1
|
Gucciardo F, Lebeau A, Pirson S, Buntinx F, Ivanova E, Blacher S, Brouillard P, Deroye J, Baudin L, Pirnay A, Morfoisse F, Villette C, Nizet C, Lallemand F, Munaut C, Alitalo K, Geris L, Vikkula M, Gautier-Isola M, Noel A. Targeting uPARAP Modifies Lymphatic Vessel Architecture and Attenuates Lymphedema. Circulation 2025; 151:1412-1429. [PMID: 40035133 PMCID: PMC12063686 DOI: 10.1161/circulationaha.124.072093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Lymphedema is an incurable disease associated with lymphatic dysfunction that causes tissue swelling and fibrosis. We investigated whether lymphedema could be attenuated by interfering with uPARAP (urokinase plasminogen activator receptor-associated protein; Mrc2 gene), an endocytic receptor involved in fibrosis and lymphangiogenesis. METHODS We generated mice with lymphatic endothelial cell (LEC)-specific uparap deficiency and compared them with constitutive knockout mice by applying a preclinical model of secondary lymphedema (SL). Computerized methods were applied for 2-dimensional and 3-dimensional image quantifications. Cellular effects of uPARAP deletion on lymphatic permeability were assessed by small interfering RNA-mediated silencing in human dermal LECs and a pharmacologic treatment targeting ROCK (Rho-associated coiled coil containing kinase), an established regulator of cell junctions. The uPARAP and vascular endothelial cadherin partnership was investigated through proximity ligation assay, coimmunoprecipitation, and immunostaining. An in silico model was generated to analyze the fluid-absorbing function of the lymphatic vasculature. To interfere with uPARAP, its downregulation was achieved in vivo through a gapmer approach. RESULTS uparap deficiency mitigated several key pathologic features of SL, including hindlimb swelling, epidermal thickening, and the accumulation and size of adipocytes. In both global and LEC-conditional uparap-deficient mice, induction of SL led to a distinctive labyrinthine vasculature, defined herein by twisted and hyperbranched vessels with overlapping cells. This topology, mainly composed of pre-collecting vessels, correlated with reduced SL, but not with change in fibrosis, highlighting the importance of uPARAP in regulating LEC functions in a lymphedematous context. In vitro, uPARAP knockdown in LECs impaired vascular endothelial growth factor C-mediated endosomal trafficking of vascular endothelial cadherin and induced overlapping cell junctions. The pharmacologic inhibition of ROCK recapitulated cell superimposition in vitro and the labyrinthine vasculature in vivo with attenuated SL. Computational modeling of labyrinthine lymphatic vasculature supported the observation on their improved fluid-absorbing function in comparison with a normal hierarchic network. These data provide proof of concept of inducing a labyrinthine topology to treat SL. For therapeutic purposes, we validated the use of an anti-uPARAP gapmer to induce a labyrinthine vasculature and attenuate SL formation. CONCLUSIONS Our findings provide evidence that downregulating uPARAP expression can induce a beneficial remodeling of lymphatic vasculature that attenuates lymphedema through a cell junction-based mechanism, offering a novel therapeutic pathway for lymphedema.
Collapse
Affiliation(s)
- Fabrice Gucciardo
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Alizée Lebeau
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Sébastien Pirson
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Florence Buntinx
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Elitsa Ivanova
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Silvia Blacher
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium (P.B., M.V.)
| | - Jonathan Deroye
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Louis Baudin
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Alexandra Pirnay
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Florent Morfoisse
- U1297-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, France (F.M.)
| | - Claire Villette
- Biomechanics Research Unit Department, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Belgium (C.V., L.G.)
| | - Christophe Nizet
- Departments of Plastic and Reconstructive Surgery (C.N.), University of Liège, Sart-Tilman, Belgium
| | | | - Carine Munaut
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum, University of Helsinki, Finland (K.A.)
| | - Liesbet Geris
- Biomechanics Research Unit Department, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Belgium (C.V., L.G.)
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium (P.B., M.V.)
- WELBIO Department, WEL Research Institute, Wavre, Belgium (M.V., A.N.)
| | - Marine Gautier-Isola
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Agnès Noel
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium (M.V., A.N.)
| |
Collapse
|
2
|
Wang L, Wang X, Wu J, Chen J, He Z, Wang J, Zhang X. Magnesium Ions Induce Endothelial Cell Differentiation into Tip Cell and Enhance Vascularized Bone Regeneration. Adv Healthc Mater 2025:e2500274. [PMID: 40346783 DOI: 10.1002/adhm.202500274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/07/2025] [Indexed: 05/12/2025]
Abstract
Vascularization has been considered an essential strategy for bone regeneration and can be promoted by magnesium ions (Mg2+). During angiogenesis, the differentiation of endothelial cells (ECs) into tip cell is a critical step since it controls the growth direction and pattern of new vascular sprouts. While several studies have noted the pro-angiogenic effects of Mg2+, however, their specific influence on tip cell formation is unclear. Therefore, this research seeks to examine the impact of Mg2+ on tip cells and elucidate the potential mechanisms involved. The results reveal that Mg2+ shows good compatibility and stimulates ECs to migrate and invade in vitro. Moreover, Mg2+ enhances EC spheroids sprouting and elevates the expression of genes linked to tip cells. The underlying mechanisms are that Mg2+ facilitates tip cell differentiation via the VEGFA-VEGFR2/Notch1 signaling pathway crosstalk and promotes migration and filopodia formation of tip cells and proliferation of stalk cells by inducing YAP nuclear translocation, culminating in the maturation of vascular networks. Furthermore, EC spheroids stimulated by Mg2+ load in hydrogel enhance vascularized bone regeneration in vivo. These findings enrich the understanding of how Mg2+ influence blood vessel formation and provide practical strategies for the development and design of magnesium-based biomaterials.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xu Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jicenyuan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zihan He
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Prosthodontics and Implantology, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Aldirawi M, Ghanbari P, Mietkowska M, März S, Odenthal-Schnittler M, Franz J, Wegner J, Currie S, Kipcke JP, Taha M, Giglmaier M, Blanque A, Schillers H, Raz E, Vestweber D, Rottner K, Schnittler H. A specific role for endothelial EPLIN-isoform-regulated actin dynamics in neutrophil transmigration. Sci Rep 2025; 15:15698. [PMID: 40325158 PMCID: PMC12053001 DOI: 10.1038/s41598-025-98192-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/09/2025] [Indexed: 05/07/2025] Open
Abstract
Proinflammatory cytokines such as TNF-α or IL-1β activate the endothelium promoting leukocyte transendothelial migration (TEM) via expression of cell adhesion molecules (CAM) and cause actin remodelling. However, the function of endothelial actin remodelling in TEM remains elusive, despite its involvement in the formation of docking structures, diapedesis pores and pore resealing. Here, we establish EPLIN-isoforms, EPLIN-β and EPLIN-α, as differential regulators of TNF-α-inducedactin-remodelling significantly affecting TEM. We find EPLIN-β-induced stress fiber formation upon TNF-α-treatment weakens endothelial junctions, upregulates junctional dynamics and facilitates intercellular gaps for TEM. Increased junctional dynamics involves branched actin filaments under the control of EPLIN-α, including docking structure formation and transmigratory pore closure. We further establish by EPLIN deletion and re-expression studies that EPLIN-α-mediated termination of branched actin filaments maintains TNF-α-induced junctional dynamics and intercellular gaps facilitating TEM. These findings highlight the critical role of TNF-α-induced differential actin dynamics, controlled by EPLIN isoforms, in TEM. These results also offer a wider understanding of inflammation-induced TEM by incorporating altered junctional dynamics alongside upregulation of cell adhesion molecules.
Collapse
Affiliation(s)
- Mohammed Aldirawi
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
| | - Parisa Ghanbari
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Magdalena Mietkowska
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Sigrid März
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
| | - Maria Odenthal-Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany
| | - Jonas Franz
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany
| | - Julian Wegner
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, 48149, Münster, Germany
| | - Silke Currie
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Jan Philip Kipcke
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany
| | - Muna Taha
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
| | - Marcus Giglmaier
- Institute of Aerodynamics and Fluid Mechanics, Technical University of Munich, Boltzmannstr. 15, 85748, Garching, Germany
| | - Anja Blanque
- Institute of Physiology, University Münster, Robert-Koch Strasse 27a, 48149, Münster, Germany
| | - Hermann Schillers
- Institute of Physiology, University Münster, Robert-Koch Strasse 27a, 48149, Münster, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, 48149, Münster, Germany
| | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany.
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany.
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany.
| |
Collapse
|
4
|
Han J, Halwachs K, West T, Larsen B, Sacks MS, Rosales AM, Zoldan J. Matrix Stiffness Regulates Mechanotransduction and Vascular Network Formation of hiPSC-Derived Endothelial Progenitors Encapsulated in 3D Hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648340. [PMID: 40291699 PMCID: PMC12027365 DOI: 10.1101/2025.04.11.648340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The mechanical properties of the extracellular matrix (ECM), particularly stiffness, regulate endothelial progenitor responses during vascular development, yet their behavior in physiologically compliant matrices (<1 kPa) remains underexplored. Using norbornene-modified hyaluronic acid (NorHA) hydrogels with tunable stiffness (190-884 Pa), we investigated how hydrogel stiffness influences cell morphology, endothelial maturation, mechanotransduction, and microvascular network formation in human induced pluripotent stem cell-derived endothelial progenitors (hiPSC-EPs). Our findings reveal a stiffness-dependent tradeoff between mechanotransduction and vascular network formation. At intermediate stiffness (551 Pa), cells exhibited the greatest increase in endothelial marker CD31 expression and Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) nuclear translocation, indicating enhanced mechanotransduction and endothelial maturation. However, this did not translate to superior plexus formation. Instead, the most compliant matrix (190 Pa) supported greater vascular connectivity, characterized by longer branches (∼0.03/volume vs. 0.015 at 551 Pa) and enhanced actin remodeling. 3D cell contraction measurements revealed a 15.6-fold higher basal displacement in compliant hydrogels, suggesting that cell-generated forces and matrix deformability collectively drive vascular morphogenesis. Unlike prior studies focusing on pathological stiffness ranges (>10 kPa), our results emphasize that vascularization is not solely driven by the most mechanotransductive environment but rather by a balance of compliance, contractility, and cell-induced remodeling. These findings underscore the need to design hydrogels that provide sufficient mechanotransduction for endothelial maturation while maintaining compliance to support dynamic vascular morphogenesis. This work provides a mechanically tuned framework for optimizing microenvironments to balance endothelial differentiation and vascular network formation in tissue engineering and regenerative medicine.
Collapse
|
5
|
Drozd M, Bruns AF, Yuldasheva NY, Maqbool A, Viswambharan H, Skromna A, Makava N, Cheng CW, Sukumar P, Eades L, Walker AMN, Griffin KJ, Galloway S, Watt NT, Haywood N, Palin V, Warmke N, Imrie H, Bridge K, Beech DJ, Wheatcroft SB, Kearney MT, Cubbon RM. Endothelial insulin-like growth factor-1 signaling regulates vascular barrier function and atherogenesis. Cardiovasc Res 2025:cvaf055. [PMID: 40171617 DOI: 10.1093/cvr/cvaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/19/2024] [Accepted: 02/15/2025] [Indexed: 04/04/2025] Open
Abstract
AIMS Progressive deposition of cholesterol in the arterial wall characterizes atherosclerosis, which underpins most cases of myocardial infarction and stroke. Insulin-like growth factor-1 (IGF-1) is a hormone that regulates systemic growth and metabolism and possesses anti-atherosclerotic properties. We asked whether endothelial-restricted augmentation of IGF-1 signaling is sufficient to suppress atherogenesis. METHODS AND RESULTS We generated mice with endothelial-restricted over-expression of human wildtype IGF-1R (hIGFREO/ApoE-/-) or a signaling defective K1003R mutant human IGF-1R (mIGFREO/ApoE-/-) and compared them to their respective ApoE-/- littermates. hIGFREO/ApoE-/- had less atherosclerosis, circulating leukocytes, arterial cholesterol uptake, and vascular leakage in multiple organs, whereas mIGFREO/ApoE-/- did not exhibit these phenomena. Overexpressing wildtype IGF-1R in human umbilical vein endothelial cells (HUVEC) altered the localization of tight junction proteins and reduced paracellular leakage across their monolayers, whilst overexpression of K1003R IGF-1R did not have these effects. Moreover, only overexpression of wildtype IGF-1R reduced HUVEC internalization of cholesterol-rich low density lipoprotein particles and increased their association of these particles with clathrin, but not caveolin-1, implicating it in vesicular uptake of lipoproteins. Endothelial overexpression of wildtype versus K1003R IGF-1R also reduced expression of YAP/TAZ target genes and nuclear localization of TAZ, which may be relevant to its impact on vascular barrier and atherogenesis. CONCLUSIONS Endothelial IGF-1 signaling modulates both para- and trans-cellular vascular barrier function. Beyond reducing atherosclerosis, this could have relevance to many diseases associated with abnormal vascular permeability.
Collapse
Affiliation(s)
- Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Alexander-Francisco Bruns
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Azhar Maqbool
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Natallia Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Lauren Eades
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Andrew M N Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Stacey Galloway
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Natalie Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Victoria Palin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Helen Imrie
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Katherine Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
6
|
Zheng Y, Nützl M, Schackel T, Chen J, Weidner N, Müller R, Puttagunta R. Biomaterial scaffold stiffness influences the foreign body reaction, tissue stiffness, angiogenesis and neuroregeneration in spinal cord injury. Bioact Mater 2025; 46:134-149. [PMID: 39760066 PMCID: PMC11700269 DOI: 10.1016/j.bioactmat.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Biomaterial scaffold engineering presents great potential in promoting axonal regrowth after spinal cord injury (SCI), yet persistent challenges remain, including the surrounding host foreign body reaction and improper host-implant integration. Recent advances in mechanobiology spark interest in optimizing the mechanical properties of biomaterial scaffolds to alleviate the foreign body reaction and facilitate seamless integration. The impact of scaffold stiffness on injured spinal cords has not been thoroughly investigated. Herein, we introduce stiffness-varied alginate anisotropic capillary hydrogel scaffolds implanted into adult rat C5 spinal cords post-lateral hemisection. Four weeks post-implantation, scaffolds with a stiffness approaching that of the spinal cord effectively minimize the host foreign body reaction via yes-associated protein (YAP) nuclear translocation. Concurrently, the softest scaffolds maximize cell infiltration and angiogenesis, fostering significant axonal regrowth but limiting the rostral-caudal linear growth. Furthermore, as measured by atomic force microscopy (AFM), the surrounding spinal cord softens when in contact with the stiffest scaffold while maintaining a physiological level in contact with the softest one. In conclusion, our findings underscore the pivotal role of stiffness in scaffold engineering for SCI in vivo, paving the way for the optimal development of efficacious biomaterial scaffolds for tissue engineering in the central nervous system.
Collapse
Affiliation(s)
- Yifeng Zheng
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, 350005, Fuzhou, China
| | - Maximilian Nützl
- Department of Physical and Theoretical Chemistry, University of Regensburg, 93053, Regensburg, Germany
| | - Thomas Schackel
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Jing Chen
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Rainer Müller
- Department of Physical and Theoretical Chemistry, University of Regensburg, 93053, Regensburg, Germany
| | - Radhika Puttagunta
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| |
Collapse
|
7
|
Jaworski D, Hundsdorfer L, Bastounis E, Constantinou I. StretchView - A Multi-Axial Cell-Stretching Device for Long-Term Automated Videomicroscopy of Living Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408853. [PMID: 39792792 PMCID: PMC11884571 DOI: 10.1002/advs.202408853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/28/2024] [Indexed: 01/12/2025]
Abstract
Incorporating mechanical stretching of cells in tissue culture is crucial for mimicking (patho)-physiological conditions and understanding the mechanobiological responses of cells, which can have significant implications in areas like tissue engineering and regenerative medicine. Despite the growing interest, most available cell-stretching devices are not compatible with automated live-cell imaging, indispensable for characterizing alterations in the dynamics of various important cellular processes. In this work, StretchView is presented, a multi-axial cell-stretching platform compatible with automated, time-resolved live-cell imaging. Using StretchView, long-term image acquisition of cells in the relaxed and stretched states is shown for the first time (experimental time of 12 h) without the need for human intervention. Homogeneous and stable strain fields are demonstrated for 18 h of cyclic stretching, highlighting the platform's versatility and robustness. As proof-of-principle, the effect of stretching on cell kinematics and spatiotemporal localization of the cell-cell adhesion protein E-cadherin is examined for MDCK cells in monolayer. First evidence of a monotonic increase in junctional E-cadherin localization upon exposure to stretch is presented using live-cell imaging data acquired during cyclic stretching, suggestive of an increase in barrier integrity of the monolayer. These findings highlight the potential of StretchView in providing insights into cell mechanobiology and beyond.
Collapse
Affiliation(s)
- David Jaworski
- Institute of Microtechnology (IMT)Technische Universität BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technische Universität BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Lara Hundsdorfer
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT)University of TübingenAuf der Morgenstelle 2872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), EXC 2124University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Effie Bastounis
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT)University of TübingenAuf der Morgenstelle 2872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), EXC 2124University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technische Universität BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technische Universität BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
8
|
Chen H, Peng C, Fang F, Li Y, Liu X, Hu Y, Wang G, Liu X, Shen Y. Angiogenesis within atherosclerotic plaques: Mechanical regulation, molecular mechanism and clinical diagnosis. MECHANOBIOLOGY IN MEDICINE 2025; 3:100114. [PMID: 40396135 PMCID: PMC12082165 DOI: 10.1016/j.mbm.2025.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/12/2024] [Accepted: 01/05/2025] [Indexed: 05/22/2025]
Abstract
Atherosclerosis (AS) is a disease characterized by focal cholesterol accumulation and insoluble inflammation in arterial intima, leading to the formation of an atherosclerotic plaque consisting of lipids, cells, and fibrous matrix. The presence of plaque can restrict or obstruct blood flow, resulting in arterial stenosis and local mechanical microenvironment changes including flow shear stress, vascular matrix stiffness, and plaque structural stress. Neovascularization within the atherosclerotic plaque plays a crucial role in both plaque growth and destabilization, potentially leading to plaque rupture and fatal embolism. However, the exact interactions between neovessels and plaque remain unclear. In this review, we provide a comprehensive analysis of the origin of intraplaque neovessels, the contributing factors, underlying molecular mechanisms, and associated signaling pathways. We specifically emphasize the role of mechanical factors contributing to angiogenesis in atherosclerotic plaques. Additionally, we summarize the imaging techniques and therapeutic strategies for intraplaque neovessels to enhance our understanding of this field.
Collapse
Affiliation(s)
- Hanxiao Chen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Chengxiu Peng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yuhao Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaran Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ying Hu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Guixue Wang
- Jinfeng Laboratory, Chongqing 401329, China
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
9
|
Giese W, Albrecht JP, Oppenheim O, Akmeriç EB, Kraxner J, Schmidt D, Harrington K, Gerhardt H. Polarity-JaM: an image analysis toolbox for cell polarity, junction and morphology quantification. Nat Commun 2025; 16:1474. [PMID: 39922822 PMCID: PMC11807127 DOI: 10.1038/s41467-025-56643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
Cell polarity involves the asymmetric distribution of cellular components such as signalling molecules and organelles within a cell, alterations in cell morphology and cell-cell contacts. Advances in fluorescence microscopy and deep learning algorithms open up a wealth of unprecedented opportunities to characterise various aspects of cell polarity, but also create new challenges for comprehensible and interpretable image data analysis workflows to fully exploit these new opportunities. Here we present Polarity-JaM, an open source package for reproducible exploratory image analysis that provides versatile methods for single cell segmentation, feature extraction and statistical analysis. We demonstrate our analysis using fluorescence image data of endothelial cells and their collective behaviour, which has been shown to be essential for vascular development and disease. The general architecture of the software allows its application to other cell types and imaging modalities, as well as seamless integration into common image analysis workflows, see https://polarityjam.readthedocs.io . We also provide a web application for circular statistics and data visualisation, available at www.polarityjam.com , and a Napari plug-in, each with a graphical user interface to facilitate exploratory analysis. We propose a holistic image analysis workflow that is accessible to the end user in bench science, enabling comprehensive analysis of image data.
Collapse
Affiliation(s)
- Wolfgang Giese
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Berlin, Germany.
| | - Jan Philipp Albrecht
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- HELMHOLTZ IMAGING, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Faculty of Mathematics and Natural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olya Oppenheim
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emir Bora Akmeriç
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Kraxner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Deborah Schmidt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- HELMHOLTZ IMAGING, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kyle Harrington
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
10
|
Guo D, Yao B, Shao W, Zuo J, Chang Z, Shi J, Hu N, Bao S, Chen M, Fan X, Li X. The Critical Role of YAP/BMP/ID1 Axis on Simulated Microgravity-Induced Neural Tube Defects in Human Brain Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410188. [PMID: 39656892 PMCID: PMC11792043 DOI: 10.1002/advs.202410188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/05/2024] [Indexed: 12/17/2024]
Abstract
Integrated biochemical and biophysical signals regulate embryonic development. Correct neural tube formation is critical for the development of central nervous system. However, the role of microgravity in neurodevelopment and its underlying molecular mechanisms remain unclear. In this study, the effects of stimulated microgravity (SMG) on the development of human brain organoids are investigated. SMG impairs N-cadherin-based adherens junction formation, leading to neural tube defects associated with dysregulated self-renewal capacity and neuroepithelial disorganization in human brain organoids. Bulk gene expression analyses reveal that SMG alters Hippo and BMP signaling in brain organoids. The neuropathological deficits in SMG-treated organoids can be rescued by regulating YAP/BMP/ID1 axis. Furthermore, sing-cell RNA sequencing data show that SMG results in perturbations in the number and function of neural stem and progenitor cell subpopulations. One of these subpopulations senses SMG cues and transmits BMP signals to the subpopulation responsible for tube morphogenesis, ultimately affecting the proliferating cell population. Finally, SMG intervention leads to persistent neurologic damage even after returning to normal gravity conditions. Collectively, this study reveals molecular and cellular abnormalities associated with SMG during human brain development, providing opportunities for countermeasures to maintain normal neurodevelopment in space.
Collapse
Affiliation(s)
- Di Guo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Wen‐Wei Shao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jia‐Chen Zuo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Zhe‐Han Chang
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jian‐Xin Shi
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Nan Hu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Shuang‐Qing Bao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Meng‐Meng Chen
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiu Fan
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiao‐Hong Li
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| |
Collapse
|
11
|
Lone M, Anwar T, Sinnett-Smith J, Jin YP, Reed EF, Rozengurt E. Antibody ligation of HLA class II induces YAP nuclear localization and formation of cytoplasmic YAP condensates in human endothelial cells. Immunohorizons 2025; 9:vlae008. [PMID: 39865973 PMCID: PMC11841970 DOI: 10.1093/immhor/vlae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/26/2024] [Indexed: 01/28/2025] Open
Abstract
Antibody (Ab) crosslinking of HLA class II (HLA II) molecules on the surface of endothelial cells (ECs) triggers proliferative and prosurvival intracellular signaling, which are implicated in promoting chronic Ab-mediated rejection (cAMR). Despite the importance of cAMR in transplant medicine, the mechanisms involved remain incompletely understood. Here, we examined the regulation of yes-associated protein (YAP) nuclear cytoplasmic localization and phosphorylation in human ECs challenged with Abs that bind HLA II, which are strongly associated with cAMR. To examine changes in YAP localization in response to Ab-mediated engagement of HLA II, we used an adenoviral vector to express the class II transactivator or treatment with interferon γ. In unstimulated ECs expressing HLA II, YAP localized mainly in the cytoplasm. Stimulation with HLA II Ab (0.1-1 µg/mL) induced marked translocation of YAP to the nucleus. HLA II signaling triggered by high concentrations of HLA II Ab (1 µg/mL) also induced prominent YAP localization in cytoplasmic punctate structures that were disassembled by exposure to 1,6-hexanediol, suggesting that these structures are biomolecular condensates. Using multiple treatments, including stimulation with serum, thrombin or HLA I Ab and conditions (eg ECs plated at different densities) indicate that formation of YAP cytoplasmic puncta can be dissociated from YAP nuclear localization and phosphorylation at Ser127, a site in YAP targeted by the Hippo kinases LATS1/2. The results revealed that HLA II signaling regulates YAP subcellular distributions in ECs and demonstrate, for the first time, that HLA II Ab selectively stimulates YAP concentration in punctate structures.
Collapse
Affiliation(s)
- Moien Lone
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Tarique Anwar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - James Sinnett-Smith
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Yi-Ping Jin
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Enrique Rozengurt
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
12
|
Huveneers S, Phng LK. Endothelial cell mechanics and dynamics in angiogenesis. Curr Opin Cell Biol 2024; 91:102441. [PMID: 39342870 DOI: 10.1016/j.ceb.2024.102441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
The efficient distribution of oxygen and metabolites is critical for embryonic development and growth as well as tissue homeostasis. This is achieved by endothelial cells forming and maintaining a closed, circulatory network of tubular blood vessels. Endothelial cells are highly plastic cells with the capability to generate diverse dynamic responses at different stages of vessel development in order to build vessel networks of tissue-specific patterns and morphologies. In this review, we discuss new conceptual advances gained from in vitro and in vivo models of angiogenesis on the control of endothelial cell dynamics. We highlight the complex interplay between mechanical cues, actin cytoskeleton and endothelial behaviors, and the emerging importance of hydrostatic pressure in complementing actin-dependent mechanisms to regulate endothelial cell mechanics and angiogenesis. Understanding these processes provides insights into vascular repair and regeneration mechanisms.
Collapse
Affiliation(s)
- Stephan Huveneers
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
| |
Collapse
|
13
|
Xia T, Pan Z, Wan H, Li Y, Mao G, Zhao J, Zhang F, Pan S. Mechanisms of mechanical stimulation in the development of respiratory system diseases. Am J Physiol Lung Cell Mol Physiol 2024; 327:L724-L739. [PMID: 39316681 DOI: 10.1152/ajplung.00122.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
During respiration, mechanical stress can initiate biological responses that impact the respiratory system. Mechanical stress plays a crucial role in the development of the respiratory system. However, pathological mechanical stress can impact the onset and progression of respiratory diseases by influencing the extracellular matrix and cell transduction processes. In this article, we explore the mechanisms by which mechanical forces communicate with and influence cells. We outline the basic knowledge of respiratory mechanics, elucidating the important role of mechanical stimulation in influencing respiratory system development and differentiation from a microscopic perspective. We also explore the potential mechanisms of mechanical transduction in the pathogenesis and development of respiratory diseases such as asthma, lung injury, pulmonary fibrosis, and lung cancer. Finally, we look forward to new research directions in cellular mechanotransduction, aiming to provide fresh insights for future therapeutic research on respiratory diseases.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoxin Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongsen Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guocai Mao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangbiao Zhang
- Department of Cardiothoracic Surgery, Lishui Municipal Central Hospital, Lishui, People's Republic of China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
14
|
Gráczer É, Pászty K, Harsányi L, Lehoczky C, Fülöp A, Varga A. BRAF Modulates the Interplay Between Cell-Cell and Cell-Extracellular Matrix Adhesions in PECAM-1-Mediated Mechanotransduction. Int J Mol Sci 2024; 25:11234. [PMID: 39457016 PMCID: PMC11508702 DOI: 10.3390/ijms252011234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Mechanotransduction, the process of how cells sense and convert mechanical stimuli into biochemical response, is crucial in the migration of leukocytes or cancer cells through the endothelium during inflammation or metastasis. Migrating cells exert forces on the endothelium through cell surface adhesion molecules, such as platelet endothelial adhesion molecule PECAM-1, and this is essential for a successful transmigration. To study PECAM-1-mediated mechanotransduction, we applied PECAM-1-antibody-coated magnetic beads and exerted about 40 pN force on the endothelial monolayer. We show that force increases cell-ECM adhesion in the cell center and is accompanied by the opening of cell-cell junctions. Upon depletion of the MEK/ERK kinase, BRAF force increases cell-ECM adhesion both at the cell periphery and in the cell center, but this does not result in the opening of cell-cell junctions. Decreasing cell-ECM adhesion in BRAF-depleted cells through FAK inhibition results in the remodeling of cell-cell junctions. Force-induced increase in cell-ECM adhesion in the cell center correlates with the activation of the transcriptional cofactor Yes-associated protein (YAP). Furthermore, the induced activation of YAP through LATS inhibition prevents junctional remodeling in control cells. Thus, the activation of YAP might determine the strength of cell-cell junctions during PECAM-1-mediated mechanotransduction.
Collapse
Affiliation(s)
- Éva Gráczer
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| | - Katalin Pászty
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| | - Laura Harsányi
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| | - Csilla Lehoczky
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | - Antónia Fülöp
- Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Andrea Varga
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| |
Collapse
|
15
|
Passier M, Bentley K, Loerakker S, Ristori T. YAP/TAZ drives Notch and angiogenesis mechanoregulation in silico. NPJ Syst Biol Appl 2024; 10:116. [PMID: 39368976 PMCID: PMC11455968 DOI: 10.1038/s41540-024-00444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/22/2024] [Indexed: 10/07/2024] Open
Abstract
Endothelial cells are key players in the cardiovascular system. Among other things, they are responsible for sprouting angiogenesis, the process of new blood vessel formation essential for both health and disease. Endothelial cells are strongly regulated by the juxtacrine signaling pathway Notch. Recent studies have shown that both Notch and angiogenesis are influenced by extracellular matrix stiffness; however, the underlying mechanisms are poorly understood. Here, we addressed this challenge by combining computational models of Notch signaling and YAP/TAZ, stiffness- and cytoskeleton-regulated mechanotransducers whose activity inhibits both Dll4 (Notch ligand) and LFng (Notch-Dll4 binding modulator). Our simulations successfully mimicked previous experiments, indicating that this YAP/TAZ-Notch crosstalk elucidates the Notch and angiogenesis mechanoresponse to stiffness. Additional simulations also identified possible strategies to control Notch activity and sprouting angiogenesis via cytoskeletal manipulations or spatial patterns of alternating stiffnesses. Our study thus inspires new experimental avenues and provides a promising modeling framework for further investigations into the role of Notch, YAP/TAZ, and mechanics in determining endothelial cell behavior during angiogenesis and similar processes.
Collapse
Affiliation(s)
- Margot Passier
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Katie Bentley
- The Francis Crick Institute, London, UK
- Department of Informatics, King's College London, London, UK
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
16
|
Brandon KD, Frank WE, Stroka KM. Junctions at the crossroads: the impact of mechanical cues on endothelial cell-cell junction conformations and vascular permeability. Am J Physiol Cell Physiol 2024; 327:C1073-C1086. [PMID: 39129490 PMCID: PMC11481987 DOI: 10.1152/ajpcell.00605.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Cells depend on precisely regulating barrier function within the vasculature to maintain physiological stability and facilitate essential substance transport. Endothelial cells achieve this through specialized adherens and tight junction protein complexes, which govern paracellular permeability across vascular beds. Adherens junctions, anchored by vascular endothelial (VE)-cadherin and associated catenins to the actin cytoskeleton, mediate homophilic adhesion crucial for barrier integrity. In contrast, tight junctions composed of occludin, claudin, and junctional adhesion molecule A interact with Zonula Occludens proteins, reinforcing intercellular connections essential for barrier selectivity. Endothelial cell-cell junctions exhibit dynamic conformations during development, maturation, and remodeling, regulated by local biochemical and mechanical cues. These structural adaptations play pivotal roles in disease contexts such as chronic inflammation, where junctional remodeling contributes to increased vascular permeability observed in conditions from cancer to cardiovascular diseases. Conversely, the brain microvasculature's specialized junctional arrangements pose challenges for therapeutic drug delivery due to their unique molecular compositions and tight organization. This commentary explores the molecular mechanisms underlying endothelial cell-cell junction conformations and their implications for vascular permeability. By highlighting recent advances in quantifying junctional changes and understanding mechanotransduction pathways, we elucidate how physical forces from cellular contacts and hemodynamic flow influence junctional dynamics.
Collapse
Affiliation(s)
- Ken D Brandon
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
| | - William E Frank
- Department of Biology, University of Puerto Rico in Ponce, Ponce, Puerto Rico
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, United States
- Biophysics Program, University of Maryland, College Park, Maryland, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, Maryland, United States
| |
Collapse
|
17
|
Zhao X, Cai Z, Luo Y, Lin Z, Wang J. Overexpression of CGRP receptor attenuates tendon graft degeneration in anterior cruciate ligament reconstruction. J Orthop Res 2024. [PMID: 39318262 DOI: 10.1002/jor.25978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Cell apoptosis or necrosis, extracellular matrix loss, and excessive inflammation may induce tendon graft degeneration. The impairment in the regeneration capability of nerve fibers and blood vessels may be the critical cause. Calcitonin gene-related peptide (CGRP), exhibiting a short half-life, favors cell proliferation, nerve fiber regeneration and angiogenesis. We aimed to investigate the effects of CGRP receptor-mediated signaling on tendon graft integrity and study if the modulation pathways are ascribed to cell proliferation, nerve fiber and blood vessel regeneration. A total of three groups in mice with ACL reconstruction were established: the control group (PBS treatment), the adenovirus vectors expressing CGRP receptor (CALCRL) treated group (Adv-Calcrl treatment), and the adenovirus vectors carrying shRNA targeting Calcrl treated group (Adv-shCalcrl treatment). The histological assessment indicated the Adv-Calcrl treatment was favored while the Adv-shCalcrl significantly impaired tendon graft integrity. TUNEL staining revealed a significant decreased number of apoptotic cells in the Adv-Calcrl group relative to the control group and the adv-shCalcrl group. Compared to the control group and the Adv-shCalcrl group, the Adv-Calcrl group showed significantly enhanced proliferation of nestin positive cells. Of note, the Adv-Calcrl treatment significantly increased EMCN expression at the tendon graft relative to the control and the Adv-shCalcrl groups, which may be ascribed to attenuation of the Hippo signaling pathway. Importantly, the Adv-Calcrl treatment significantly increased sensory nerve fibers and also PIEZO2 levels. Our results demonstrate the activation of CGRP receptor-mediated signaling attenuated tendon graft degeneration, which was ascribed to enhanced proliferation of Nestin positive cells, angiogenesis, and nerve fiber outgrowth.
Collapse
Affiliation(s)
- Xibang Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhaoji Cai
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ying Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhousheng Lin
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
18
|
Gao S, Thillaikumaran T, Dominguez MH, Giang W, Hayes K, Chen X, Pace J, Bockman J, Jathan D, Sung D, Narayan S, Frankfurter M, Mericko-Ishizuka P, Yang J, Castro M, Potente M, Kahn ML. YAP/TAZ signaling in allantois-derived cells is required for placental vascularization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613151. [PMID: 39345443 PMCID: PMC11429833 DOI: 10.1101/2024.09.15.613151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Normal placental development and angiogenesis are crucial for fetal growth and maternal health during pregnancy. However, molecular regulation of placental angiogenesis has been difficult to study due to a lack of specific genetic tools that isolate the placenta from the embryo and yolk sac. To address this gap in knowledge we recently developed Hoxa13 Cre mice in which Cre is expressed in allantois-derived cells, including placental endothelial and stromal cells. Mice lacking the transcriptional regulators Yes-associated protein (YAP) and PDZ-binding motif (TAZ) in allantois-derived cells exhibit embryonic lethality at midgestation with compromised placental vasculature. snRNA-seq analysis revealed transcriptional changes in placental stromal cells and endothelial cells. YAP/TAZ mutants exhibited significantly reduced placental stromal cells prior to the endothelial architectural change, highlighting the role of these cells in placental vascular growth. These results reveal a central role for YAP/TAZ signaling during placental vascular growth and implicate Hoxa13 -derived placental stromal cells as a critical component of placental vascularization.
Collapse
|
19
|
Tan YH, Habing KM, Riesterer JL, Stempinski ES, Lewis SH, Pfeifer CS, Malhotra SV, Nakayama KH. Engineered nanofibrillar collagen with tunable biophysical properties for myogenic, endothelial, and osteogenic cell guidance. Acta Biomater 2024; 186:95-107. [PMID: 39117115 PMCID: PMC11407781 DOI: 10.1016/j.actbio.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
A goal of regenerative engineering is the rational design of materials to restore the structure-function relationships that drive reparative programs in damaged tissues. Despite the widespread use of extracellular matrices for engineering tissues, their application has been limited by a narrow range of tunable features. The primary objective of this study is to develop a versatile platform for evaluating tissue-specific cellular interactions using Type I collagen scaffolds with highly tunable biophysical properties. The kinetics of collagen fibrillogenesis were modulated through a combination of varied shear rate and pH during neutralization, to achieve a broad range of fibril anisotropy, porosity, diameter, and storage modulus. The role that each of these properties play in guiding muscle, bone, and vascular cell types was comprehensively identified, and informed the in vitro generation of three distinct musculoskeletal engineered constructs. Myogenesis was highly regulated by smaller fibrils and larger storage moduli, endothelial inflammatory phenotype was predominantly guided by fibril anisotropy, and osteogenesis was enhanced by highly porous collagen with larger fibrils. This study introduces a novel approach for dynamically modulating Type I collagen materials and provides a robust platform for investigating cell-material interactions, offering insights for the future rational design of tissue-specific regenerative biomaterials. STATEMENT OF SIGNIFICANCE: The biophysical properties of regenerative materials facilitate key cell-substrate interactions that can guide the morphology, phenotype, and biological response of cells. In this study, we describe the fabrication of an engineered collagen hydrogel that can be modified to exhibit control over a wide range of biophysical features, including fibril organization and size, nanoscale porosity, and mechanics. We identified the unique combination of collagen features that optimally promote regenerative muscle, bone, and vascular cell types while also delineating the properties that hinder these same cellular responses. This study presents a highly accessible method to control the biophysical properties of collagen hydrogels that can be adapted for a broad range of tissue engineering and regenerative applications.
Collapse
Affiliation(s)
- Yong How Tan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Krista M Habing
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Jessica L Riesterer
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Erin S Stempinski
- Multiscale Microscopy Core, Oregon Health & Science University, Portland, OR, USA
| | - Steven H Lewis
- Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Carmem S Pfeifer
- Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Sanjay V Malhotra
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Karina H Nakayama
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA; Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
20
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza induces lung lymphangiogenesis independent of YAP/TAZ activity in lymphatic endothelial cells. Sci Rep 2024; 14:21324. [PMID: 39266641 PMCID: PMC11393066 DOI: 10.1038/s41598-024-72115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases twofold at 7 days post-influenza infection (dpi) and threefold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA.
| | - Senegal Carty
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Fengzhi Shao
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Jhonatan Henao-Vasquez
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alexandra B Ysasi
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Michelle Zeng
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Anne Hinds
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Ming Lo
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Comparative Pathology Laboratory, Boston University National Emerging and Infectious Disease Laboratories, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Matthew R Jones
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alan Fine
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| |
Collapse
|
21
|
Keshavarz M, Smith Q. Gelatin-Mediated Vascular Self-Assembly via a YAP-MMP Signaling Axis. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2402360. [PMID: 39583865 PMCID: PMC11583535 DOI: 10.1002/adfm.202402360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Indexed: 11/26/2024]
Abstract
Tissue self-assembly relies on the interplay between structural cues imparted by the extracellular matrix and instructive chemical factors that guide cellular signaling pathways. Here, we report that endothelial cell-laden gelatin-based hydrogels with optimized mechanical and chemical properties facilitate de novo vasculogenesis and recruitment of endogenous blood vessels in vivo. We demonstrate that these engineered matrices, with tailored viscoelastic features and stiffness, drive vascular self-assembly in a yes-associated protein mechanosensing-dependent manner through αvβ3 integrin and matrix metalloproteinase 2 activity. Our research highlights how the extracellular matrix, in the form of gelatin-based hydrogels with adjustable stress relaxation rates, drive vascular morphogenesis in the absence of growth factor supplementation, lending to a minimalistic platform for discretizing features of the microenvironment niche. Collectively, these results demonstrate a testbed that enables mechanistic evaluation of morphogenetic processes. Specifically, our results show how mechanical cues impact signaling pathways that modulate vascular remodeling, a critical tissue engineering paradigm needed for the translational application of vascularized grafts for regenerative medicine applications.
Collapse
Affiliation(s)
- Mozhgan Keshavarz
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, United States
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, United States
| |
Collapse
|
22
|
Surendran V, Safarulla S, Griffith C, Ali R, Madan A, Polacheck W, Chandrasekaran A. Magnetically Integrated Tumor-Vascular Interface System to Mimic Pro-angiogenic Endothelial Dysregulations for On-Chip Drug Testing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47075-47088. [PMID: 39196896 PMCID: PMC11403600 DOI: 10.1021/acsami.4c01766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
The tumor-vascular interface is a critical component of the tumor microenvironment that regulates all of the dynamic interactions between a growing tumor and the endothelial lining of the surrounding vasculature. In this paper, we report the design and development of a custom-engineered tumor-vascular interface system for investigating the early stage tumor-mediated pro-angiogenic dysfunctional behavior of the endothelium. Using representative endothelial cells and triple negative breast cancer cell lines, we established a biomimetic interface between a three-dimensional tumor tissue across a mature, functional endothelial barrier using a magnetically hybrid-integrated tumor-vascular interface system, wherein vasculature-like features containing a monolayer of endothelial cell culture on porous microfluidic channel surfaces were magnetically attached to tumor spheroids generated on a composite polymer-hydrogel microwell plate and embedded in a collagen matrix. Tumor-mediated endothelial microdynamics were characterized by their hallmark behavior such as loss of endothelial adherens junctions, increased cell density, proliferation, and changes in cell spreading and corroborated with endothelial YAP/TAZ nuclear translocation. We further confirm the feasibility of drug-mediated reversal of this pro-angiogenic endothelial organization through two different signaling mechanisms, namely, inhibition of the vascular endothelial growth factor pathway and the Notch signaling pathway, thereby demonstrating the utility of the tumor-vascular interface platform for rapid, early stage prediction of antiangiogenic drug efficacy. Overall, our work emphasizes the importance of our strategic engineering approach for identifying some unique, physiologically relevant aspects of the tumor-vascular interface, which are otherwise difficult to implement using standard in vitro approaches.
Collapse
Affiliation(s)
- Vikram Surendran
- Bioinspired Microengineering (BIOME) Laboratory, Department of Chemical, Biological and Bio Engineering, North Carolina A&T State University, Greensboro, North Carolina 27265, United States
| | - Simrit Safarulla
- Bioinspired Microengineering (BIOME) Laboratory, Department of Chemical, Biological and Bio Engineering, North Carolina A&T State University, Greensboro, North Carolina 27265, United States
| | - Christian Griffith
- Joint Department of Biomedical Engineering, UNC Chapel Hill─NC State University, Chapel Hill, North Carolina 27599, United States
| | - Reem Ali
- Bioinspired Microengineering (BIOME) Laboratory, Department of Chemical, Biological and Bio Engineering, North Carolina A&T State University, Greensboro, North Carolina 27265, United States
| | - Ankit Madan
- MedStar Southern Maryland Hospital Center, MedStar Georgetown Cancer Institute, Clinton, Maryland 20735, United States
| | - William Polacheck
- Joint Department of Biomedical Engineering, UNC Chapel Hill─NC State University, Chapel Hill, North Carolina 27599, United States
| | - Arvind Chandrasekaran
- Bioinspired Microengineering (BIOME) Laboratory, Department of Chemical, Biological and Bio Engineering, North Carolina A&T State University, Greensboro, North Carolina 27265, United States
| |
Collapse
|
23
|
Xu J, Wang J, Zhang H, Chen Y, Zhang X, Zhang Y, Xie M, Xiao J, Qiu J, Wang G. Coupled single-cell and bulk RNA-seq analysis reveals the engulfment role of endothelial cells in atherosclerosis. Genes Dis 2024; 11:101250. [PMID: 39022128 PMCID: PMC11252887 DOI: 10.1016/j.gendis.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 10/28/2023] [Accepted: 12/05/2023] [Indexed: 07/20/2024] Open
Abstract
The clearance of apoptotic cell debris, containing professional phagocytosis and non-professional phagocytosis, is essential for maintaining the homeostasis of healthy tissues. Here, we discovered that endothelial cells could engulf apoptotic cell debris in atherosclerotic plaque. Single-cell RNA sequencing (RNA-seq) has revealed a unique endothelial cell subpopulation in atherosclerosis, which was strongly associated with vascular injury-related pathways. Moreover, integrated analysis of three vascular injury-related RNA-seq datasets showed that the expression of scavenger receptor class B type 1 (SR-B1) was up-regulated and specifically enriched in the phagocytosis pathway under vascular injury circumstances. Single-cell RNA-seq and bulk RNA-seq indicate that SR-B1 was highly expressed in a unique endothelial cell subpopulation of mouse aorta and strongly associated with the reorganization of cellular adherent junctions and cytoskeleton which were necessary for phagocytosis. Furthermore, SR-B1 was strongly required for endothelial cells to engulf apoptotic cell debris in atherosclerotic plaque of both mouse and human aorta. Overall, this study demonstrated that apoptotic cell debris could be engulfed by endothelial cells through SR-B1 and associated with the reorganization of cellular adherent junctions and cytoskeleton.
Collapse
Affiliation(s)
- Jianxiong Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Hongping Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Ying Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Ming Xie
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Jun Xiao
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
24
|
Tan C, Ge ZD, Kurup S, Dyakiv Y, Liu T, Muller WA, Kume T. FOXC1 and FOXC2 Ablation Causes Abnormal Valvular Endothelial Cell Junctions and Lymphatic Vessel Formation in Myxomatous Mitral Valve Degeneration. Arterioscler Thromb Vasc Biol 2024; 44:1944-1959. [PMID: 38989578 PMCID: PMC11335087 DOI: 10.1161/atvbaha.124.320316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Mitral valve (MV) disease including myxomatous degeneration is the most common form of valvular heart disease with an age-dependent frequency. Genetic evidence indicates that mutations of the human transcription factor FOXC1 are associated with MV defects, including MV regurgitation. In this study, we sought to determine whether murine Foxc1 and its closely related factor, Foxc2, are required in valvular endothelial cells (VECs) for the maintenance of MV leaflets, including VEC junctions and the stratified trilaminar ECM (extracellular matrix). METHODS Adult mice carrying tamoxifen-inducible, vascular endothelial cell (EC), and lymphatic EC-specific, compound Foxc1;Foxc2 mutations (ie, EC-Foxc-DKO and lymphatic EC-Foxc-DKO mice, respectively) were used to study the function of Foxc1 and Foxc2 in the maintenance of MVs. The EC and lymphatic EC mutations of Foxc1/c2 were induced at 7 to 8 weeks of age by tamoxifen treatment, and abnormalities in the MVs of these mutant mice were assessed via whole-mount immunostaining, immunohistochemistry/RNAscope, Movat pentachrome/Masson Trichrome staining, and Evans blue injection. RESULTS EC deletions of Foxc1 and Foxc2 in mice resulted in abnormally extended and thicker MVs by causing defects in the regulation of ECM organization with increased proteoglycan and decreased collagen. Notably, reticular adherens junctions were found in VECs of control MV leaflets, and these reticular structures were severely disrupted in EC-Foxc-DKO mice. PROX1 (prospero homeobox protein 1), a key regulator in a subset of VECs on the fibrosa side of MVs, was downregulated in EC-Foxc1/c2 mutant VECs. Furthermore, we determined the precise location of lymphatic vessels in murine MVs, and these lymphatic vessels were aberrantly expanded and dysfunctional in EC-Foxc1/c2 mutant MVs. Lymphatic EC deletion of Foxc1/c2 also resulted in similar structural/ECM abnormalities as seen in EC-Foxc1/c2 mutant MVs. CONCLUSIONS Our results indicate that Foxc1 and Foxc2 are required for maintaining the integrity of the MV, including VEC junctions, ECM organization, and lymphatic vessel formation/function to prevent myxomatous MV degeneration.
Collapse
Affiliation(s)
- Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Zhi-Dong Ge
- Departments of Pediatrics, Surgery, and Pathology, Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago (Z.-D.G.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Shreya Kurup
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
- Honors College, University of Illinois at Chicago (S.K.)
| | - Yaryna Dyakiv
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ting Liu
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - William A. Muller
- Department of Pathology (W.A.M.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute (C.T., S.K., Y.D., T.L., T.K.), Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
25
|
Mai Y, Kobayashi Y, Kitahata H, Seo T, Nohara T, Itamoto S, Mai S, Kumamoto J, Nagayama M, Nishie W, Ujiie H, Natsuga K. Patterning in stratified epithelia depends on cell-cell adhesion. Life Sci Alliance 2024; 7:e202402893. [PMID: 39025524 PMCID: PMC11258421 DOI: 10.26508/lsa.202402893] [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: 06/19/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Epithelia consist of proliferating and differentiating cells that often display patterned arrangements. However, the mechanism regulating these spatial arrangements remains unclear. Here, we show that cell-cell adhesion dictates multicellular patterning in stratified epithelia. When cultured keratinocytes, a type of epithelial cell in the skin, are subjected to starvation, they spontaneously develop a pattern characterized by areas of high and low cell density. Pharmacological and knockout experiments show that adherens junctions are essential for patterning, whereas the mathematical model that only considers local cell-cell adhesion as a source of attractive interactions can form regions with high/low cell density. This phenomenon, called cell-cell adhesion-induced patterning (CAIP), influences cell differentiation and proliferation through Yes-associated protein modulation. Starvation, which induces CAIP, enhances the stratification of the epithelia. These findings highlight the intrinsic self-organizing property of epithelial cells.
Collapse
Affiliation(s)
- Yosuke Mai
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuaki Kobayashi
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
- Department of Mathematics, Faculty of Science, Josai University, Sakado, Japan
| | - Hiroyuki Kitahata
- Department of Physics, Graduate School of Science, Chiba University, Chiba, Japan
| | - Takashi Seo
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takuma Nohara
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sota Itamoto
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shoko Mai
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Junichi Kumamoto
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Masaharu Nagayama
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Wataru Nishie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ken Natsuga
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
26
|
Al-Nuaimi DA, Rütsche D, Abukar A, Hiebert P, Zanetti D, Cesarovic N, Falk V, Werner S, Mazza E, Giampietro C. Hydrostatic pressure drives sprouting angiogenesis via adherens junction remodelling and YAP signalling. Commun Biol 2024; 7:940. [PMID: 39097636 PMCID: PMC11297954 DOI: 10.1038/s42003-024-06604-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
Endothelial cell physiology is governed by its unique microenvironment at the interface between blood and tissue. A major contributor to the endothelial biophysical environment is blood hydrostatic pressure, which in mechanical terms applies isotropic compressive stress on the cells. While other mechanical factors, such as shear stress and circumferential stretch, have been extensively studied, little is known about the role of hydrostatic pressure in the regulation of endothelial cell behavior. Here we show that hydrostatic pressure triggers partial and transient endothelial-to-mesenchymal transition in endothelial monolayers of different vascular beds. Values mimicking microvascular pressure environments promote proliferative and migratory behavior and impair barrier properties that are characteristic of a mesenchymal transition, resulting in increased sprouting angiogenesis in 3D organotypic model systems ex vivo and in vitro. Mechanistically, this response is linked to differential cadherin expression at the adherens junctions, and to an increased YAP expression, nuclear localization, and transcriptional activity. Inhibition of YAP transcriptional activity prevents pressure-induced sprouting angiogenesis. Together, this work establishes hydrostatic pressure as a key modulator of endothelial homeostasis and as a crucial component of the endothelial mechanical niche.
Collapse
Affiliation(s)
| | - Dominic Rütsche
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland
| | - Asra Abukar
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland
| | - Paul Hiebert
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
- Centre for Biomedicine, Hull York Medical School, The University of Hull, Hull, HU6 7RX, UK
| | - Dominik Zanetti
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Nikola Cesarovic
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zürich, Switzerland
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zürich, Switzerland
| | - Sabine Werner
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Edoardo Mazza
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland.
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland.
| | - Costanza Giampietro
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland.
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland.
| |
Collapse
|
27
|
Kim D, Olson JM, Cooper JA. N-cadherin dynamically regulates pediatric glioma cell migration in complex environments. J Cell Biol 2024; 223:e202401057. [PMID: 38477830 PMCID: PMC10937189 DOI: 10.1083/jcb.202401057] [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: 01/11/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that intercellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.
Collapse
Affiliation(s)
- Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James M. Olson
- Clinical Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jonathan A. Cooper
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
28
|
Zhang Y, Ren Y, Li X, Li M, Fu M, Zhou W, Yu Y, Xiong Y. A review on decoding the roles of YAP/TAZ signaling pathway in cardiovascular diseases: Bridging molecular mechanisms to therapeutic insights. Int J Biol Macromol 2024; 271:132473. [PMID: 38795886 DOI: 10.1016/j.ijbiomac.2024.132473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) serve as transcriptional co-activators that dynamically shuttle between the cytoplasm and nucleus, resulting in either the suppression or enhancement of their downstream gene expression. Recent emerging evidence demonstrates that YAP/TAZ is strongly implicated in the pathophysiological processes that contribute to cardiovascular diseases (CVDs). In the cardiovascular system, YAP/TAZ is involved in the orchestration of a range of biological processes such as oxidative stress, inflammation, proliferation, and autophagy. Furthermore, YAP/TAZ has been revealed to be closely associated with the initiation and development of various cardiovascular diseases, including atherosclerosis, pulmonary hypertension, myocardial fibrosis, cardiac hypertrophy, and cardiomyopathy. In this review, we delve into recent studies surrounding YAP and TAZ, along with delineating their roles in contributing to the pathogenesis of CVDs with a link to various physiological processes in the cardiovascular system. Additionally, we highlight the current potential drugs targeting YAP/TAZ for CVDs therapy and discuss their challenges for translational application. Overall, this review may offer novel insights for understanding and treating cardiovascular disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Xiaofang Li
- Department of Gastroenterology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Man Li
- Department of Endocrinology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Mingdi Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Wenjing Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
29
|
Pratt SJP, Plunkett CM, Kuzu G, Trinh T, Barbara J, Choconta P, Quackenbush D, Huynh T, Smith A, Barnes SW, New J, Pierce J, Walker JR, Mainquist J, King FJ, Elliott J, Hammack S, Decker RS. A high throughput cell stretch device for investigating mechanobiology in vitro. APL Bioeng 2024; 8:026129. [PMID: 38938688 PMCID: PMC11210978 DOI: 10.1063/5.0206852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Mechanobiology is a rapidly advancing field, with growing evidence that mechanical signaling plays key roles in health and disease. To accelerate mechanobiology-based drug discovery, novel in vitro systems are needed that enable mechanical perturbation of cells in a format amenable to high throughput screening. Here, both a mechanical stretch device and 192-well silicone flexible linear stretch plate were designed and fabricated to meet high throughput technology needs for cell stretch-based applications. To demonstrate the utility of the stretch plate in automation and screening, cell dispensing, liquid handling, high content imaging, and high throughput sequencing platforms were employed. Using this system, an assay was developed as a biological validation and proof-of-concept readout for screening. A mechano-transcriptional stretch response was characterized using focused gene expression profiling measured by RNA-mediated oligonucleotide Annealing, Selection, and Ligation with Next-Gen sequencing. Using articular chondrocytes, a gene expression signature containing stretch responsive genes relevant to cartilage homeostasis and disease was identified. The possibility for integration of other stretch sensitive cell types (e.g., cardiovascular, airway, bladder, gut, and musculoskeletal), in combination with alternative phenotypic readouts (e.g., protein expression, proliferation, or spatial alignment), broadens the scope of high throughput stretch and allows for wider adoption by the research community. This high throughput mechanical stress device fills an unmet need in phenotypic screening technology to support drug discovery in mechanobiology-based disease areas.
Collapse
Affiliation(s)
- Stephen J. P. Pratt
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | | | - Guray Kuzu
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Ton Trinh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joshua Barbara
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Paula Choconta
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Doug Quackenbush
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Truc Huynh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Anders Smith
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - S. Whitney Barnes
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joel New
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Pierce
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - John R. Walker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Mainquist
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Frederick J. King
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Jimmy Elliott
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Scott Hammack
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Rebekah S. Decker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| |
Collapse
|
30
|
Ritsvall O, Albinsson S. Emerging role of YAP/TAZ in vascular mechanotransduction and disease. Microcirculation 2024; 31:e12838. [PMID: 38011540 DOI: 10.1111/micc.12838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
Cells have an incredible ability to physically interact with neighboring cells and their environment. They can detect and respond to mechanical forces by converting mechanical stimuli into biochemical signals in a process known as mechanotransduction. This is a key process for the adaption of vascular smooth muscle and endothelial cells to altered flow and pressure conditions. Mechanical stimuli, referring to a physical force exerted on cells, are primarily sensed by transmembrane proteins and the actin cytoskeleton, which initiate a cascade of intracellular events, including the activation of signaling pathways, ion channels, and transcriptional regulators. Recent work has highlighted an important role of the transcriptional coactivators YAP/TAZ for mechanotransduction in vascular cells. Interestingly, the activity of YAP/TAZ decreases with age, providing a potential mechanism for the detrimental effects of aging in the vascular wall. In this review, we summarize the current knowledge on the functional role of YAP and TAZ in vascular endothelial and smooth muscle cells for mechanotransduction in homeostasis and disease. In particular, the review is focused on in vivo observations from conditional knockout (KO) models of YAP/TAZ and the potential implications these studies may have for our understanding of vascular disease development.
Collapse
Affiliation(s)
- Olivia Ritsvall
- Department of Experimental Medical Science, Molecular Vascular Physiology, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science, Molecular Vascular Physiology, Lund University, Lund, Sweden
| |
Collapse
|
31
|
Hooglugt A, van der Stoel MM, Shapeti A, Neep BF, de Haan A, van Oosterwyck H, Boon RA, Huveneers S. DLC1 promotes mechanotransductive feedback for YAP via RhoGAP-mediated focal adhesion turnover. J Cell Sci 2024; 137:jcs261687. [PMID: 38563084 PMCID: PMC11112125 DOI: 10.1242/jcs.261687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Angiogenesis is a tightly controlled dynamic process demanding a delicate equilibrium between pro-angiogenic signals and factors that promote vascular stability. The spatiotemporal activation of the transcriptional co-factors YAP (herein referring to YAP1) and TAZ (also known WWTR1), collectively denoted YAP/TAZ, is crucial to allow for efficient collective endothelial migration in angiogenesis. The focal adhesion protein deleted-in-liver-cancer-1 (DLC1) was recently described as a transcriptional downstream target of YAP/TAZ in endothelial cells. In this study, we uncover a negative feedback loop between DLC1 expression and YAP activity during collective migration and sprouting angiogenesis. In particular, our study demonstrates that signaling via the RhoGAP domain of DLC1 reduces nuclear localization of YAP and its transcriptional activity. Moreover, the RhoGAP activity of DLC1 is essential for YAP-mediated cellular processes, including the regulation of focal adhesion turnover, traction forces, and sprouting angiogenesis. We show that DLC1 restricts intracellular cytoskeletal tension by inhibiting Rho signaling at the basal adhesion plane, consequently reducing nuclear YAP localization. Collectively, these findings underscore the significance of DLC1 expression levels and its function in mitigating intracellular tension as a pivotal mechanotransductive feedback mechanism that finely tunes YAP activity throughout the process of sprouting angiogenesis.
Collapse
Affiliation(s)
- Aukie Hooglugt
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081HZ Amsterdam, the Netherlands
| | - Miesje M. van der Stoel
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
| | - Apeksha Shapeti
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, 3001 Leuven, Belgium
| | - Beau F. Neep
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, 1081HZ Amsterdam, the Netherlands
| | - Annett de Haan
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
| | - Hans van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, 3001 Leuven, Belgium
- KU Leuven, Prometheus, Division of Skeletal Tissue Engineering, 3000 Leuven, Belgium
| | - Reinier A. Boon
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081HZ Amsterdam, the Netherlands
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt am Main, Germany
- Goethe University, Institute of Cardiovascular Regeneration, 60590 Frankfurt am Main, Germany
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
| |
Collapse
|
32
|
Mehl J, Farahani SK, Brauer E, Klaus‐Bergmann A, Thiele T, Ellinghaus A, Bartels‐Klein E, Koch K, Schmidt‐Bleek K, Petersen A, Gerhardt H, Vogel V, Duda GN. External Mechanical Stability Regulates Hematoma Vascularization in Bone Healing Rather than Endothelial YAP/TAZ Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307050. [PMID: 38273642 PMCID: PMC10987120 DOI: 10.1002/advs.202307050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/21/2023] [Indexed: 01/27/2024]
Abstract
Bone fracture healing is regulated by mechanobiological cues. Both, extracellular matrix (ECM) deposition and microvascular assembly determine the dynamics of the regenerative processes. Mechanical instability as by inter-fragmentary shear or compression is known to influence early ECM formation and wound healing. However, it remains unclear how these external cues shape subsequent ECM and microvascular network assembly. As transcriptional coactivators, the mechanotransducers yes-associated protein 1 (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) translate physical cues into downstream signaling events, yet their role in sprouting angiogenesis into the hematoma after injury is unknown. Using bone healing as model system for scar-free regeneration, the role of endothelial YAP/TAZ in combination with tuning the extrinsic mechanical stability via fracture fixation is investigated. Extrinsically imposed shear across the gap delayed hematoma remodeling and shaped the morphology of early collagen fiber orientations and microvascular networks, suggesting that enhanced shear increased the nutrient exchange in the hematoma. In contrast, endothelial YAP/TAZ deletion has little impact on the overall vascularization of the fracture gap, yet slightly increases the collagen fiber deposition under semi-rigid fixation. Together, these data provide novel insights into the respective roles of endothelial YAP/TAZ and extrinsic mechanical cues in orchestrating the process of bone regeneration.
Collapse
Affiliation(s)
- Julia Mehl
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Laboratory of Applied MechanobiologyDepartment of Health Sciences and TechnologyETH ZurichZurich8092Switzerland
| | - Saeed Khomeijani Farahani
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Erik Brauer
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Alexandra Klaus‐Bergmann
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Tobias Thiele
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Agnes Ellinghaus
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Eireen Bartels‐Klein
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Katharina Koch
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Katharina Schmidt‐Bleek
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Ansgar Petersen
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Holger Gerhardt
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Viola Vogel
- Laboratory of Applied MechanobiologyDepartment of Health Sciences and TechnologyETH ZurichZurich8092Switzerland
| | - Georg N. Duda
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| |
Collapse
|
33
|
Ma N, Wibowo YC, Wirtz P, Baltus D, Wieland T, Jansen S. Tankyrase inhibition interferes with junction remodeling, induces leakiness, and disturbs YAP1/TAZ signaling in the endothelium. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1763-1789. [PMID: 37741944 PMCID: PMC10858845 DOI: 10.1007/s00210-023-02720-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023]
Abstract
Tankyrase inhibitors are increasingly considered for therapeutic use in malignancies that are characterized by high intrinsic β-catenin activity. However, how tankyrase inhibition affects the endothelium after systemic application remains poorly understood. In this study, we aimed to investigate how the tankyrase inhibitor XAV939 affects endothelial cell function and the underlying mechanism involved. Endothelial cell function was analyzed using sprouting angiogenesis, endothelial cell migration, junctional dynamics, and permeability using human umbilical vein endothelial cells (HUVEC) and explanted mouse retina. Underlying signaling was studied using western blot, immunofluorescence, and qPCR in HUVEC in addition to luciferase reporter gene assays in human embryonic kidney cells. XAV939 treatment leads to altered junctional dynamics and permeability as well as impaired endothelial migration. Mechanistically, XAV939 increased stability of the angiomotin-like proteins 1 and 2, which impedes the nuclear translocation of YAP1/TAZ and consequently suppresses TEAD-mediated transcription. Intriguingly, XAV939 disrupts adherens junctions by inducing RhoA-Rho dependent kinase (ROCK)-mediated F-actin bundling, whereas disruption of F-actin bundling through the ROCK inhibitor H1152 restores endothelial cell function. Unexpectedly, this was accompanied by an increase in nuclear TAZ and TEAD-mediated transcription, suggesting differential regulation of YAP1 and TAZ by the actin cytoskeleton in endothelial cells. In conclusion, our findings elucidate the complex relationship between the actin cytoskeleton, YAP1/TAZ signaling, and endothelial cell function and how tankyrase inhibition disturbs this well-balanced signaling.
Collapse
Affiliation(s)
- Nan Ma
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Yohanes Cakrapradipta Wibowo
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Phillip Wirtz
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Doris Baltus
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany.
- DZHK, German Center for Cardiovascular Research, partner site Heidelberg/Mannheim, Mannheim, Germany.
| | - Sepp Jansen
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| |
Collapse
|
34
|
Mannion AJ, Zhao H, Zhang Y, von Wright Y, Bergman O, Roy J, Saharinen P, Holmgren L. Regulation of YAP Promotor Accessibility in Endothelial Mechanotransduction. Arterioscler Thromb Vasc Biol 2024; 44:666-689. [PMID: 38299356 PMCID: PMC10880945 DOI: 10.1161/atvbaha.123.320300] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Endothelial cells are constantly exposed to mechanical forces in the form of fluid shear stress, extracellular stiffness, and cyclic strain. The mechanoresponsive activity of YAP (Yes-associated protein) and its role in vascular development are well described; however, whether changes to transcription or epigenetic regulation of YAP are involved in these processes remains unanswered. Furthermore, how mechanical forces are transduced to the nucleus to drive transcriptional reprogramming in endothelial cells is poorly understood. The YAP target gene, AmotL2 (angiomotin-like 2), is a junctional mechanotransducer that connects cell-cell junctions to the nuclear membrane via the actin cytoskeleton. METHODS We applied mechanical manipulations including shear flow, stretching, and substrate stiffness to endothelial cells to investigate the role of mechanical forces in modulating YAP transcription. Using in vitro and in vivo endothelial depletion of AmotL2, we assess nuclear morphology, chromatin organization (using transposase-accessible chromatin sequencing), and whole-mount immunofluorescent staining of the aorta to determine the regulation and functionality of YAP. Finally, we use genetic and chemical inhibition to uncouple the nuclear-cytoskeletal connection to investigate the role of this pathway on YAP transcription. RESULTS Our results reveal that mechanical forces sensed at cell-cell junctions by the YAP target gene AmotL2 are directly involved in changes in global chromatin accessibility and activity of the histone methyltransferase EZH2, leading to modulation of YAP promotor activity. Functionally, shear stress-induced proliferation of endothelial cells in vivo was reliant on AmotL2 and YAP/TAZ (transcriptional coactivator with PDZ-binding motif) expression. Mechanistically, uncoupling of the nuclear-cytoskeletal connection from junctions and focal adhesions led to altered nuclear morphology, chromatin accessibility, and YAP promotor activity. CONCLUSIONS Our findings reveal a role for AmotL2 and nuclear-cytoskeletal force transmission in modulating the epigenetic and transcriptional regulation of YAP to maintain a mechano-enforced positive feedback loop of vascular homeostasis. These findings may offer an explanation as to the proinflammatory phenotype that leads to aneurysm formation observed in AmotL2 endothelial deletion models.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
- Department of Cell and Tissue Dynamics, Max Planck Institute of Molecular Biomedicine, Münster, Germany (A.J.M.)
| | - Honglei Zhao
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| | - Yuanyuan Zhang
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| | - Ylva von Wright
- Wihuri Research Institute, Biomedicum Helsinki, Finland (Y.v.W., P.S.)
| | - Otto Bergman
- Medicine (O.B.), Karolinska Institute, Stockholm, Sweden
| | - Joy Roy
- Molecular Medicine and Surgery (J.R.), Karolinska Institute, Stockholm, Sweden
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden (J.R.)
| | - Pipsa Saharinen
- Wihuri Research Institute, Biomedicum Helsinki, Finland (Y.v.W., P.S.)
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Finland (P.S.)
| | - Lars Holmgren
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
35
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza Induces Lung Lymphangiogenesis Independent of YAP/TAZ Activity in Lymphatic Endothelial Cells. RESEARCH SQUARE 2024:rs.3.rs-3951689. [PMID: 38463972 PMCID: PMC10925403 DOI: 10.21203/rs.3.rs-3951689/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases 2-fold at 7 days post-influenza infection (dpi) and 3-fold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Boston University Chobanian and Avedisian School of Medicine
| | - Senegal Carty
- Boston University Chobanian and Avedisian School of Medicine
| | - Fengzhi Shao
- Boston University Chobanian and Avedisian School of Medicine
| | | | | | - Michelle Zeng
- Boston University Chobanian and Avedisian School of Medicine
| | - Anne Hinds
- Boston University Chobanian and Avedisian School of Medicine
| | - Ming Lo
- Boston University Chobanian and Avedisian School of Medicine
| | | | | | - Matthew R Jones
- Boston University Chobanian and Avedisian School of Medicine
| | - Alan Fine
- Boston University Chobanian and Avedisian School of Medicine
| |
Collapse
|
36
|
Yan Y, Qin X, Zheng Y, Jin T, Hu Y, An Q, Leng B. Decreased PDLIM1 expression in endothelial cells contributes to the development of intracranial aneurysm. Vasc Med 2024; 29:5-16. [PMID: 38334094 DOI: 10.1177/1358863x231218210] [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: 02/10/2024]
Abstract
INTRODUCTION Intracranial aneurysm (IA) is a common vascular enlargement that occurs in the wall of cerebral vessels and frequently leads to fatal subarachnoid hemorrhage. PDZ and LIM domain protein 1 (PDLIM1) is a cytoskeletal protein that functions as a platform for multiple protein complex formation. However, whether PDLIM is involved in the pathogenesis of IA remains poorly understood. METHODS Loss-of-function and gain-of-function strategies were employed to determine the in vitro roles of PDLIM1 in vascular endothelial cells (VECs). A rat model of IA was generated to study the role of PDLIM1 in vivo. Gene expression profiling, Western blotting, and dual luciferase reporter assays were performed to uncover the underlying cellular mechanism. Clinical IA samples were used to determine the expression of PDLIM1 and its downstream signaling molecules. RESULTS PDLIM1 expression was reduced in the endothelial cells of IA and was regulated by Yes-associated protein 1 (YAP1). Genetic silencing of PDLIM1 inhibited the viability, migratory ability, and tube formation ability of VECs. Opposite results were obtained by ectopic expression of PDLIM1. Additionally, PDLIM1 overexpression mitigated IA in vivo. Mechanistic investigations revealed that PDLIM1 promoted the transcriptional activity of β-catenin and induced the expression of v-myc myelocytomatosis viral oncogene homolog (MYC) and cyclin D1 (CCND1). In clinical settings, reduced expression of PDLIM1 and β-catenin downstream target genes was observed in human IA samples. CONCLUSION Our study indicates that YAP1-dependent expression of PDLIM1 can inhibit IA development by modulating the activity of the Wnt/β-catenin signaling pathway and that PDLIM1 deficiency in VECs may represent a potential marker of aggressive disease.
Collapse
Affiliation(s)
- Yan Yan
- Department of Neurosurgery, The 1st Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuanfeng Qin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongtao Zheng
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Jin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingzhu An
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing Leng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Kim D, Olson JM, Cooper JA. N-cadherin dynamically regulates pediatric glioma cell migration in complex environments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.04.535599. [PMID: 38260559 PMCID: PMC10802396 DOI: 10.1101/2023.04.04.535599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that inter-cellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.
Collapse
Affiliation(s)
- Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James M Olson
- Clinical Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
| | - Jonathan A Cooper
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| |
Collapse
|
38
|
Guo Y, Zhang S, Wang D, Heng BC, Deng X. Role of cell rearrangement and related signaling pathways in the dynamic process of tip cell selection. Cell Commun Signal 2024; 22:24. [PMID: 38195565 PMCID: PMC10777628 DOI: 10.1186/s12964-023-01364-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/25/2023] [Indexed: 01/11/2024] Open
Abstract
Angiogenesis is a complex, highly-coordinated and multi-step process of new blood vessel formation from pre-existing blood vessels. When initiated, the sprouting process is spearheaded by the specialized endothelial cells (ECs) known as tip cells, which guide the organization of accompanying stalk cells and determine the function and morphology of the finally-formed blood vessels. Recent studies indicate that the orchestration and coordination of angiogenesis involve dynamic tip cell selection, which is the competitive selection of cells to lead the angiogenic sprouts. Therefore, this review attempt to summarize the underlying mechanisms involved in tip cell specification in a dynamic manner to enable readers to gain a systemic and overall understanding of tip cell formation, involving cooperative interaction of cell rearrangement with Notch and YAP/TAZ signaling. Various mechanical and chemical signaling cues are integrated to ensure the right number of cells at the right place during angiogenesis, thereby precisely orchestrating morphogenic functions that ensure correct patterning of blood vessels. Video Abstract.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
39
|
Kobayashi S, Cox AG, Harvey KF, Hogan BM. Vasculature is getting Hip(po): Hippo signaling in vascular development and disease. Dev Cell 2023; 58:2627-2640. [PMID: 38052179 DOI: 10.1016/j.devcel.2023.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/29/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023]
Abstract
The Hippo signaling pathway regulates developmental organ growth, regeneration, and cell fate decisions. Although the role of the Hippo pathway, and its transcriptional effectors YAP and TAZ, has been well documented in many cell types and species, only recently have the roles for this pathway come to light in vascular development and disease. Experiments in mice, zebrafish, and in vitro have uncovered roles for the Hippo pathway, YAP, and TAZ in vasculogenesis, angiogenesis, and lymphangiogenesis. In addition, the Hippo pathway has been implicated in vascular cancers and cardiovascular diseases, thus identifying it as a potential therapeutic target for the treatment of these conditions. However, despite recent advances, Hippo's role in the vasculature is still underappreciated compared with its role in epithelial tissues. In this review, we appraise our current understanding of the Hippo pathway in blood and lymphatic vessel development and highlight the current knowledge gaps and opportunities for further research.
Collapse
Affiliation(s)
- Sakurako Kobayashi
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew G Cox
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kieran F Harvey
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
40
|
Cao J, Zhang Z, Zhou L, Luo M, Li L, Li B, Nice EC, He W, Zheng S, Huang C. Oncofetal reprogramming in tumor development and progression: novel insights into cancer therapy. MedComm (Beijing) 2023; 4:e427. [PMID: 38045829 PMCID: PMC10693315 DOI: 10.1002/mco2.427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
Emerging evidence indicates that cancer cells can mimic characteristics of embryonic development, promoting their development and progression. Cancer cells share features with embryonic development, characterized by robust proliferation and differentiation regulated by signaling pathways such as Wnt, Notch, hedgehog, and Hippo signaling. In certain phase, these cells also mimic embryonic diapause and fertilized egg implantation to evade treatments or immune elimination and promote metastasis. Additionally, the upregulation of ATP-binding cassette (ABC) transporters, including multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 1 (MRP1), and breast cancer-resistant protein (BCRP), in drug-resistant cancer cells, analogous to their role in placental development, may facilitate chemotherapy efflux, further resulting in treatment resistance. In this review, we concentrate on the underlying mechanisms that contribute to tumor development and progression from the perspective of embryonic development, encompassing the dysregulation of developmental signaling pathways, the emergence of dormant cancer cells, immune microenvironment remodeling, and the hyperactivation of ABC transporters. Furthermore, we synthesize and emphasize the connections between cancer hallmarks and embryonic development, offering novel insights for the development of innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Jiangjun Cao
- West China School of Basic Medical Sciences and Forensic Medicine, and Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Zhe Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Diseasethe First Affiliated HospitalSchool of MedicineZhejiang UniversityZhejiangChina
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Department of Infectious Diseasesthe Second Affiliated HospitalInstitute for Viral Hepatitis, Chongqing Medical UniversityChongqingChina
| | - Maochao Luo
- West China School of Basic Medical Sciences and Forensic Medicine, and Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Lei Li
- Department of anorectal surgeryHospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese MedicineChengduChina
| | - Bowen Li
- West China School of Basic Medical Sciences and Forensic Medicine, and Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
| | - Weifeng He
- State Key Laboratory of TraumaBurn and Combined InjuryInstitute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Shaojiang Zheng
- Hainan Cancer Medical Center of The First Affiliated Hospital, the Hainan Branch of National Clinical Research Center for Cancer, Hainan Engineering Research Center for Biological Sample Resources of Major DiseasesHainan Medical UniversityHaikouChina
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Women and Children's Medical Center, Key Laboratory of Emergency and Trauma of Ministry of EducationHainan Medical UniversityHaikouChina
| | - Canhua Huang
- West China School of Basic Medical Sciences and Forensic Medicine, and Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
41
|
Liu J, Wei Q, Man K, Liang C, Zhou Y, Liu X, Xin HB, Yang Y. Nanofibrous Membrane Promotes and Sustains Vascular Endothelial Barrier Function. ACS APPLIED BIO MATERIALS 2023; 6:4988-4997. [PMID: 37862245 DOI: 10.1021/acsabm.3c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023]
Abstract
The vascular endothelium serves as a physical barrier between the circulating blood and surrounding tissue and acts as a critical regulator of various physiological processes. In vitro models involving vasculature rely on the maintenance of the endothelial barrier function. In this study, we fabricated 2D aligned nanofibrous membranes with distinct pore sizes via electrospinning and investigated the effect of membrane pore size on endothelial barrier function. Our results demonstrated that the use of the nanofibrous membranes promoted the formation of a tight vascular endothelium and sustained barrier function for over one month in comparison with conventional transwell setups. Moreover, the examination of the nucleocytoplasmic localization of yes-associated protein (YAP) in the endothelial cells indicated that nanofibrous membrane promoted YAP expression and its nuclear localization, critical to endothelial barrier function. Furthermore, the comparison of permeability between random and aligned nanofibrous membranes underscored the importance of pore size in preserving barrier function. Our findings offer a valuable strategy for creating more physiologically relevant in vitro vascular models and contribute to the understanding of endothelial barrier formation and maintenance mechanisms.
Collapse
Affiliation(s)
- Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Qiang Wei
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Cindy Liang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yuting Zhou
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266073, P. R. China
| | - Xiaohua Liu
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, Missouri 65211, United States
| | - Hong-Bo Xin
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
42
|
Chen L, Qiu X, Dupre A, Pellon-Cardenas O, Fan X, Xu X, Rout P, Walton KD, Burclaff J, Zhang R, Fang W, Ofer R, Logerfo A, Vemuri K, Bandyopadhyay S, Wang J, Barbet G, Wang Y, Gao N, Perekatt AO, Hu W, Magness ST, Spence JR, Verzi MP. TGFB1 induces fetal reprogramming and enhances intestinal regeneration. Cell Stem Cell 2023; 30:1520-1537.e8. [PMID: 37865088 PMCID: PMC10841757 DOI: 10.1016/j.stem.2023.09.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/03/2023] [Accepted: 09/28/2023] [Indexed: 10/23/2023]
Abstract
The gut epithelium has a remarkable ability to recover from damage. We employed a combination of high-throughput sequencing approaches, mouse genetics, and murine and human organoids and identified a role for TGFB signaling during intestinal regeneration following injury. At 2 days following irradiation (IR)-induced damage of intestinal crypts, a surge in TGFB1 expression is mediated by monocyte/macrophage cells at the location of damage. The depletion of macrophages or genetic disruption of TGFB signaling significantly impaired the regenerative response. Intestinal regeneration is characterized by the induction of a fetal-like transcriptional signature during repair. In organoid culture, TGFB1 treatment was necessary and sufficient to induce the fetal-like/regenerative state. Mesenchymal cells were also responsive to TGFB1 and enhanced the regenerative response. Mechanistically, pro-regenerative factors, YAP/TEAD and SOX9, are activated in the epithelium exposed to TGFB1. Finally, pre-treatment with TGFB1 enhanced the ability of primary epithelial cultures to engraft into damaged murine colon, suggesting promise for cellular therapy.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China.
| | - Xia Qiu
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Abigail Dupre
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Oscar Pellon-Cardenas
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Xiaojiao Fan
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Xiaoting Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Prateeksha Rout
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Katherine D Walton
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Chapel Hill, NC 27695, USA; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Ruolan Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Wenxin Fang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Rachel Ofer
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Alexandra Logerfo
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Kiranmayi Vemuri
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA
| | - Sheila Bandyopadhyay
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102, USA
| | - Jianming Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University-New Brunswick, New Brunswick, NJ 08903, USA
| | - Gaetan Barbet
- Child Health Institute of New Jersey, Rutgers University-New Brunswick, New Brunswick, NJ 08901, USA
| | - Yan Wang
- Center for Translation Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Nan Gao
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102, USA
| | - Ansu O Perekatt
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University-New Brunswick, New Brunswick, NJ 08903, USA
| | - Scott T Magness
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Chapel Hill, NC 27695, USA; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Michael P Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 00854, USA; Rutgers Cancer Institute of New Jersey, Rutgers University-New Brunswick, New Brunswick, NJ 08903, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University-New Brunswick, New Brunswick, NJ 08901, USA; NIEHS Center for Environmental Exposures and Disease (CEED), Rutgers EOHSI, Piscataway, NJ 08854, USA.
| |
Collapse
|
43
|
Romeo SG, Secco I, Schneider E, Reumiller CM, Santos CXC, Zoccarato A, Musale V, Pooni A, Yin X, Theofilatos K, Trevelin SC, Zeng L, Mann GE, Pathak V, Harkin K, Stitt AW, Medina RJ, Margariti A, Mayr M, Shah AM, Giacca M, Zampetaki A. Human blood vessel organoids reveal a critical role for CTGF in maintaining microvascular integrity. Nat Commun 2023; 14:5552. [PMID: 37689702 PMCID: PMC10492781 DOI: 10.1038/s41467-023-41326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
The microvasculature plays a key role in tissue perfusion and exchange of gases and metabolites. In this study we use human blood vessel organoids (BVOs) as a model of the microvasculature. BVOs fully recapitulate key features of the human microvasculature, including the reliance of mature endothelial cells on glycolytic metabolism, as concluded from metabolic flux assays and mass spectrometry-based metabolomics using stable tracing of 13C-glucose. Pharmacological targeting of PFKFB3, an activator of glycolysis, using two chemical inhibitors results in rapid BVO restructuring, vessel regression with reduced pericyte coverage. PFKFB3 mutant BVOs also display similar structural remodelling. Proteomic analysis of the BVO secretome reveal remodelling of the extracellular matrix and differential expression of paracrine mediators such as CTGF. Treatment with recombinant CTGF recovers microvessel structure. In this work we demonstrate that BVOs rapidly undergo restructuring in response to metabolic changes and identify CTGF as a critical paracrine regulator of microvascular integrity.
Collapse
Affiliation(s)
- Sara G Romeo
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Ilaria Secco
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Edoardo Schneider
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Christina M Reumiller
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Celio X C Santos
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Anna Zoccarato
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Vishal Musale
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Aman Pooni
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Xiaoke Yin
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Konstantinos Theofilatos
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Silvia Cellone Trevelin
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Lingfang Zeng
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Giovanni E Mann
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Varun Pathak
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Kevin Harkin
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Alan W Stitt
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Reinhold J Medina
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Andriana Margariti
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Mauro Giacca
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Anna Zampetaki
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK.
| |
Collapse
|
44
|
Nan W, He Y, Wang S, Zhang Y. Molecular mechanism of VE-cadherin in regulating endothelial cell behaviour during angiogenesis. Front Physiol 2023; 14:1234104. [PMID: 37601629 PMCID: PMC10433914 DOI: 10.3389/fphys.2023.1234104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Vascular endothelial (VE)-cadherin, an endothelium-specific adhesion protein, is found in the junctions between endothelial cells (ECs). It's crucial to maintain the homogeneity of ECs. Keeping and controlling the contact between ECs is essential. In addition to its adhesive function, VE-cadherin plays important roles in vascular development, permeability, and tumour angiogenesis. Signal transfer, cytoskeletal reconstruction, and contractile integrating, which are crucial for constructing and maintaining monolayer integrity as well as for repair and regeneration, are the foundation of endothelial cell (EC) junctional dynamics. The molecular basis of adhesion junctions (AJs), which are closely related and work with actin filaments, is provided by the VE-cadherin-catenin complex. They can activate intracellular signals that drive ECs to react or communicate structural changes to junctions. An increasing number of molecules, including the vascular endothelial growth factor receptor 2 (VEGFR2) and vascular endothelial protein tyrosine phosphatase (VE-PTP), have been connected to VE-cadherin in addition to the conventional VE-cadherin-catenin complex. This review demonstrates significant progress in our understanding of the molecular mechanisms that affect VE-cadherin's function in the regulation of EC behaviour during angiogenesis. The knowledge of the molecular processes that control VE-cadherin's role in the regulation of EC behaviour during angiogenesis has recently advanced, as shown in this review.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
45
|
Rodríguez TC, Kwan S, Smith JL, Dadafarin S, Wu CH, Sontheimer EJ, Xue W. Multiomics characterization of mouse hepatoblastoma identifies yes-associated protein 1 target genes. Hepatology 2023; 78:58-71. [PMID: 35932276 PMCID: PMC10205091 DOI: 10.1002/hep.32713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Hepatoblastoma (HB) is the most common primary liver malignancy in childhood and lacks targeted therapeutic options. We previously engineered, to our knowledge, the first yes-associated protein 1 (YAP1) S127A -inducible mouse model of HB, demonstrating tumor regression and redifferentiation after YAP1 withdrawal through genome-wide enhancer modulation. Probing accessibility, transcription, and YAP1 binding at regulatory elements in HB tumors may provide more insight into YAP1-driven tumorigenesis and expose exploitable vulnerabilities in HB. APPROACH AND RESULTS Using a multiomics approach, we integrated high-throughput transcriptome and chromatin profiling of our murine HB model to identify dynamic activity at candidate cis -regulatory elements (cCREs). We observed that 1301 of 305,596 cCREs exhibit "tumor-modified" (TM) accessibility in HB. We mapped 241 TM enhancers to corresponding genes using accessibility and histone H3K27Ac profiles. Anti-YAP1 cleavage under targets and tagmentation in tumors revealed 66 YAP1-bound TM cCRE/gene pairs, 31 of which decrease expression after YAP1 withdrawal. We validated the YAP1-dependent expression of a putative YAP1 target, Jun dimerization protein 2 (JDP2), in human HB cell lines using YAP1 and LATS1/2 small interfering RNA knockdown. We also confirmed YAP1-induced activity of the Jdp2 TM enhancer in vitro and discovered an analogous human enhancer in silico. Finally, we used transcription factor (TF) footprinting to identify putative YAP1 cofactors and characterize HB-specific TF activity genome wide. CONCLUSIONS Our chromatin-profiling techniques define the regulatory frameworks underlying HB and identify YAP1-regulated gene/enhancer pairs. JDP2 is an extensively validated target with YAP1-dependent expression in human HB cell lines and hepatic malignancies.
Collapse
Affiliation(s)
- Tomás C. Rodríguez
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - SuetYan Kwan
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Jordan L. Smith
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | | | - Chern-Horng Wu
- Division of Internal Medicine and Primary Care, Tufts Medical Center, 800 Washington, Boston, MA, 02111
| | - Erik J. Sontheimer
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
46
|
Constantinou I, Bastounis EE. Cell-stretching devices: advances and challenges in biomedical research and live-cell imaging. Trends Biotechnol 2023; 41:939-950. [PMID: 36604290 DOI: 10.1016/j.tibtech.2022.12.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/04/2023]
Abstract
Basic human functions such as breathing and digestion require mechanical stretching of cells and tissues. However, when it comes to laboratory experiments, the mechanical stretching that cells experience in the body is not often replicated, limiting the biomimetic nature of the studies and the relevance of results. Herein, we establish the importance of mechanical stretching during in vitro investigations by reviewing seminal works performed using cell-stretching platforms, highlighting important outcomes of these works as well as the engineering characteristics of the platforms used. Emphasis is placed on the compatibility of cell-stretching devices (CSDs) with live-cell imaging as well as their limitations and on the research advancements that could arise from live-cell imaging performed during cell stretching.
Collapse
Affiliation(s)
- Iordania Constantinou
- Institute of Microtechnology (IMT), Technische Universität Braunschweig, Alte Salzdahlumer Str. 203, 38124 Braunschweig, Germany; Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany.
| | - Effie E Bastounis
- Institute of Microbiology and Infection Medicine (IMIT), Eberhard Karls University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" EXC 2124, Eberhard Karls University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany.
| |
Collapse
|
47
|
Pontes B, Mendes FA. Mechanical Properties of Glioblastoma: Perspectives for YAP/TAZ Signaling Pathway and Beyond. Diseases 2023; 11:86. [PMID: 37366874 DOI: 10.3390/diseases11020086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Glioblastoma is a highly aggressive brain tumor with a poor prognosis. Recent studies have suggested that mechanobiology, the study of how physical forces influence cellular behavior, plays an important role in glioblastoma progression. Several signaling pathways, molecules, and effectors, such as focal adhesions, stretch-activated ion channels, or membrane tension variations, have been studied in this regard. Also investigated are YAP/TAZ, downstream effectors of the Hippo pathway, which is a key regulator of cell proliferation and differentiation. In glioblastoma, YAP/TAZ have been shown to promote tumor growth and invasion by regulating genes involved in cell adhesion, migration, and extracellular matrix remodeling. YAP/TAZ can be activated by mechanical cues such as cell stiffness, matrix rigidity, and cell shape changes, which are all altered in the tumor microenvironment. Furthermore, YAP/TAZ have been shown to crosstalk with other signaling pathways, such as AKT, mTOR, and WNT, which are dysregulated in glioblastoma. Thus, understanding the role of mechanobiology and YAP/TAZ in glioblastoma progression could provide new insights into the development of novel therapeutic strategies. Targeting YAP/TAZ and mechanotransduction pathways in glioblastoma may offer a promising approach to treating this deadly disease.
Collapse
Affiliation(s)
- Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Fabio A Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
48
|
Choi S, Hong SP, Bae JH, Suh SH, Bae H, Kang KP, Lee HJ, Koh GY. Hyperactivation of YAP/TAZ Drives Alterations in Mesangial Cells through Stabilization of N-Myc in Diabetic Nephropathy. J Am Soc Nephrol 2023; 34:809-828. [PMID: 36724799 PMCID: PMC10125647 DOI: 10.1681/asn.0000000000000075] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 12/18/2022] [Indexed: 01/26/2023] Open
Abstract
SIGNIFICANCE STATEMENT Mesangial cells (MCs) in the kidney are essential to maintaining glomerular integrity, and their impairment leads to major glomerular diseases including diabetic nephropathy (DN). Although high blood glucose elicits abnormal alterations in MCs, the underlying mechanism is poorly understood. We show that YAP/TAZ are increased in MCs of patients with DN and two animal models of DN. High glucose directly induces activation of YAP/TAZ through the canonical Hippo pathway in cultured MCs. Hyperactivation of YAP/TAZ in mouse MCs recapitulates the hallmarks of DN. Activated YAP/TAZ bind and stabilize N-Myc, one of the Myc family. N-Myc stabilization leads to aberrant enhancement of its transcriptional activity and to MC impairments. Our findings shed light on how high blood glucose in diabetes mellitus leads to DN and support a rationale that lowering blood glucose in diabetes mellitus could delay DN pathogenesis. BACKGROUND Mesangial cells (MCs) in the kidney are central to maintaining glomerular integrity, and their impairment leads to major glomerular diseases, including diabetic nephropathy (DN). Although high blood glucose elicits abnormal alterations in MCs, the underlying molecular mechanism is poorly understood. METHODS Immunolocalization of YAP/TAZ and pathological features of PDGFRβ + MCs were analyzed in the glomeruli of patients with DN, in Zucker diabetic fatty rats, and in Lats1/2i ΔPβ mice. RiboTag bulk-RNA sequencing and transcriptomic analysis of gene expression profiles of the isolated MCs from control and Lats1/2iΔPβ mice were performed. Immunoprecipitation analysis and protein stability of N-Myc were performed by the standard protocols. RESULTS YAP and TAZ, the final effectors of the Hippo pathway, are highly increased in MCs of patients with DN and in Zucker diabetic fatty rats. Moreover, high glucose directly induces activation of YAP/TAZ through the canonical Hippo pathway in cultured MCs. Hyperactivation of YAP/TAZ in mouse model MCs recapitulates the hallmarks of DN, including excessive proliferation of MCs and extracellular matrix deposition, endothelial cell impairment, glomerular sclerosis, albuminuria, and reduced glomerular filtration rate. Mechanistically, activated YAP/TAZ bind and stabilize N-Myc protein, one of the Myc family of oncogenes. N-Myc stabilization leads to aberrant enhancement of its transcriptional activity and eventually to MC impairments and DN pathogenesis. CONCLUSIONS Our findings shed light on how high blood glucose in diabetes mellitus leads to DN and support a rationale that lowering blood glucose in diabetes mellitus could delay DN pathogenesis.
Collapse
Affiliation(s)
- Seunghyeok Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seon Pyo Hong
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jung Hyun Bae
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Sang Heon Suh
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hosung Bae
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Research Institute of Clinical Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Hyuek Jong Lee
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| |
Collapse
|
49
|
Kang JH, Jang M, Seo SJ, Choi A, Shin D, Seo S, Lee SH, Kim HN. Mechanobiological Adaptation to Hyperosmolarity Enhances Barrier Function in Human Vascular Microphysiological System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206384. [PMID: 36808839 PMCID: PMC10161024 DOI: 10.1002/advs.202206384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Indexed: 05/06/2023]
Abstract
In infectious disease such as sepsis and COVID-19, blood vessel leakage treatment is critical to prevent fatal progression into multi-organ failure and ultimately death, but the existing effective therapeutic modalities that improve vascular barrier function are limited. Here, this study reports that osmolarity modulation can significantly improve vascular barrier function, even in an inflammatory condition. 3D human vascular microphysiological systems and automated permeability quantification processes for high-throughput analysis of vascular barrier function are utilized. Vascular barrier function is enhanced by >7-folds with 24-48 h hyperosmotic exposure (time window of emergency care; >500 mOsm L-1 ) but is disrupted after hypo-osmotic exposure (<200 mOsm L-1 ). By integrating genetic and protein level analysis, it is shown that hyperosmolarity upregulates vascular endothelial-cadherin, cortical F-actin, and cell-cell junction tension, indicating that hyperosmotic adaptation mechanically stabilizes the vascular barrier. Importantly, improved vascular barrier function following hyperosmotic exposure is maintained even after chronic exposure to proinflammatory cytokines and iso-osmotic recovery via Yes-associated protein signaling pathways. This study suggests that osmolarity modulation may be a unique therapeutic strategy to proactively prevent infectious disease progression into severe stages via vascular barrier function protection.
Collapse
Affiliation(s)
- Joon Ho Kang
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Minjeong Jang
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Su Jin Seo
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Department of Chemical EngineeringKwangwoon UniversitySeoul01897Republic of Korea
| | - Andrew Choi
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Daeeun Shin
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- School of Mechanical EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Suyoung Seo
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Program in Nano Science and TechnologyGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul08826Republic of Korea
| | - Soo Hyun Lee
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
50
|
Parab S, Setten E, Astanina E, Bussolino F, Doronzo G. The tissue-specific transcriptional landscape underlines the involvement of endothelial cells in health and disease. Pharmacol Ther 2023; 246:108418. [PMID: 37088448 DOI: 10.1016/j.pharmthera.2023.108418] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/23/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Endothelial cells (ECs) that line vascular and lymphatic vessels are being increasingly recognized as important to organ function in health and disease. ECs participate not only in the trafficking of gases, metabolites, and cells between the bloodstream and tissues but also in the angiocrine-based induction of heterogeneous parenchymal cells, which are unique to their specific tissue functions. The molecular mechanisms regulating EC heterogeneity between and within different tissues are modeled during embryogenesis and become fully established in adults. Any changes in adult tissue homeostasis induced by aging, stress conditions, and various noxae may reshape EC heterogeneity and induce specific transcriptional features that condition a functional phenotype. Heterogeneity is sustained via specific genetic programs organized through the combinatory effects of a discrete number of transcription factors (TFs) that, at the single tissue-level, constitute dynamic networks that are post-transcriptionally and epigenetically regulated. This review is focused on outlining the TF-based networks involved in EC specialization and physiological and pathological stressors thought to modify their architecture.
Collapse
Affiliation(s)
- Sushant Parab
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elisa Setten
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elena Astanina
- Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy.
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| |
Collapse
|