D1 exosomes. Comparative proteomics revealed that 21D1 exosomes contained VEGF-associated proteins, while MDCKYBX1 exosomes were enriched with activated Rac1 and PAK2. To validate, 2F-2B cells and HUVECs were pre-treated with PAK inhibitors prior to exosome supplementation. PAK inhibition nullified the effects of MDCKYBX1 exosomes by reducing the tube length and branching to baseline levels. By contrast, the effects of 21D1 exosomes were not significantly decreased. Our results demonstrate for the first time that oncogenic cells undergoing EMT can communicate with endothelial cells via exosomes, and establish exosomal Rac1/PAK2 as angiogenic promoters that may function from early stages of the metastatic cascade.
Collapse
Affiliation(s)
- Shashi K Gopal
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Eric G Hanssen
- Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hong-Jian Zhu
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Rommel A Mathias
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Abstract
Angiogenesis and vascular remodeling are essential for the establishment of vascular networks during organogenesis. Here we show that the Hippo signaling pathway effectors YAP and TAZ are required, in a gene dosage-dependent manner, for the proliferation and migration of vascular endothelial cells (ECs) during retinal angiogenesis. Intriguingly, nuclear translocation of YAP and TAZ induced by Lats1/2-deletion blocked endothelial migration and phenocopied Yap/Taz-deficient mutants. Furthermore, overexpression of a cytoplasmic form of YAP (YAPS127D) partially rescued the migration defects caused by loss of YAP and TAZ function. Finally, we found that cytoplasmic YAP positively regulated the activity of the small GTPase CDC42, deletion of which caused severe defects in endothelial migration. These findings uncover a previously unrecognized role of cytoplasmic YAP/TAZ in promoting cell migration by activating CDC42 and provide insight into how Hippo signaling in ECs regulates angiogenesis.
Collapse
|
25
|
Ahmad SM. Conserved signaling mechanisms in Drosophila heart development. Dev Dyn 2017; 246:641-656. [PMID: 28598558 PMCID: PMC11546222 DOI: 10.1002/dvdy.24530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/06/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022] Open
Abstract
Signal transduction through multiple distinct pathways regulates and orchestrates the numerous biological processes comprising heart development. This review outlines the roles of the FGFR, EGFR, Wnt, BMP, Notch, Hedgehog, Slit/Robo, and other signaling pathways during four sequential phases of Drosophila cardiogenesis-mesoderm migration, cardiac mesoderm establishment, differentiation of the cardiac mesoderm into distinct cardiac cell types, and morphogenesis of the heart and its lumen based on the proper positioning and cell shape changes of these differentiated cardiac cells-and illustrates how these same cardiogenic roles are conserved in vertebrates. Mechanisms bringing about the regulation and combinatorial integration of these diverse signaling pathways in Drosophila are also described. This synopsis of our present state of knowledge of conserved signaling pathways in Drosophila cardiogenesis and the means by which it was acquired should facilitate our understanding of and investigations into related processes in vertebrates. Developmental Dynamics 246:641-656, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shaad M. Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
26
|
Botulinum Toxin A Upregulates Rac1, Cdc42, and RhoA Gene Expression in a Dose-Dependent Manner: In Vivo and in Vitro Study. J Craniofac Surg 2016; 27:516-20. [PMID: 26963302 DOI: 10.1097/scs.0000000000002272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
UNLABELLED Angiogenesis is the development of new capillaries from existing blood vessels and is a prerequisite for the wound-healing process. Many lines of scientific evidences have shown that complicated roles of small guanosine triphosphatases (GTPases) (ras-related C3 botulinum toxin substrate 1 [Rac1], cell division control protein 42 [Cdc42], and ras homolog gene family, member A [RhoA]) in regulation of signal transduction pathways exist to transmit distinct cellular effects on the modulation of actin cytoskeleton remodeling such as cell cycle progression, cell survival, and cell motility. In addition, these small GTPases activate mitogen-activated protein kinase kinase kinases (MAP3Ks) leading to activated mitogen-activated protein kinase kinases (MAPKK), mitogen-activated protein kinase (MAPK), and various transcription factors such as vascular endothelial growth factor with involvement of MAPK signaling pathways.In this study, the authors hypothesized that botulinum toxin A increases angiogenesis via the expression of small GTPases in vivo and in vitro studies.In vivo experiment, 24 Sprague-Dawley rats were randomly divided into 2 groups: a control group and a botulinum toxin A group. Five days prior to superiorly based transverse rectus abdominis myocutaneous flap elevation, the botulinum toxin A (BoTA) group was pretreated with BoTA, while the control group was pretreated with normal saline. quantitative real-time polymerase chain reaction was performed to evaluate the expression of Rac1, RhoA, and Cdc42.The angiogenic effects of botulinum toxin A on human dermal fibroblasts were measured in vitro experiment. To understand the mechanism of botulinum toxin A on small GTPases production of fibroblasts, Rac1, Cdc42, and RhoA were measured using qRT-PCR.The relative messenger ribonucleic acid expression of Rac1, RhoA, and Cdc42 was significantly higher in the BoTA group than in the control group, in every zone and pedicle muscle, on postoperative days 1, 3, and 5. Levels of these molecules increased significantly in human dermal fibroblasts grown in the presence of BoTA compared with control group over 5 IU.Our in vivo and in vitro studies suggest that administration of BoTA upregulates the expression of RhoA, Rac1, and Cdc42 in a dose-dependent manner. MAPK signaling pathway might be involved in BoTA-induced angiogenesis mechanism. LEVEL OF EVIDENCE N/A.
Collapse
|
27
|
Lv J, Zeng J, Zhao W, Cheng Y, Zhang L, Cai S, Hu G, Chen Y. Cdc42 regulates LPS-induced proliferation of primary pulmonary microvascular endothelial cells via ERK pathway. Microvasc Res 2016; 109:45-53. [PMID: 27769693 DOI: 10.1016/j.mvr.2016.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/21/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND After stimulation due to injury, cell division cycle protein 42 (Cdc42) restores and enhances barrier functions by strengthening intercellular adherens junctions; however, its influence on cell proliferation after injury remains unknown. OBJECTIVE In this study, we sought to investigate the effect of stimulation using small doses of lipopolysaccharide (LPS) on the proliferation of pulmonary microvascular endothelial cells (PMVECs). METHODS We stimulated PMVECs with different doses of LPS and evaluated the effects on cell proliferation. We also constructed a primary gene-knockout cell line lacking Cdc42 to verify the role of Cdc42 in regulating the proliferation of PMVECs that were stimulated using LPS and to explore related signaling pathways. RESULTS Stimulating PMVECs with small doses of LPS increased proliferation. Cdc42 is involved in regulating this process, which was mediated by the extracellular regulated protein kinase (ERK) pathway. CONCLUSIONS Cdc42 plays a role in regulating the proliferation of PMVECs stimulated with small doses of LPS, and this regulation involves the ERK pathway.
Collapse
Affiliation(s)
- Jiawen Lv
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junchao Zeng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wen Zhao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuanxiong Cheng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences Southern Medical University, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Shaoxi Cai
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guodong Hu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yinghua Chen
- Department of Histology and Embryology, School of Basic Medical Sciences Southern Medical University, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
28
|
Barry DM, Koo Y, Norden PR, Wylie LA, Xu K, Wichaidit C, Azizoglu DB, Zheng Y, Cobb MH, Davis GE, Cleaver O. Rasip1-Mediated Rho GTPase Signaling Regulates Blood Vessel Tubulogenesis via Nonmuscle Myosin II. Circ Res 2016; 119:810-26. [PMID: 27486147 DOI: 10.1161/circresaha.116.309094] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE Vascular tubulogenesis is essential to cardiovascular development. Within initial vascular cords of endothelial cells, apical membranes are established and become cleared of cell-cell junctions, thereby allowing continuous central lumens to open. Rasip1 (Ras-interacting protein 1) is required for apical junction clearance, as well as for regulation of Rho GTPase (enzyme that hydrolyzes GTP) activity. However, it remains unknown how activities of different Rho GTPases are coordinated by Rasip1 to direct tubulogenesis. OBJECTIVE The aim of this study is to determine the mechanisms downstream of Rasip1 that drive vascular tubulogenesis. METHODS AND RESULTS Using conditional mouse mutant models and pharmacological approaches, we dissect GTPase pathways downstream of Rasip1. We show that clearance of endothelial cell apical junctions during vascular tubulogenesis depends on Rasip1, as well as the GTPase Cdc42 (cell division control protein 42 homolog) and the kinase Pak4 (serine/threonine-protein kinase 4). Genetic deletion of Rasip1 or Cdc42, or inhibition of Pak4, all blocks endothelial cell tubulogenesis. By contrast, inactivation of RhoA (Ras homologue gene family member A) signaling leads to vessel overexpansion, implicating actomyosin contractility in control of lumen diameter. Interestingly, blocking activity of NMII (nonmuscle myosin II) either before, or after, lumen morphogenesis results in dramatically different tubulogenesis phenotypes, suggesting time-dependent roles. CONCLUSIONS Rasip1 controls different pools of GTPases, which in turn regulate different pools of NMII to coordinate junction clearance (remodeling) and actomyosin contractility during vascular tubulogenesis. Rasip1 promotes activity of Cdc42 to activate Pak4, which in turn activates NMII, clearing apical junctions. Once lumens open, Rasip1 suppresses actomyosin contractility via inhibition of RhoA by Arhgap29, allowing controlled expansion of vessel lumens during embryonic growth. These findings elucidate the stepwise processes regulated by Rasip1 through downstream Rho GTPases and NMII.
Collapse
Affiliation(s)
- David M Barry
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Yeon Koo
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Pieter R Norden
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Lyndsay A Wylie
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Ke Xu
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Chonlarat Wichaidit
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - D Berfin Azizoglu
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Yi Zheng
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Melanie H Cobb
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - George E Davis
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.)
| | - Ondine Cleaver
- From the Department of Molecular Biology and Center for Regenerative Science and Medicine (D.M.B., Y.K., K.X., D.B.A., O.C.) and Department of Pharmacology (C.W., M.H.C.), University of Texas Southwestern Medical Center, Dallas; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia (P.R.N., G.E.D.); Department of Biology, University of North Carolina, Chapel Hill (L.A.W.); and Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, OH (Y.Z.).
| |
Collapse
|
29
|
The Role of RhoJ in Endothelial Cell Biology and Tumor Pathology. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6386412. [PMID: 27556037 PMCID: PMC4983353 DOI: 10.1155/2016/6386412] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/24/2016] [Accepted: 06/15/2016] [Indexed: 02/04/2023]
Abstract
Background. RhoJ, an endothelially expressed member of Cdc42 (cell division cycle 42) subfamily of Rho GTPase, plays an important role in endocytic pathway, adipocyte differentiation, endothelial motility, tube formation, and focal adhesion. RhoJ is a selective and effective therapeutic target in tumor tissues or retinopathy. Methods. A systematic review was related to “small Rho GTPase” or “RhoJ” with “endothelial motility, tube formation and focal adhesion” and “tumor therapy”. This led to many cross-references involving RhoJ and these data have been incorporated into the following study. Results. We have grouped the role of RhoJ according to three main effects: RhoJ regulates endocytic pathway and adipocyte differentiation in early studies, and RhoJ shows an important role in endothelial cell biology; furthermore, RhoJ blockade serves as a target in tumor vasculature and enhances the effects of anticancer drug. Conclusions. More research is necessary to understand the role of RhoJ in many aspects, on the basis of current knowledge of the role of RhoJ blockade in tumor vessels, there are opportunities for the therapy of tumor, and RhoJ is expressed outside tumour vasculature and is involved in wound healing. Taking advantage of the opportunities could result in a development in tumor therapy.
Collapse
|
30
|
Brinkmann BF, Steinbacher T, Hartmann C, Kummer D, Pajonczyk D, Mirzapourshafiyi F, Nakayama M, Weide T, Gerke V, Ebnet K. VE-cadherin interacts with cell polarity protein Pals1 to regulate vascular lumen formation. Mol Biol Cell 2016; 27:2811-21. [PMID: 27466317 PMCID: PMC5025268 DOI: 10.1091/mbc.e16-02-0127] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
Blood vessel tubulogenesis requires the establishment of apicobasal polarity of endothelial cells. A novel interaction is described of the cell adhesion molecule VE-cadherin with the cell polarity protein Pals1. The activity of VE-cadherin in regulation of endothelial lumen formation depends on its interaction with both Pals1 and Par3. Blood vessel tubulogenesis requires the formation of stable cell-to-cell contacts and the establishment of apicobasal polarity of vascular endothelial cells. Cell polarity is regulated by highly conserved cell polarity protein complexes such as the Par3-aPKC-Par6 complex and the CRB3-Pals1-PATJ complex, which are expressed by many different cell types and regulate various aspects of cell polarity. Here we describe a functional interaction of VE-cadherin with the cell polarity protein Pals1. Pals1 directly interacts with VE-cadherin through a membrane-proximal motif in the cytoplasmic domain of VE-cadherin. VE-cadherin clusters Pals1 at cell–cell junctions. Mutating the Pals1-binding motif in VE-cadherin abrogates the ability of VE-cadherin to regulate apicobasal polarity and vascular lumen formation. In a similar way, deletion of the Par3-binding motif at the C-terminus of VE-cadherin impairs apicobasal polarity and vascular lumen formation. Our findings indicate that the biological activity of VE-cadherin in regulating endothelial polarity and vascular lumen formation is mediated through its interaction with the two cell polarity proteins Pals1 and Par3.
Collapse
Affiliation(s)
- Benjamin F Brinkmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Tim Steinbacher
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany
| | - Christian Hartmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Denise Pajonczyk
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany
| | - Fatemeh Mirzapourshafiyi
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Masanori Nakayama
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Weide
- Department of Internal Medicine D, Division of Molecular Nephrology, University Hospital Münster, Albert-Schweitzer-Campus 1, University of Münster, 48419 Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, 48419 Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| |
Collapse
|
31
|
Soulavie F, Sundaram MV. Auto-fusion and the shaping of neurons and tubes. Semin Cell Dev Biol 2016; 60:136-145. [PMID: 27436685 DOI: 10.1016/j.semcdb.2016.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
Abstract
Cells adopt specific shapes that are necessary for specific functions. For example, some neurons extend elaborate arborized dendrites that can contact multiple targets. Epithelial and endothelial cells can form tiny seamless unicellular tubes with an intracellular lumen. Recent advances showed that cells can auto-fuse to acquire those specific shapes. During auto-fusion, a cell merges two parts of its own plasma membrane. In contrast to cell-cell fusion or macropinocytic fission, which result in the merging or formation of two separate membrane bound compartments, auto-fusion preserves one compartment, but changes its shape. The discovery of auto-fusion in C. elegans was enabled by identification of specific protein fusogens, EFF-1 and AFF-1, that mediate cell-cell fusion. Phenotypic characterization of eff-1 and aff-1 mutants revealed that fusogen-mediated fusion of two parts of the same cell can be used to sculpt dendritic arbors, reconnect two parts of an axon after injury, or form a hollow unicellular tube. Similar auto-fusion events recently were detected in vertebrate cells, suggesting that auto-fusion could be a widely used mechanism for shaping neurons and tubes.
Collapse
Affiliation(s)
- Fabien Soulavie
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104,United States
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104,United States.
| |
Collapse
|
32
|
Sundaram MV, Cohen JD. Time to make the doughnuts: Building and shaping seamless tubes. Semin Cell Dev Biol 2016; 67:123-131. [PMID: 27178486 DOI: 10.1016/j.semcdb.2016.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/04/2016] [Accepted: 05/06/2016] [Indexed: 10/21/2022]
Abstract
A seamless tube is a very narrow-bore tube that is composed of a single cell with an intracellular lumen and no adherens or tight junctions along its length. Many capillaries in the vertebrate vascular system are seamless tubes. Seamless tubes also are found in invertebrate organs, including the Drosophila trachea and the Caenorhabditis elegans excretory system. Seamless tube cells can be less than a micron in diameter, and they can adopt very simple "doughnut-like" shapes or very complex, branched shapes comparable to those of neurons. The unusual topology and varied shapes of seamless tubes raise many basic cell biological questions about how cells form and maintain such structures. The prevalence of seamless tubes in the vascular system means that answering such questions has significant relevance to human health. In this review, we describe selected examples of seamless tubes in animals and discuss current models for how seamless tubes develop and are shaped, focusing particularly on insights that have come from recent studies in Drosophila and C. elegans.
Collapse
Affiliation(s)
- Meera V Sundaram
- Dept. of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Jennifer D Cohen
- Dept. of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
|
34
|
Rasip1 is essential to blood vessel stability and angiogenic blood vessel growth. Angiogenesis 2016; 19:173-90. [PMID: 26897025 DOI: 10.1007/s10456-016-9498-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/03/2016] [Indexed: 01/09/2023]
Abstract
Cardiovascular function depends on patent, continuous and stable blood vessel formation by endothelial cells (ECs). Blood vessel development initiates by vasculogenesis, as ECs coalesce into linear aggregates and organize to form central lumens that allow blood flow. Molecular mechanisms underlying in vivo vascular 'tubulogenesis' are only beginning to be unraveled. We previously showed that the GTPase-interacting protein called Rasip1 is required for the formation of continuous vascular lumens in the early embryo. Rasip1(-/-) ECs exhibit loss of proper cell polarity and cell shape, disrupted localization of EC-EC junctions and defects in adhesion of ECs to extracellular matrix. In vitro studies showed that Rasip1 depletion in cultured ECs blocked tubulogenesis. Whether Rasip1 is required in blood vessels after their initial formation remained unclear. Here, we show that Rasip1 is essential for vessel formation and maintenance in the embryo, but not in quiescent adult vessels. Rasip1 is also required for angiogenesis in three models of blood vessel growth: in vitro matrix invasion, retinal blood vessel growth and directed in vivo angiogenesis assays. Rasip1 is thus necessary in growing embryonic blood vessels, postnatal angiogenic sprouting and remodeling, but is dispensable for maintenance of established blood vessels, making it a potential anti-angiogenic therapeutic target.
Collapse
|
35
|
Li Q, Zhang Y, Pluchon P, Robens J, Herr K, Mercade M, Thiery JP, Yu H, Viasnoff V. Extracellular matrix scaffolding guides lumen elongation by inducing anisotropic intercellular mechanical tension. Nat Cell Biol 2016; 18:311-8. [PMID: 26878396 DOI: 10.1038/ncb3310] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/08/2016] [Indexed: 02/07/2023]
Abstract
The de novo formation of secretory lumens plays an important role during organogenesis. It involves the establishment of a cellular apical pole and the elongation of luminal cavities. The molecular parameters controlling cell polarization have been heavily scrutinized. In particular, signalling from the extracellular matrix (ECM) proved essential to the proper localization of the apical pole by directed protein transport. However, little is known about the regulation of the shape and the directional development of lumen into tubes. We demonstrate that the spatial scaffolding of cells by ECM can control tube shapes and can direct their elongation. We developed a minimal organ approach comprising of hepatocyte doublets cultured in artificial microniches to precisely control the spatial organization of cellular adhesions in three dimensions. This approach revealed a mechanism by which the spatial repartition of integrin-based adhesion can elicit an anisotropic intercellular mechanical stress guiding the osmotically driven elongation of lumens in the direction of minimal tension. This mechanical guidance accounts for the different morphologies of lumen in various microenvironmental conditions.
Collapse
Affiliation(s)
- Qiushi Li
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Yue Zhang
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Perrine Pluchon
- Department of Biological Sciences, National University of Singapore, Singapore 117411, Singapore
| | - Jeffrey Robens
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Keira Herr
- Institute of Molecular Cell Biology, A∗STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Myriam Mercade
- Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés, Université de Toulouse, INSA, INRA, CNRS, 31077 Toulouse, France
| | - Jean-Paul Thiery
- Institute of Molecular Cell Biology, A∗STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore 117597, Singapore.,Institute of Bioengineering and Nanotechnology (IBN), Agency for Science, Technology and Research, Singapore 138669, Singapore
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore 117411, Singapore.,Institute of Molecular Cell Biology, A∗STAR, 61 Biopolis Drive, Singapore 138673, Singapore.,CNRS UMI3639, Singapore 117411, Singapore
| |
Collapse
|
36
|
Norden PR, Kim DJ, Barry DM, Cleaver OB, Davis GE. Cdc42 and k-Ras Control Endothelial Tubulogenesis through Apical Membrane and Cytoskeletal Polarization: Novel Stimulatory Roles for GTPase Effectors, the Small GTPases, Rac2 and Rap1b, and Inhibitory Influence of Arhgap31 and Rasa1. PLoS One 2016; 11:e0147758. [PMID: 26812085 PMCID: PMC4728208 DOI: 10.1371/journal.pone.0147758] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/07/2016] [Indexed: 01/18/2023] Open
Abstract
A critical and understudied property of endothelial cells is their ability to form lumens and tube networks. Although considerable information has been obtained concerning these issues, including the role of Cdc42 and Rac1 and their effectors such as Pak2, Pak4, Par6b, and co-regulators such as integrins, MT1-MMP and Par3; many key questions remain that are necessary to elucidate molecular and signaling requirements for this fundamental process. In this work, we identify new small GTPase regulators of EC tubulogenesis including k-Ras, Rac2 and Rap1b that act in conjunction with Cdc42 as well as the key downstream effectors, IQGAP1, MRCKβ, beta-Pix, GIT1, and Rasip1 (which can assemble into multiprotein complexes with key regulators including α2β1 integrin and MT1-MMP). In addition, we identify the negative regulators, Arhgap31 (by inactivating Cdc42 and Rac) and Rasa1 (by inactivating k-Ras) and the positive regulator, Arhgap29 (by inactivating RhoA) which play a major functional role during the EC tubulogenic process. Human EC siRNA suppression or mouse knockout of Rasip1 leads to identical phenotypes where ECs form extensive cord networks, but cannot generate lumens or tubes. Essential roles for these molecules during EC tubulogenesis include; i) establishment of asymmetric EC cytoskeletal polarization (subapical distribution of acetylated tubulin and basal membrane distribution of F-actin); and ii) directed membrane trafficking of pinocytic vacuoles or other intracellular vesicles along acetylated tubulin tracks to the developing apical membrane surface. Cdc42 co-localizes subapically with acetylated tubulin, while Rac1 and k-Ras strongly label vacuole/ vesicle membranes which accumulate and fuse together in a polarized, perinuclear manner. We observe polarized apical membrane and subapical accumulation of key GTPases and effectors regulating EC lumen formation including Cdc42, Rac1, Rac2, k-Ras, Rap1b, activated c-Raf and Rasip1 to control EC tube network assembly. Overall, this work defines novel key regulators and their functional roles during human EC tubulogenesis.
Collapse
Affiliation(s)
- Pieter R. Norden
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Dae Joong Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - David M. Barry
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Ondine B. Cleaver
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - George E. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| |
Collapse
|
37
|
Bowers S, Norden P, Davis G. Molecular Signaling Pathways Controlling Vascular Tube Morphogenesis and Pericyte-Induced Tube Maturation in 3D Extracellular Matrices. ADVANCES IN PHARMACOLOGY 2016; 77:241-80. [DOI: 10.1016/bs.apha.2016.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
The alternative splicing factor Nova2 regulates vascular development and lumen formation. Nat Commun 2015; 6:8479. [PMID: 26446569 PMCID: PMC4633719 DOI: 10.1038/ncomms9479] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/27/2015] [Indexed: 12/19/2022] Open
Abstract
Vascular lumen formation is a fundamental step during angiogenesis; yet, the molecular mechanisms underlying this process are poorly understood. Recent studies have shown that neural and vascular systems share common anatomical, functional and molecular similarities. Here we show that the organization of endothelial lumen is controlled at the post-transcriptional level by the alternative splicing (AS) regulator Nova2, which was previously considered to be neural cell-specific. Nova2 is expressed during angiogenesis and its depletion disrupts vascular lumen formation in vivo. Similarly, Nova2 depletion in cultured endothelial cells (ECs) impairs the apical distribution and the downstream signalling of the Par polarity complex, resulting in altered EC polarity, a process required for vascular lumen formation. These defects are linked to AS changes of Nova2 target exons affecting the Par complex and its regulators. Collectively, our results reveal that Nova2 functions as an AS regulator in angiogenesis and is a novel member of the ‘angioneurins' family. The alternative splicing factor Nova2 is best known for its pivotal function in the brain. Giampietro et al. reveal an important role for Nova2 in the regulation of alternative splicing of transcripts in the vascular endothelium that are crucial for the maintenance of endothelial cell polarity and vessel lumen formation in zebrafish.
Collapse
|
39
|
Masci VL, Taddei AR, Gambellini G, Giorgi F, Fausto AM. Ultrastructural investigation on fibroblast interaction with collagen scaffold. J Biomed Mater Res A 2015; 104:272-82. [PMID: 26375405 DOI: 10.1002/jbm.a.35563] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/25/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
Collagen-based scaffolds are used as temporary or permanent coverings to help wound healing. Under natural conditions, wound healing is affected by such factors as cell types, growth factors and several components of the extracellular matrix. Due to the complexity of the cell-to-matrix interaction, many cell based mechanisms regulating wound healing in vivo are not yet properly understood. However, the whole process can be partially simulated in vitro to determine how cells interact with the collagen scaffold in relation to such features as physico-chemical properties, matrix architecture and fiber stability. Under these conditions, cell migration into the collagen matrix can be easily assessed and causally correlated with these features. In this study, we aimed at providing a structural analysis of how NIH3T3 fibroblasts migrate and proliferate in vitro when seeded on a native type-I collagen scaffold. To this end, samples were collected at regular time intervals and analyzed by light microscopy (LM), scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Through this experimental approach we demonstrate that collagen is gradually frayed into progressively thinner fibrils as fibroblasts migrate into the matrix, embrace the collagen fibers with long filopodia and form large intracellular vacuoles. A key role in this process is also played by microvesicles shed from the fibroblast plasma membrane and spread over long distances inside the collagen matrix. These observations indicate that a native type-I equine collagen provides favorable conditions for simulating collagen processing in vitro and eventually for unraveling the mechanisms controlling cell uptake and intracellular degradation.
Collapse
Affiliation(s)
- Valentina Laghezza Masci
- Department for Innovation in Biological, Agrifood and Forestry Systems, Tuscia University, Viterbo, 01100, Italy
| | - Anna Rita Taddei
- Section of Electron Microscopy, Great Equipment Center, Tuscia University, Viterbo, 01100, Italy
| | - Gabriella Gambellini
- Section of Electron Microscopy, Great Equipment Center, Tuscia University, Viterbo, 01100, Italy
| | | | - Anna Maria Fausto
- Department for Innovation in Biological, Agrifood and Forestry Systems, Tuscia University, Viterbo, 01100, Italy
| |
Collapse
|
40
|
Activation of Apoptotic Signal in Endothelial Cells through Intracellular Signaling Molecules Blockade in Tumor-Induced Angiogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:908757. [PMID: 26346668 PMCID: PMC4539440 DOI: 10.1155/2015/908757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/07/2015] [Accepted: 06/11/2015] [Indexed: 11/25/2022]
Abstract
Tumor-induced angiogenesis is the bridge between avascular and vascular tumor growth phases. In tumor-induced angiogenesis, endothelial cells start to migrate and proliferate toward the tumor and build new capillaries toward the tumor. There are two stages for sprout extension during angiogenesis. The first stage is prior to anastomosis, when single sprouts extend. The second stage is after anastomosis when closed flow pathways or loops are formed and blood flows in the closed loops. Prior to anastomosis, biochemical and biomechanical signals from extracellular matrix regulate endothelial cell phenotype; however, after anastomosis, blood flow is the main regulator of endothelial cell phenotype. In this study, the critical signaling pathways of each stage are introduced. A Boolean network model is used to map environmental and flow induced signals to endothelial cell phenotype (proliferation, migration, apoptosis, and lumen formation). Using the Boolean network model, blockade of intracellular signaling molecules of endothelial cell is investigated prior to and after anastomosis and the cell fate is obtained in each case. Activation of apoptotic signal in endothelial cell can prevent the extension of new vessels and may inhibit angiogenesis. It is shown that blockade of a few signaling molecules in endothelial cell activates apoptotic signal that are proposed as antiangiogenic strategies.
Collapse
|
41
|
Yu JA, Castranova D, Pham VN, Weinstein BM. Single-cell analysis of endothelial morphogenesis in vivo. Development 2015; 142:2951-61. [PMID: 26253401 PMCID: PMC4582182 DOI: 10.1242/dev.123174] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/27/2015] [Indexed: 12/19/2022]
Abstract
Vessel formation has been extensively studied at the tissue level, but the difficulty in imaging the endothelium with cellular resolution has hampered study of the morphogenesis and behavior of endothelial cells (ECs) in vivo. We are using endothelial-specific transgenes and high-resolution imaging to examine single ECs in zebrafish. By generating mosaics with transgenes that simultaneously mark endothelial nuclei and membranes we are able to definitively identify and study the morphology and behavior of individual ECs during vessel sprouting and lumen formation. Using these methods, we show that developing trunk vessels are composed of ECs of varying morphology, and that single-cell analysis can be used to quantitate alterations in morphology and dynamics in ECs that are defective in proper guidance and patterning. Finally, we use single-cell analysis of intersegmental vessels undergoing lumen formation to demonstrate the coexistence of seamless transcellular lumens and single or multicellular enclosed lumens with autocellular or intercellular junctions, suggesting that heterogeneous mechanisms contribute to vascular lumen formation in vivo. The tools that we have developed for single EC analysis should facilitate further rigorous qualitative and quantitative analysis of EC morphology and behavior in vivo.
Collapse
Affiliation(s)
- Jianxin A Yu
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
42
|
The Formin FMNL3 Controls Early Apical Specification in Endothelial Cells by Regulating the Polarized Trafficking of Podocalyxin. Curr Biol 2015; 25:2325-31. [PMID: 26299518 DOI: 10.1016/j.cub.2015.07.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/27/2015] [Accepted: 07/16/2015] [Indexed: 01/11/2023]
Abstract
Angiogenesis is the fundamental process by which new blood vessels form from pre-existing vasculature. It plays a critical role in the formation of the vasculature during development and is triggered in response to tissue hypoxia in adult organisms. This process requires complex and coordinated regulation of the endothelial cell cytoskeleton to control cell shape and polarity. In our previous work, we showed that the cytoskeletal regulator FMNL3/FRL2 controls the alignment of stabilized microtubules during polarized endothelial cell elongation and that depletion of FMNL3 retards elongation of the intersegmental vessels in zebrafish. Recent work has shown that FMNL3 is also needed for vascular lumen formation, a critical element of the formation of functional vessels. Here, we show that FMNL3 interacts with Cdc42 and RhoJ, two Rho family GTPases known to be required for lumen formation. FMNL3 and RhoJ are concentrated at the early apical surface, or AMIS, and regulate the formation of radiating actin cables from this site. In diverse biological systems, formins mediate polarized trafficking through the generation of similar actin filaments tracks. We show that FMNL3 and RhoJ are required for polarized trafficking of podocalyxin to the early apical surface--an important event in vascular lumenogenesis.
Collapse
|
43
|
WU WENJUAN, ZHANG XIZHI, LV HOUNING, LIAO YUEXIA, ZHANG WEICHENG, CHENG HAICHAO, DENG ZIJING, SHEN JINGYUAN, YUAN QING, ZHANG YU, SHEN WEIGAN. Identification of immediate early response protein 2 as a regulator of angiogenesis through the modulation of endothelial cell motility and adhesion. Int J Mol Med 2015; 36:1104-10. [DOI: 10.3892/ijmm.2015.2310] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 08/07/2015] [Indexed: 11/06/2022] Open
|
44
|
Barry DM, Xu K, Meadows SM, Zheng Y, Norden PR, Davis GE, Cleaver O. Cdc42 is required for cytoskeletal support of endothelial cell adhesion during blood vessel formation in mice. Development 2015; 142:3058-70. [PMID: 26253403 DOI: 10.1242/dev.125260] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 07/29/2015] [Indexed: 12/11/2022]
Abstract
The Rho family of small GTPases has been shown to be required in endothelial cells (ECs) during blood vessel formation. However, the underlying cellular events controlled by different GTPases remain unclear. Here, we assess the cellular mechanisms by which Cdc42 regulates mammalian vascular morphogenesis and maintenance. In vivo deletion of Cdc42 in embryonic ECs (Cdc42(Tie2KO)) results in blocked lumen formation and endothelial tearing, leading to lethality of mutant embryos by E9-10 due to failed blood circulation. Similarly, inducible deletion of Cdc42 (Cdc42(Cad5KO)) at mid-gestation blocks angiogenic tubulogenesis. By contrast, deletion of Cdc42 in postnatal retinal vessels leads to aberrant vascular remodeling and sprouting, as well as markedly reduced filopodia formation. We find that Cdc42 is essential for organization of EC adhesion, as its loss results in disorganized cell-cell junctions and reduced focal adhesions. Endothelial polarity is also rapidly lost upon Cdc42 deletion, as seen by failed localization of apical podocalyxin (PODXL) and basal actin. We link observed failures to a defect in F-actin organization, both in vitro and in vivo, which secondarily impairs EC adhesion and polarity. We also identify Cdc42 effectors Pak2/4 and N-WASP, as well as the actomyosin machinery, to be crucial for EC actin organization. This work supports the notion of Cdc42 as a central regulator of the cellular machinery in ECs that drives blood vessel formation.
Collapse
Affiliation(s)
- David M Barry
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Ke Xu
- Department SCRB, Harvard University, Cambridge, MA 02138, USA
| | - Stryder M Meadows
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA 70118, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Pieter R Norden
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
45
|
Bazmara H, Soltani M, Sefidgar M, Bazargan M, Mousavi Naeenian M, Rahmim A. Blood flow and endothelial cell phenotype regulation during sprouting angiogenesis. Med Biol Eng Comput 2015; 54:547-58. [DOI: 10.1007/s11517-015-1341-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/01/2015] [Indexed: 11/24/2022]
|
46
|
Chaki SP, Barhoumi R, Rivera GM. Actin remodeling by Nck regulates endothelial lumen formation. Mol Biol Cell 2015; 26:3047-60. [PMID: 26157164 PMCID: PMC4551318 DOI: 10.1091/mbc.e15-06-0338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/01/2015] [Indexed: 01/03/2023] Open
Abstract
Nck-dependent actin remodeling enables endothelial morphogenesis by promoting cell elongation and proper organization of VE-cadherin intercellular junctions. Nck determines spatiotemporal patterns of Cdc42/aPKC activation to regulate endothelial apical-basal polarity and lumen formation. Multiple angiogenic cues modulate phosphotyrosine signaling to promote vasculogenesis and angiogenesis. Despite its functional and clinical importance, how vascular cells integrate phosphotyrosine-dependent signaling to elicit cytoskeletal changes required for endothelial morphogenesis remains poorly understood. The family of Nck adaptors couples phosphotyrosine signals with actin dynamics and therefore is well positioned to orchestrate cellular processes required in vascular formation and remodeling. Culture of endothelial cells in three-dimensional collagen matrices in the presence of VEGF stimulation was combined with molecular genetics, optical imaging, and biochemistry to show that Nck-dependent actin remodeling promotes endothelial cell elongation and proper organization of VE-cadherin intercellular junctions. Major morphogenetic defects caused by abrogation of Nck signaling included loss of endothelial apical-basal polarity and impaired lumenization. Time-lapse imaging using a Förster resonance energy transfer biosensor, immunostaining with phospho-specific antibodies, and GST pull-down assays showed that Nck determines spatiotemporal patterns of Cdc42/aPKC activation during endothelial morphogenesis. Our results demonstrate that Nck acts as an important hub integrating angiogenic cues with cytoskeletal changes that enable endothelial apical-basal polarization and lumen formation. These findings point to Nck as an emergent target for effective antiangiogenic therapy.
Collapse
Affiliation(s)
- Sankar P Chaki
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843-4467
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4467
| | - Gonzalo M Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843-4467
| |
Collapse
|
47
|
AECHL-1, a novel triterpenoid, targets tumor neo-vasculature and impairs the endothelial cell cytoskeleton. Angiogenesis 2015; 18:283-99. [PMID: 25952529 PMCID: PMC4472952 DOI: 10.1007/s10456-015-9466-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/29/2015] [Indexed: 11/26/2022]
Abstract
Tumor angiogenesis is characterized by abnormal vessel morphology leading to erratic and insufficient delivery of chemotherapeutics and oxygen, making the tumor core not only highly hypoxic but also unresponsive toward treatment. Such hypoxic conditions promote tumor aggressiveness, leading to the establishment of metastatic disease. Most anti-angiogenic treatments aim toward the destruction of tumor vasculature, which proves countereffective by further increasing its aggressive nature. Hence, developing drugs which target or regulate these processes might lead to a better delivery of chemotherapeutics resulting in tumor shrinkage. Plant-derived natural compounds having a bioactive ingredient, especially triterpenoids, have been known to possess anticancer properties. AECHL-1, a recently isolated novel triterpenoid with proven anticancer potential, is seemingly noncytotoxic toward HEK 293 and HUVECs. Also, cytotoxicity was absent during in vivo studies involving intraperitoneal injections with 5 µg/kg body weight AECHL-1 on SCID mice. When used at subtoxic doses, it was found to be effective in suppression of neo-vessel formation as demonstrated in the chick chorioallantoic membrane, rat aortic rings, Matrigel plugs and xenograft tumors implanted in SCID mice. Tumor vasculature from AECHL-1-treated mice showed greater mural cell coverage and relatively normalized architecture. Investigations into the molecular mechanisms responsible for these observations revealed an effect on the actin cytoskeleton of stimulated HUVECs as well as the VEGFR2-mediated MAPK pathway. AECHL-1 could effectively distinguish between stimulated and nonstimulated endothelial cells. AECHL-1 could also downregulate HIF-1α expression and VEGF secretion under hypoxic conditions, thus reducing the fears of unnecessarily aggravating tumor metastasis as a result of anti-angiogenic therapy. Results obtained from the aforementioned studies make it clear that though AECHL-1 shows promise in discouraging and pruning neo-vasculature, it may not affect existing vasculature as the doses used for the assays are significantly lower than the ones causing endothelial cell death and has potential to be considered as a candidate for therapeutic drug development.
Collapse
|
48
|
Laurenzana A, Fibbi G, Chillà A, Margheri G, Del Rosso T, Rovida E, Del Rosso M, Margheri F. Lipid rafts: integrated platforms for vascular organization offering therapeutic opportunities. Cell Mol Life Sci 2015; 72:1537-57. [PMID: 25552244 PMCID: PMC11113367 DOI: 10.1007/s00018-014-1814-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/12/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023]
Abstract
Research on the nanoscale membrane structures known as lipid rafts is relevant to the fields of cancer biology, inflammation and ischaemia. Lipid rafts recruit molecules critical to signalling and regulation of the invasion process in malignant cells, the leukocytes that provide immunity in inflammation and the endothelial cells that build blood and lymphatic vessels, as well as the patterning of neural networks. As angiogenesis is a common denominator, regulation of receptors and signalling molecules critical to angiogenesis is central to the design of new approaches aimed at reducing, promoting or normalizing the angiogenic process. The goal of this review is to highlight some of the key issues that indicate the involvement of endothelial cell lipid rafts at each step of so-called 'sprouting angiogenesis', from stimulation of the vascular endothelial growth factor to the choice of tip cells, activation of migratory and invasion pathways, recruitment of molecules that guide axons in vascular patterning and maturation of blood vessels. Finally, the review addresses opportunities for future studies to define how these lipid domains (and their constituents) may be manipulated to stimulate the so-called 'normalization' of vascular networks within tumors, and be identified as the main target, enabling the development of more efficient chemotherapeutics and cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Laurenzana
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Gabriella Fibbi
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Anastasia Chillà
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Giancarlo Margheri
- Institute of Complex Systems (ISC), Consiglio Nazionale delle Ricerche (CNR), Florence, Italy
| | - Tommaso Del Rosso
- Department of Physics, Pontificia Universidade Catolica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisabetta Rovida
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Mario Del Rosso
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
- Istituto Toscano Tumori, Florence, Italy
| | - Francesca Margheri
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
49
|
Charpentier MS, Tandon P, Trincot CE, Koutleva EK, Conlon FL. A distinct mechanism of vascular lumen formation in Xenopus requires EGFL7. PLoS One 2015; 10:e0116086. [PMID: 25705891 PMCID: PMC4338030 DOI: 10.1371/journal.pone.0116086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/04/2014] [Indexed: 01/03/2023] Open
Abstract
During vertebrate blood vessel development, lumen formation is the critical process by which cords of endothelial cells transition into functional tubular vessels. Here, we use Xenopus embryos to explore the cellular and molecular mechanisms underlying lumen formation of the dorsal aorta and the posterior cardinal veins, the primary major vessels that arise via vasculogenesis within the first 48 hours of life. We demonstrate that endothelial cells are initially found in close association with one another through the formation of tight junctions expressing ZO-1. The emergence of vascular lumens is characterized by elongation of endothelial cell shape, reorganization of junctions away from the cord center to the periphery of the vessel, and onset of Claudin-5 expression within tight junctions. Furthermore, unlike most vertebrate vessels that exhibit specialized apical and basal domains, we show that early Xenopus vessels are not polarized. Moreover, we demonstrate that in embryos depleted of the extracellular matrix factor Epidermal Growth Factor-Like Domain 7 (EGFL7), an evolutionarily conserved factor associated with vertebrate vessel development, vascular lumens fail to form. While Claudin-5 localizes to endothelial tight junctions of EGFL7-depleted embryos in a timely manner, endothelial cells of the aorta and veins fail to undergo appropriate cell shape changes or clear junctions from the cell-cell contact. Taken together, we demonstrate for the first time the mechanisms by which lumens are generated within the major vessels in Xenopus and implicate EGFL7 in modulating cell shape and cell-cell junctions to drive proper lumen morphogenesis.
Collapse
Affiliation(s)
- Marta S. Charpentier
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Panna Tandon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Claire E. Trincot
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Elitza K. Koutleva
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Frank L. Conlon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
50
|
Rytlewski JA, Alejandra Aldon M, Lewis EW, Suggs LJ. Mechanisms of tubulogenesis and endothelial phenotype expression by MSCs. Microvasc Res 2015; 99:26-35. [PMID: 25711526 DOI: 10.1016/j.mvr.2015.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 12/03/2014] [Accepted: 02/11/2015] [Indexed: 11/17/2022]
Abstract
Stem cell-based therapies are a promising new avenue for treating ischemic disease and chronic wounds. Mesenchymal stem cells (MSCs) have a proven ability to augment the neovascularization processes necessary for wound healing and are widely popular as an autologous source of progenitor cells. Our lab has previously reported on PEGylated fibrin as a unique hydrogel that promotes spontaneous tubulogenesis of encapsulated MSCs without exogenous factors. However, the mechanisms underlying this process have remained unknown. To better understand the therapeutic value of PEGylated fibrin delivery of MSCs, we sought to clarify the relationship between biomaterial properties and cell behavior. Here we find that fibrin PEGylation does not dramatically alter the macroscopic mechanical properties of the fibrin-based matrix (less than 10% difference). It does, however, dramatically reduce the rate of diffusion through the gel matrix. PEGylated fibrin enhances the tubulogenic growth of encapsulated MSCs demonstrating fluid-filled lumens by interconnected MSCs. Image analysis gave a value of 4320 ± 1770 μm total network length versus 618 ± 443 μm for unmodified fibrin. PEGylation promotes the endothelial phenotype of encapsulated MSCs--compared to unmodified fibrin--as evidenced by higher levels of endothelial markers (von Willebrand factor, 2.2-fold; vascular endothelial cadherin, 1.8-fold) and vascular endothelial growth factor (VEGF, up to 1.8-fold). Prospective analysis of underlying molecular pathways demonstrated that this endothelial-like MSC behavior is sensitively modulated by hypoxic stress, but not VEGF supplementation as evidenced by a significant increase in VEGF and MMP-2 secretion per cell under hypoxia. Further gain-of-function studies under hypoxic stress demonstrated that hypoxia culture of MSCs in unmodified fibrin could increase both vWF and VE-cadherin levels to values that were not significantly different than cells cultured in PEGylated fibrin. This result corroborated our hypothesis that the diffusion-limited environment of PEGylated fibrin is augmenting endothelial differentiation cues provided by unmodified fibrin. However, MSC networks lack platelet endothelial cell adhesion molecule-1 (PECAM-1) expression, which indicates incomplete differentiation towards an endothelial cell type. Collectively, the data here supports a revised understanding of MSC-derived neovascularization that contextualizes their behavior and utility as a hybrid endothelial-stromal cell type, with mixed characteristics of both populations.
Collapse
Affiliation(s)
- Julie A Rytlewski
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Stop C0800, Austin, TX 78712, USA.
| | - M Alejandra Aldon
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Stop C0800, Austin, TX 78712, USA.
| | - Evan W Lewis
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Stop C0800, Austin, TX 78712, USA.
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Stop C0800, Austin, TX 78712, USA.
| |
Collapse
|