1
|
Abedinzadeh M, Nazari A, Vahidi SA, Pourmasumi S, Khorramdelazad H. Critical Roles of VEGFR1, VEGFR2, VEGFR3, BAX, and BCL-2 in the Pathogenesis of Varicose Veins: Unveiling Molecular Mechanisms. Am J Mens Health 2025; 19:15579883251321588. [PMID: 40079569 PMCID: PMC11907509 DOI: 10.1177/15579883251321588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 03/15/2025] Open
Abstract
Varicocele is characterized by the abnormal dilation of veins within the testicular pampiniform plexus, contributing to inflammation, pain, and infertility in males. The precise roles of vascular endothelial growth factor receptors (VEGFRs), B-cell lymphoma 2 (BCL-2), and BCL-2-associated X-protein (BAX) in the pathology of varicocele still need to be clarified. This study sought to investigate the protein expression levels of VEGFR1, VEGFR2, VEGFR3, BCL-2, and BAX in varicose and healthy vessels from patients diagnosed with varicocele. Tissue samples were collected from 20 varicose veins and 20 healthy vessels from patients diagnosed with varicocele. Western blotting was utilized to quantify VEGFR1, VEGFR2, VEGFR3, BCL-2, and BAX protein levels. Analysis revealed a statistically significant increase in VEGFR3 protein expression within varicose veins compared to healthy vessels (p = .0473), while no significant differences were observed in the levels of VEGFR1 and VEGFR2 between the two groups. Concerning apoptotic signaling proteins, no significant differences were noted in the individual expression levels of BAX and BCL-2; however, the BAX/BCL-2 ratio was approximately 1.29 in varicose vessels. This ratio, exceeding 1.0, may suggest a pro-apoptotic shift in varicose veins and indicates a potential involvement of apoptosis in the pathology of varicocele. These findings suggest that VEGFR3 may play a pivotal role in the pathogenesis of varicocele and could contribute to vascular alterations associated with this condition. Furthermore, the elevated BAX/BCL-2 ratio implies a pro-apoptotic environment within varicose veins, thereby implicating apoptosis as a possible mechanism in the development of varicocele. Further exploration of VEGFR3-related signaling pathways and apoptotic markers may yield valuable insights for identifying therapeutic targets in managing varicocele.
Collapse
Affiliation(s)
- Mehdi Abedinzadeh
- Department of Urology, Shahid Rahnemoon Hospital, Shahid Sadoughi University of Medical Sciences and Health Service, Yazd, Iran
| | - Alireza Nazari
- Social Determinants of Health Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Andrology Research Center, Yazd Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seraj-Aldin Vahidi
- Andrology Research Center, Yazd Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Soheila Pourmasumi
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Clinical Research Development Unit, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
2
|
Cho WJ, Elbasiony E, Singh A, Mittal SK, Chauhan SK. IL-36γ Augments Ocular Angiogenesis by Promoting the Vascular Endothelial Growth Factor-Vascular Endothelial Growth Factor Receptor Axis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1740-1749. [PMID: 36740182 PMCID: PMC10616713 DOI: 10.1016/j.ajpath.2023.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/05/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023]
Abstract
Prevention of inflammatory angiogenesis is critical for suppressing chronic inflammation and inhibiting inflammatory tissue damage. Angiogenesis is particularly detrimental to the cornea because pathologic growth of new blood vessels can lead to marked vision impairment and even loss of vision. The expression of proinflammatory cytokines by injured tissues exacerbates the inflammatory cascade, including angiogenesis. IL-36 cytokine, a subfamily of the IL-1 superfamily, consists of three proinflammatory agonists, IL-36α, IL-36β, and IL-36γ, and an IL-36 receptor antagonist (IL-36Ra). Data from the current study indicate that human vascular endothelial cells constitutively expressed the cognate IL-36 receptor. The current investigation, for the first time, characterized the direct contribution of IL-36γ to various angiogenic processes. IL-36γ up-regulated the expression of vascular endothelial growth factors (VEGFs) and their receptors VEGFR2 and VEGFR3 by human vascular endothelial cells, suggesting that IL-36γ mediates the VEGF-VEGFR signaling by endothelial cells. Moreover, by using a naturally occurring antagonist IL-36Ra in a murine model of inflammatory angiogenesis, this study demonstrated that blockade of endogenous IL-36γ signaling results in significant retardation of inflammatory angiogenesis. The current investigation on the proangiogenic function of IL-36γ provides novel evidence of the development of IL-36γ-targeting strategies to hamper inflammatory angiogenesis.
Collapse
Affiliation(s)
- WonKyung J Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Aastha Singh
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
3
|
Bokhari SMZ, Hamar P. Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors. Int J Mol Sci 2023; 24:13317. [PMID: 37686121 PMCID: PMC10487419 DOI: 10.3390/ijms241713317] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Vascular endothelial growth factors (VEGFs) are the key regulators of vasculogenesis in normal and oncological development. VEGF-A is the most studied angiogenic factor secreted by malignant tumor cells under hypoxic and inflammatory stress, which made VEGF-A a rational target for anticancer therapy. However, inhibition of VEGF-A by monoclonal antibody drugs led to the upregulation of VEGF-D. VEGF-D was primarily described as a lymphangiogenic factor; however, VEGF-D's blood angiogenic potential comparable to VEGF-A has already been demonstrated in glioblastoma and colorectal carcinoma. These findings suggested a role for VEGF-D in facilitating malignant tumor growth by bypassing the anti-VEGF-A antiangiogenic therapy. Owing to its high mitogenic ability, higher affinity for VEGFR-2, and higher expression in cancer, VEGF-D might even be a stronger angiogenic driver and, hence, a better therapeutic target than VEGF-A. In this review, we summarized the angiogenic role of VEGF-D in blood vasculogenesis and its targetability as an antiangiogenic therapy in cancer.
Collapse
Affiliation(s)
| | - Peter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
| |
Collapse
|
4
|
Fernandez-Flores A, Cassarino D, Colmenero I. Vascular Malformations: A Histopathologic and Conceptual Appraisal. ACTAS DERMO-SIFILIOGRAFICAS 2023; 114:T213-T228. [PMID: 36690143 DOI: 10.1016/j.ad.2022.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 01/22/2023] Open
Abstract
In the field of vascular anomalies, distinguishing between vascular malformations and tumors has become crucial for a correct therapeutic approach. However, the differential diagnosis between these two groups is not always well explained in classical texts, mainly because many vascular malformations are still known with old names that suggest a tumoral nature. Also, genetic and pathogenic knowledge of these entities has greatly increased in recent decades, so researchers and clinicians now have a better understanding of vascular malformations. In this paper, we present the main histopathological tips to recognize and identify a vascular malformation as such. We also contextualize such information in the clinical and pathogenic knowledge for a better understanding of these entities.
Collapse
Affiliation(s)
- A Fernandez-Flores
- Departamento de Histopatología, Hospital Universitario El Bierzo, Ponferrada, León, España; Departamento de Patología Celular, Hospital de la Reina, Ponferrada, León, España; Departamento de Investigación, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidad de A Coruña, A Coruña, España.
| | - D Cassarino
- Los Angeles Medical Center (LAMC), Southern California Kaiser Permanente, Department of Dermatology, Los Ángeles, CA, EE. UU
| | - I Colmenero
- Departamento de Histopatología Pediátrica, Hospital Universitario Niño Jesús, Madrid, España
| |
Collapse
|
5
|
Fernandez-Flores A, Cassarino D, Colmenero I. Vascular Malformations: A Histopathologic and Conceptual Appraisal. ACTAS DERMO-SIFILIOGRAFICAS 2023; 114:213-228. [PMID: 36309042 DOI: 10.1016/j.ad.2022.10.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 12/13/2022] Open
Abstract
In the field of vascular anomalies, distinguishing between vascular malformations and tumors has become crucial for a correct therapeutic approach. However, the differential diagnosis between these two groups is not always well explained in classical texts, mainly because many vascular malformations are still known with old names that suggest a tumoral nature. Also, genetic and pathogenic knowledge of these entities has greatly increased in recent decades, so researchers and clinicians now have a better understanding of vascular malformations. In this paper, we present the main histopathological tips to recognize and identify a vascular malformation as such. We also contextualize such information in the clinical and pathogenic knowledge for a better understanding of these entities.
Collapse
Affiliation(s)
- A Fernandez-Flores
- Department of Histopathology, University Hospital El Bierzo, Ponferrada, Spain; Department of Cellular Pathology, Hospital de la Reina, Ponferrada, Spain; Research Department, Institute for Biomedical Research of A Coruña (INIBIC), University of A Coruña (UDC), A Coruña, Spain.
| | - D Cassarino
- Los Angeles Medical Center (LAMC), Southern California Kaiser Permanente, Department of Dermatology, Los Angeles, CA, USA
| | - I Colmenero
- Department of Pediatric Histopathology, University Hospital Niño Jesús, Madrid, Spain
| |
Collapse
|
6
|
McCright J, Naiknavare R, Yarmovsky J, Maisel K. Targeting Lymphatics for Nanoparticle Drug Delivery. Front Pharmacol 2022; 13:887402. [PMID: 35721179 PMCID: PMC9203826 DOI: 10.3389/fphar.2022.887402] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/25/2022] Open
Abstract
The lymphatics transport material from peripheral tissues to lymph nodes, where immune responses are formed, before being transported into systemic circulation. With key roles in transport and fluid homeostasis, lymphatic dysregulation is linked to diseases, including lymphedema. Fluid within the interstitium passes into initial lymphatic vessels where a valve system prevents fluid backflow. Additionally, lymphatic endothelial cells produce key chemokines, such as CCL21, that direct the migration of dendritic cells and lymphocytes. As a result, lymphatics are an attractive delivery route for transporting immune modulatory treatments to lymph nodes where immunotherapies are potentiated in addition to being an alternative method of reaching systemic circulation. In this review, we discuss the physiology of lymphatic vessels and mechanisms used in the transport of materials from peripheral tissues to lymph nodes. We then summarize nanomaterial-based strategies to take advantage of lymphatic transport functions for delivering therapeutics to lymph nodes or systemic circulation. We also describe opportunities for targeting lymphatic endothelial cells to modulate transport and immune functions.
Collapse
Affiliation(s)
| | | | | | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, MD, United States
| |
Collapse
|
7
|
Kawashima T, Ji RC, Itoh Y, Agata N, Sasai N, Murakami T, Sokabe M, Hamada F, Kawakami K. Morphological and biochemical changes of lymphatic vessels in the soleus muscle of mice after hindlimb unloading. Muscle Nerve 2021; 64:620-628. [PMID: 34409627 DOI: 10.1002/mus.27402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 11/07/2022]
Abstract
INTRODUCTION/AIMS Lymphatic vessels are responsible for the removal of metabolic waste from body tissues. They also play a crucial role in skeletal muscle functioning thorough their high-energy metabolism. In this study we investigated whether disuse muscle atrophy induced by hindlimb unloading is associated with an alteration in the number of lymphatic vessels and differential expression of lymphangiogenic factors in the soleus muscle. METHODS Male C57BL/6 mice were subjected to tail suspension (TS) for 2 or 4 weeks to induce soleus muscle atrophy. After TS, lymphatic and blood capillaries in the soleus muscle were visualized and counted by double staining with LYVE-1 and CD31. The protein and mRNA levels of vascular endothelial growth factor (VEGF)-C, VEGF-D, and vascular endothelial growth factor receptor-3 were measured by Western blotting and real-time reverse transcript polymerase chain reaction, respectively. RESULTS TS for 2 weeks resulted in a significant decrease in the number of blood capillaries compared with controls. However, there was no significant change in the number of lymphatic capillaries. By contrast, TS for 4 weeks resulted in a significant decrease in the number of lymphatic and blood capillaries. We observed a significant decrease in the mRNA levels of VEGF-C and VEGF-D in mice subjected to TS for 4 weeks. DISCUSSION The decrease of intramuscular lymphatic vessels may a crucial role in the process of muscle atrophy.
Collapse
Affiliation(s)
- Takafumi Kawashima
- Department of Human Anatomy, Graduate School of Medicine, Oita University, Yufu, Japan
| | - Rui-Cheng Ji
- Department of Physical Therapy, Graduate School of Medicine, Oita University, Oita, Japan
| | - Yuta Itoh
- Faculty of Rehabilitation Science, Nagoya Gakuin University, Nagoya, Japan
| | - Nobuhide Agata
- Faculty of Health and Medical Sciences, Tokoha University, Hamamatsu, Japan
| | - Nobuaki Sasai
- Department of Physical Therapy, Graduate School of Medical Science & Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Japan
| | - Taro Murakami
- Faculty of Wellness, Shigakkan University, Ohbu, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumihiko Hamada
- Department of Human Anatomy, Graduate School of Medicine, Oita University, Yufu, Japan
| | - Keisuke Kawakami
- Department of Physical Therapy, Graduate School of Medicine, Oita University, Oita, Japan
| |
Collapse
|
8
|
Brinks J, van Dijk EHC, Klaassen I, Schlingemann RO, Kielbasa SM, Emri E, Quax PHA, Bergen AA, Meijer OC, Boon CJF. Exploring the choroidal vascular labyrinth and its molecular and structural roles in health and disease. Prog Retin Eye Res 2021; 87:100994. [PMID: 34280556 DOI: 10.1016/j.preteyeres.2021.100994] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022]
Abstract
The choroid is a key player in maintaining ocular homeostasis and plays a role in a variety of chorioretinal diseases, many of which are poorly understood. Recent advances in the field of single-cell RNA sequencing have yielded valuable insights into the properties of choroidal endothelial cells (CECs). Here, we review the role of the choroid in various physiological and pathophysiological mechanisms, focusing on the role of CECs. We also discuss new insights regarding the phenotypic properties of CECs, CEC subpopulations, and the value of measuring transcriptomics in primary CEC cultures derived from post-mortem eyes. In addition, we discuss key phenotypic, structural, and functional differences that distinguish CECs from other endothelial cells such as retinal vascular endothelial cells. Understanding the specific clinical and molecular properties of the choroid will shed new light on the pathogenesis of the broad clinical range of chorioretinal diseases such as age-related macular degeneration, central serous chorioretinopathy and other diseases within the pachychoroid spectrum, uveitis, and diabetic choroidopathy. Although our knowledge is still relatively limited with respect to the clinical features and molecular pathways that underlie these chorioretinal diseases, we summarise new approaches and discuss future directions for gaining new insights into these sight-threatening diseases and highlight new therapeutic strategies such as pluripotent stem cell‒based technologies and gene therapy.
Collapse
Affiliation(s)
- J Brinks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - E H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - I Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - R O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - S M Kielbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - E Emri
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - P H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - A A Bergen
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - O C Meijer
- Department of Medicine, Division of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands
| | - C J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Li W, Gauthier JM, Tong AY, Terada Y, Higashikubo R, Frye CC, Harrison MS, Hashimoto K, Bery AI, Ritter JH, Nava RG, Puri V, Wong BW, Lavine KJ, Bharat A, Krupnick AS, Gelman AE, Kreisel D. Lymphatic drainage from bronchus-associated lymphoid tissue in tolerant lung allografts promotes peripheral tolerance. J Clin Invest 2021; 130:6718-6727. [PMID: 33196461 DOI: 10.1172/jci136057] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 09/03/2020] [Indexed: 12/29/2022] Open
Abstract
Tertiary lymphoid organs are aggregates of immune and stromal cells including high endothelial venules and lymphatic vessels that resemble secondary lymphoid organs and can be induced at nonlymphoid sites during inflammation. The function of lymphatic vessels within tertiary lymphoid organs remains poorly understood. During lung transplant tolerance, Foxp3+ cells accumulate in tertiary lymphoid organs that are induced within the pulmonary grafts and are critical for the local downregulation of alloimmune responses. Here, we showed that tolerant lung allografts could induce and maintain tolerance of heterotopic donor-matched hearts through pathways that were dependent on the continued presence of the transplanted lung. Using lung retransplantation, we showed that Foxp3+ cells egressed from tolerant lung allografts via lymphatics and were recruited into donor-matched heart allografts. Indeed, survival of the heart allografts was dependent on lymphatic drainage from the tolerant lung allograft to the periphery. Thus, our work indicates that cellular trafficking from tertiary lymphoid organs regulates immune responses in the periphery. We propose that these findings have important implications for a variety of disease processes that are associated with the induction of tertiary lymphoid organs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jon H Ritter
- Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | | | | | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | | | - Andrew E Gelman
- Departments of Surgery.,Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Departments of Surgery.,Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Screening for potential targets to reduce stenosis in bioprosthetic heart valves. Sci Rep 2021; 11:2464. [PMID: 33510256 PMCID: PMC7843970 DOI: 10.1038/s41598-021-81340-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/24/2020] [Indexed: 12/02/2022] Open
Abstract
Progressive stenosis is one of the main factors that limit the lifetime of bioprosthetic valved conduits. To improve long-term performance we aimed to identify targets that inhibit pannus formation on conduit walls. From 11 explanted, obstructed, RNAlater presevered pulmonary valved conduits, we dissected the thickened conduit wall and the thin leaflet to determine gene expression-profiles using ultra deep sequencing. Differential gene expression between pannus and leaflet provided the dataset that was screened for potential targets. Promising target candidates were immunohistologically stained to see protein abundance and the expressing cell type(s). While immunostainings for DDR2 and FGFR2 remained inconclusive, EGFR, ErbB4 and FLT4 were specifically expressed in a subset of tissue macrophages, a cell type known to regulate the initiation, maintenance, and resolution of tissue repair. Taken toghether, our data suggest EGFR, ErbB4 and FLT4 as potential target candidates to limit pannus formation in bioprosthestic replacement valves.
Collapse
|
11
|
Arciniegas E, Carrillo LM, Rojas H, Pineda J, Ramírez R, Reyes O, Chopite M, Rocheta A. Plump endothelial cells integrated into pre-existing venules contribute to the formation of 'mother' and 'daughter' vessels in pyogenic granuloma: possible role of galectin-1, -3 and -8. Scars Burn Heal 2021; 7:2059513120986687. [PMID: 33796337 PMCID: PMC7841855 DOI: 10.1177/2059513120986687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Pyogenic granuloma (PG) is a reactive inflammatory vascular lesion of the skin and mucous membranes, characterised by the presence of enlarged venules and seamed and seamless capillaries with plump endothelial cells (EC), and numerous macrophages. EC activation upregulates the synthesis of galectins and induces their translocation to the EC surface promoting angiogenesis and lymphangiogenesis, particularly galectin-1 (Gal-1), Gal-3 and Gal-8. However, the presence and distribution of Gal-1, -3 and -8, as well as their implications in the pathogenesis of PG, has not been considered. MATERIALS AND METHODS Eight biopsies from patients diagnosed with PG were selected. The presence of PECAM-1/CD31, IL-1β, VEGF-C, VEGFR-2, VEGFR-3, integrin β1, CD44, fibronectin and Gal-1, -3 and -8 was assessed by immunofluorescence staining using confocal laser scanning microscopy. RESULTS AND DISCUSSION Immunostaining revealed that these molecules were present in the enlarged venules with plump ECs, in some macrophages and other immune cells. We propose that macrophages release VEGF-A and VEGF-C inducing VEGFR-2/VEGFR-3 expression and activation, leading macrophages to transdifferentiate into plump ECs that might integrate into pre-existing venules, contributing to the formation of enlarged venules with transluminal bridges and capillaries. EC activation, induced by certain cytokines, has been shown to stimulate galectin expression and changes in the cellular localisation through association and activation of specific EC surface glycoproteins. Therefore, it is plausible that Gal-1, -3 and -8, acting in a concerted manner, could be mediating the transdifferentiation of macrophages into plump ECs and facilitating their migration and incorporation into the new vessels. LAY SUMMARY In this study, immunostaining of pyogenic granuloma (PG) tissue sections showed immunoreactivity for PECAM-1/CD31, IL-1β, VEGF-C, VEGFR-2 and VEGFR-3, and galectin-1, -3 and -8 in enlarged venules with plump endothelial cells (EC), as well as in some macrophages and other immune cells. Interestingly, enlarged and thin-walled transient vessels lined by PECAM-1/CD31 and VEGFR-2 immunopositive ECs that form from pre-existing normal venules in response to VEGF-A (called 'mother' vessels [MV]) and that undergo intraluminal bridging evolving into various types of capillaries (called 'daughter' vessels [DV]) have been observed in benign and malignant tumours, in physiological and pathological angiogenesis as well as in vascular malformations, suggesting an important role for VEGF-A and VEGFR-2 in such a process. However, it is not only the mechanisms by which the MVs evolve in different types of DVs that remains to be elucidated, but also whether the cells that form intraluminal bridges proceed from locally activated ECs or whether they are derived from bone marrow precursors or from resident macrophages.Given that the formation of homodimers by Gal-1 and Gal-8 and pentamers by Gal-3 to generate gal-glycan lattices at the cell surface and in the extracellular space has been shown, it is possible that in PG tissue Gal-1, -3 and -8, through their binding partners, form a supramolecular structure at the surface of ECs and plump ECs, macrophages and in the extracellular space that might be mediating the transdifferentiation of macrophages into plump ECs and facilitating the migration and incorporation of these cells into the pre-existing venules, thus contributing to the formation of MVs and DVs.
Collapse
Affiliation(s)
- Enrique Arciniegas
- Institute of Biomedicine, Central University of Venezuela, Caracas, Venezuela
| | - Luz Marina Carrillo
- Institute of Biomedicine, Central University of Venezuela, Caracas, Venezuela
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Héctor Rojas
- Institute of Immunology, Central University of Venezuela, Caracas, Venezuela
| | - Jacinto Pineda
- Institute of Anatomy and Pathology, Central University of Venezuela, Caracas, Venezuela
| | - Richard Ramírez
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Oscar Reyes
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Marina Chopite
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Albani Rocheta
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| |
Collapse
|
12
|
Tacconi C, He Y, Ducoli L, Detmar M. Epigenetic regulation of the lineage specificity of primary human dermal lymphatic and blood vascular endothelial cells. Angiogenesis 2020; 24:67-82. [PMID: 32918672 PMCID: PMC7921079 DOI: 10.1007/s10456-020-09743-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
Abstract
Lymphatic and blood vascular endothelial cells (ECs) share several molecular and developmental features. However, these two cell types possess distinct phenotypic signatures, reflecting their different biological functions. Despite significant advances in elucidating how the specification of lymphatic and blood vascular ECs is regulated at the transcriptional level during development, the key molecular mechanisms governing their lineage identity under physiological or pathological conditions remain poorly understood. To explore the epigenomic signatures in the maintenance of EC lineage specificity, we compared the transcriptomic landscapes, histone composition (H3K4me3 and H3K27me3) and DNA methylomes of cultured matched human primary dermal lymphatic and blood vascular ECs. Our findings reveal that blood vascular lineage genes manifest a more ‘repressed’ histone composition in lymphatic ECs, whereas DNA methylation at promoters is less linked to the differential transcriptomes of lymphatic versus blood vascular ECs. Meta-analyses identified two transcriptional regulators, BCL6 and MEF2C, which potentially govern endothelial lineage specificity. Notably, the blood vascular endothelial lineage markers CD34, ESAM and FLT1 and the lymphatic endothelial lineage markers PROX1, PDPN and FLT4 exhibited highly differential epigenetic profiles and responded in distinct manners to epigenetic drug treatments. The perturbation of histone and DNA methylation selectively promoted the expression of blood vascular endothelial markers in lymphatic endothelial cells, but not vice versa. Overall, our study reveals that the fine regulation of lymphatic and blood vascular endothelial transcriptomes is maintained via several epigenetic mechanisms, which are crucial to the maintenance of endothelial cell identity.
Collapse
Affiliation(s)
- Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland
| | - Yuliang He
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland.
| |
Collapse
|
13
|
Pawlak JB, Bálint L, Lim L, Ma W, Davis RB, Benyó Z, Soares MJ, Oliver G, Kahn ML, Jakus Z, Caron KM. Lymphatic mimicry in maternal endothelial cells promotes placental spiral artery remodeling. J Clin Invest 2020; 129:4912-4921. [PMID: 31415243 DOI: 10.1172/jci120446] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/08/2019] [Indexed: 12/27/2022] Open
Abstract
Molecular heterogeneity of endothelial cells underlies their highly specialized functions during changing physiological conditions within diverse vascular beds. For example, placental spiral arteries (SAs) undergo remarkable remodeling to meet the ever-growing demands of the fetus - a process which is deficient in preeclampsia. The extent to which maternal endothelial cells coordinate with immune cells and pregnancy hormones to promote SA remodeling remains largely unknown. Here we found that remodeled SAs expressed the lymphatic markers PROX1, LYVE1, and VEGFR3, mimicking lymphatic identity. Uterine natural killer (uNK) cells, which are required for SA remodeling and secrete VEGFC, were both sufficient and necessary for VEGFR3 activation in vitro and in mice lacking uNK cells, respectively. Using Flt4Chy/+ mice with kinase inactive VEGFR3 and Vegfcfl/fl Vav1-Cre mice, we demonstrated that SA remodeling required VEGFR3 signaling, and that disrupted maternal VEGFR3 signaling contributed to late-gestation fetal growth restriction. Collectively, we identified a novel instance of lymphatic mimicry by which maternal endothelial cells promote SA remodeling, furthering our understanding of the vascular heterogeneity employed for the mitigation of pregnancy complications such as fetal growth restriction and preeclampsia.
Collapse
Affiliation(s)
- John B Pawlak
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - László Bálint
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary.,MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Lillian Lim
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wanshu Ma
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Reema B Davis
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Zoltán Benyó
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Michael J Soares
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Center for Perinatal Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri, USA
| | - Guillermo Oliver
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary.,MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
14
|
Abokyi S, To CH, Lam TT, Tse DY. Central Role of Oxidative Stress in Age-Related Macular Degeneration: Evidence from a Review of the Molecular Mechanisms and Animal Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7901270. [PMID: 32104539 PMCID: PMC7035553 DOI: 10.1155/2020/7901270] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 01/18/2020] [Indexed: 11/17/2022]
Abstract
Age-related macular degeneration (AMD) is a common cause of visual impairment in the elderly. There are very limited therapeutic options for AMD with the predominant therapies targeting vascular endothelial growth factor (VEGF) in the retina of patients afflicted with wet AMD. Hence, it is important to remind readers, especially those interested in AMD, about current studies that may help to develop novel therapies for other stages of AMD. This study, therefore, provides a comprehensive review of studies on human specimens as well as rodent models of the disease, to identify and analyze the molecular mechanisms behind AMD development and progression. The evaluation of this information highlights the central role that oxidative damage in the retina plays in contributing to major pathways, including inflammation and angiogenesis, found in the AMD phenotype. Following on the debate of oxidative stress as the earliest injury in the AMD pathogenesis, we demonstrated how the targeting of oxidative stress-associated pathways, such as autophagy and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, might be the futuristic direction to explore in the search of an effective treatment for AMD, as the dysregulation of these mechanisms is crucial to oxidative injury in the retina. In addition, animal models of AMD have been discussed in great detail, with their strengths and pitfalls included, to assist inform in the selection of suitable models for investigating any of the molecular mechanisms.
Collapse
Affiliation(s)
- Samuel Abokyi
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong
- Department of Optometry, University of Cape Coast, Ghana
| | - Chi-Ho To
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong
| | - Tim T. Lam
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong
| | - Dennis Y. Tse
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong
| |
Collapse
|
15
|
ArabSheibani M, Seifi S, Salehinejad J, Bijani A. Expression of CD34, VEGFR3 and eosinophil density in selected odontogenic tumors- a pilot study. J Oral Biol Craniofac Res 2019; 10:367-371. [PMID: 31687323 DOI: 10.1016/j.jobcr.2019.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 05/11/2019] [Accepted: 09/13/2019] [Indexed: 01/04/2023] Open
Abstract
Background Limited statistically and clinically significant studies have been down on connective tissue factors in the odontogenic tumors. Therefore, the aim of this study was to determine the biological behavior of two selected epithelial odontogenic tumors (Ameloblastoma and Adenomatoid odontogenic tumor) by detecting CD34, VEGFR3 and eosinophil densities. Methods In this cross-sectional study, paraffin blocks including 20 cases of ameloblastoma and 20 (AOT), were selected. Totally, 4 sections were prepared for hematoxylin-eosin, Congo red staining, immunohistochemistry with CD34 and VEGFR3. Expression of VEGFR3, CD34 and lymphatic, blood vessels and eosinophil densities was examined. Results The mean of blood, lymphatic vessels and eosinophils densities in ameloblastoma were 14.9 ± 6.4, 4.4 ± 2 and 3.2 ± 2.7, respectively; and in AOT, they were 8.9 ± 3.4, 3.6 ± 1.3 and 1.2 ± 07, respectively. There was a significant difference in eosinophils and blood vessels densities between the two lesions (p = 0.005; p = 0.003). By increasing the density of eosinophils, the density of the blood vessels increased in both lesions (r = 0.539, P = 0.001) There was no positive relationship between eosinophils and lymphatic vessels densities in the two above mentioned odontogenic lesions (p = 0.288, R = 0.191). Conclusion It can be suggested that tumor angiogenesis and eosinophil densities may play a more effective role than lymphangiogenesis in local invasive behavior of ameloblastoma rather than AOT.
Collapse
Affiliation(s)
- MohammadSina ArabSheibani
- Resident of Oral and Maxillofacial Pathology Department, School of Dentistry, Babol University of Medical Sciences, Babol, Iran
| | - Safoura Seifi
- Oral Health Research Center, Health Research Institute,Babol University of Medical Sciences, Babol, Iran
| | - Jahanshah Salehinejad
- Full Professor of Oral and Maxillofacial Pathology Department, School of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Bijani
- Social Determinant of Health Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
16
|
Hayashi KG, Hosoe M, Fujii S, Kanahara H, Sakumoto R. Temporal expression and localization of vascular endothelial growth factor family members in the bovine uterus during peri-implantation period. Theriogenology 2019; 133:56-64. [PMID: 31059929 DOI: 10.1016/j.theriogenology.2019.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/25/2018] [Accepted: 04/15/2019] [Indexed: 11/30/2022]
Abstract
The aim of this study was to determine endometrial mRNA expression patterns and uterine protein localizations of vascular endothelial growth factor (VEGF) ligands (VEGFA, VEGFB, VEGFC, and VEGFD) and their receptors (VEGFR1, soluble VEGFR1 (sVEGFR1), VEGFR2, and VEGFR3) during the peri-implantation period in cows. The number of blood and lymphatic vessels in the bovine uterus was also investigated. Bovine uterine tissues were collected from pregnant animals on days 15, 18, and 27 after artificial insemination and from non-pregnant animals on days 15 and 18 of the estrous cycle (day 0 = day of estrus). The mRNA expression level of VEGFA, VEGFR1, sVEGFR1, and VEGFR3 were higher on day 18 than on day 15 in the non-pregnant group. On day 18, the levels of mRNA expression of these genes were higher in the non-pregnant group than in the pregnant group. VEGFB mRNA expression levels was higher on day 15 than on days 18 and 27 of gestation and was higher in the pregnant group than in the non-pregnant group on day 15. Using immunohistochemistry, VEGF ligands and their receptors were found in luminal epithelium, glandular epithelium, stroma, and blood vessels of the endometrium. In addition, VEGFA, VEGFD, and VEGFR3 were also detected in the uterine myometrium. In the pregnant group, the number of blood vessels in the endometrium increased from day 15 to 18 and was greater than that of the non-pregnant group on day 18. Our results demonstrate that the VEGF family is expressed and regulated in the bovine uterus during the peri-implantation period, which may be associated with uterine functions, including vascular remodeling in maternal recognition of pregnancy and implantation.
Collapse
Affiliation(s)
- Ken-Go Hayashi
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan
| | - Misa Hosoe
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan
| | - Shiori Fujii
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan
| | - Hiroko Kanahara
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan
| | - Ryosuke Sakumoto
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan.
| |
Collapse
|
17
|
Breslin JW, Yang Y, Scallan JP, Sweat RS, Adderley SP, Murfee WL. Lymphatic Vessel Network Structure and Physiology. Compr Physiol 2018; 9:207-299. [PMID: 30549020 PMCID: PMC6459625 DOI: 10.1002/cphy.c180015] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The lymphatic system is comprised of a network of vessels interrelated with lymphoid tissue, which has the holistic function to maintain the local physiologic environment for every cell in all tissues of the body. The lymphatic system maintains extracellular fluid homeostasis favorable for optimal tissue function, removing substances that arise due to metabolism or cell death, and optimizing immunity against bacteria, viruses, parasites, and other antigens. This article provides a comprehensive review of important findings over the past century along with recent advances in the understanding of the anatomy and physiology of lymphatic vessels, including tissue/organ specificity, development, mechanisms of lymph formation and transport, lymphangiogenesis, and the roles of lymphatics in disease. © 2019 American Physiological Society. Compr Physiol 9:207-299, 2019.
Collapse
Affiliation(s)
- Jerome W. Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Richard S. Sweat
- Department of Biomedical Engineering, Tulane University, New Orleans, LA
| | - Shaquria P. Adderley
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - W. Lee Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, FL
| |
Collapse
|
18
|
The role of placental growth factor (PlGF) and its receptor system in retinal vascular diseases. Prog Retin Eye Res 2018; 69:116-136. [PMID: 30385175 DOI: 10.1016/j.preteyeres.2018.10.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family. Upon binding to VEGF- and neuropilin-receptor sub-types, PlGF modulates a range of neural, glial and vascular cell responses that are distinct from VEGF-A. As PlGF expression is selectively associated with pathological angiogenesis and inflammation, its blockade does not affect the healthy vasculature. PlGF actions have been extensively described in tumor biology but more recently there has been accumulating preclinical evidence that indicates that this growth factor could have an important role in retinal diseases. High levels of PlGF have been found in aqueous humor, vitreous and/or retina of patients exhibiting retinopathies, especially those with diabetic retinopathy (DR) and neovascular age-related macular degeneration (nvAMD). Expression of this growth factor seems to correlate closely with many of the key pathogenic features of early and late retinopathy in preclinical models. For example, studies using genetic modification and/or pharmacological treatment to block PlGF in the laser-induced choroidal neovascularization (CNV) model, oxygen-induced retinopathy model, as well as various murine diabetic models, have shown that PlGF deletion or inhibition can reduce neovascularization, retinal leakage, inflammation and gliosis, without affecting vascular development or inducing neuronal degeneration. Moreover, an inhibitory effect of PlGF blockade on retinal scarring in the mouse CNV model has also been recently demonstrated and was found to be unique for PlGF inhibition, as compared to various VEGF inhibition strategies. Together, these preclinical results suggest that anti-PlGF therapy might have advantages over anti-VEGF treatment, and that it may have clinical applications as a standalone treatment or in combination with anti-VEGF. Additional clinical studies are clearly needed to further elucidate the role of PlGF and its potential as a therapeutic target in ocular diseases.
Collapse
|
19
|
Dallinga MG, Yetkin-Arik B, Kayser RP, Vogels IMC, Nowak-Sliwinska P, Griffioen AW, van Noorden CJF, Klaassen I, Schlingemann RO. IGF2 and IGF1R identified as novel tip cell genes in primary microvascular endothelial cell monolayers. Angiogenesis 2018; 21:823-836. [PMID: 29951828 PMCID: PMC6208896 DOI: 10.1007/s10456-018-9627-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022]
Abstract
Tip cells, the leading cells of angiogenic sprouts, were identified in cultures of human umbilical vein endothelial cells (HUVECs) by using CD34 as a marker. Here, we show that tip cells are also present in primary human microvascular endothelial cells (hMVECs), a more relevant endothelial cell type for angiogenesis. By means of flow cytometry, immunocytochemistry, and qPCR, it is shown that endothelial cell cultures contain a dynamic population of CD34+ cells with many hallmarks of tip cells, including filopodia-like extensions, elevated mRNA levels of known tip cell genes, and responsiveness to stimulation with VEGF and inhibition by DLL4. Furthermore, we demonstrate that our in vitro tip cell model can be exploited to investigate cellular and molecular mechanisms in tip cells and to discover novel targets for anti-angiogenesis therapy in patients. Small interfering RNA (siRNA) was used to knockdown gene expression of the known tip cell genes angiopoietin 2 (ANGPT2) and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), which resulted in similar effects on tip cells and sprouting as compared to inhibition of tip cells in vivo. Finally, we identified two novel tip cell-specific genes in CD34+ tip cells in vitro: insulin-like growth factor 2 (IGF2) and IGF-1-receptor (IGF1R). Knockdown of these genes resulted in a significant decrease in the fraction of tip cells and in the extent of sprouting in vitro and in vivo. In conclusion, this study shows that by using our in vitro tip cell model, two novel essential tip cells genes are identified.
Collapse
Affiliation(s)
- Marchien G Dallinga
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Richelle P Kayser
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Ilse M C Vogels
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands.
- Ocular Angiogenesis Group, Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 15, Room L3-154, 1105 AZ, Amsterdam, The Netherlands.
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| |
Collapse
|
20
|
Asosingh K, Weiss K, Queisser K, Wanner N, Yin M, Aronica M, Erzurum S. Endothelial cells in the innate response to allergens and initiation of atopic asthma. J Clin Invest 2018; 128:3116-3128. [PMID: 29911993 DOI: 10.1172/jci97720] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 05/01/2018] [Indexed: 01/03/2023] Open
Abstract
Protease-activated receptor 2 (PAR-2), an airway epithelial pattern recognition receptor (PRR), participates in the genesis of house dust mite-induced (HDM-induced) asthma. Here, we hypothesized that lung endothelial cells and proangiogenic hematopoietic progenitor cells (PACs) that express high levels of PAR-2 contribute to the initiation of atopic asthma. HDM extract (HDME) protease allergens were found deep in the airway mucosa and breaching the endothelial barrier. Lung endothelial cells and PACs released the Th2-promoting cytokines IL-1α and GM-CSF in response to HDME, and the endothelium had PAC-derived VEGF-C-dependent blood vessel sprouting. Blockade of the angiogenic response by inhibition of VEGF-C signaling lessened the development of inflammation and airway remodeling in the HDM model. Reconstitution of the bone marrow in WT mice with PAR-2-deficient bone marrow also reduced airway inflammation and remodeling. Adoptive transfer of PACs that had been exposed to HDME induced angiogenesis and Th2 inflammation with remodeling similar to that induced by allergen challenge. Our findings identify that lung endothelium and PACs in the airway sense allergen and elicit an angiogenic response that is central to the innate nonimmune origins of Th2 inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Mei Yin
- Imaging Core, Lerner Research Institute, and
| | - Mark Aronica
- Department of Inflammation and Immunity.,Respiratory Institute, the Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Inflammation and Immunity.,Respiratory Institute, the Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
21
|
Rampogu S, Baek A, Zeb A, Lee KW. Exploration for novel inhibitors showing back-to-front approach against VEGFR-2 kinase domain (4AG8) employing molecular docking mechanism and molecular dynamics simulations. BMC Cancer 2018. [PMID: 29514608 PMCID: PMC5842552 DOI: 10.1186/s12885-018-4050-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Angiogenesis is a process of formation of new blood vessels and is an important criteria demonstrated by cancer cells. Over a period of time, these cancer cells infect the other parts of the healthy body by a process called progression. The objective of the present article is to identify a drug molecule that inhibits angiogenesis and progression. Methods In this pursuit, ligand based pharmacophore virtual screening was employed, generating a pharmacophore model, Hypo1 consisting of four features. Furthermore, this Hypo1 was validated recruiting, Fischer’s randomization, test set method and decoy set method. Later, Hypo1 was allowed to screen databases such as Maybridge, Chembridge, Asinex and NCI and were further filtered by ADMET filters and Lipinski’s Rule of Five. A total of 699 molecules that passed the above criteria, were challenged against 4AG8, an angiogenic drug target employing GOLD v5.2.2. Results The results rendered by molecular docking, DFT and the MD simulations showed only one molecule (Hit) obeyed the back-to-front approach. This molecule displayed a dock score of 89.77, involving the amino acids, Glu885 and Cys919, Asp1046, respectively and additionally formed several important hydrophobic interactions. Furthermore, the identified lead molecule showed interactions with key residues when challenged with CDK2 protein, 1URW. Conclusion The lead candidate showed several interactions with the crucial residues of both the targets. Furthermore, we speculate that the residues Cys919 and Leu83 are important in the development of dual inhibitor. Therefore, the identified lead molecule can act as a potential inhibitor for angiogenesis and progression. Electronic supplementary material The online version of this article (10.1186/s12885-018-4050-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shailima Rampogu
- Division of Applied Life Science (BK21 Plus Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju, 52828, Republic of Korea
| | - Ayoung Baek
- Division of Applied Life Science (BK21 Plus Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju, 52828, Republic of Korea
| | - Amir Zeb
- Division of Applied Life Science (BK21 Plus Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju, 52828, Republic of Korea
| | - Keun Woo Lee
- Division of Applied Life Science (BK21 Plus Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju, 52828, Republic of Korea.
| |
Collapse
|
22
|
Dussik CM, Hockley M, Grozić A, Kaneko I, Zhang L, Sabir MS, Park J, Wang J, Nickerson CA, Yale SH, Rall CJ, Foxx-Orenstein AE, Borror CM, Sandrin TR, Jurutka PW. Gene Expression Profiling and Assessment of Vitamin D and Serotonin Pathway Variations in Patients With Irritable Bowel Syndrome. J Neurogastroenterol Motil 2018; 24:96-106. [PMID: 29291611 PMCID: PMC5753908 DOI: 10.5056/jnm17021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 08/03/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIMS Irritable bowel syndrome (IBS) is a multifaceted disorder that afflicts millions of individuals worldwide. IBS is currently diagnosed based on the presence/duration of symptoms and systematic exclusion of other conditions. A more direct manner to identify IBS is needed to reduce healthcare costs and the time required for accurate diagnosis. The overarching objective of this work is to identify gene expression-based biological signatures and biomarkers of IBS. METHODS Gene transcripts from 24 tissue biopsy samples were hybridized to microarrays for gene expression profiling. A combination of multiple statistical analyses was utilized to narrow the raw microarray data to the top 200 differentially expressed genes between IBS versus control subjects. In addition, quantitative polymerase chain reaction was employed for validation of the DNA microarray data. Gene ontology/pathway enrichment analysis was performed to investigate gene expression patterns in biochemical pathways. Finally, since vitamin D has been shown to modulate serotonin production in some models, the relationship between serum vitamin D and IBS was investigated via 25-hydroxyvitamin D (25[OH]D) chemiluminescence immunoassay. RESULTS A total of 858 genetic features were identified with differential expression levels between IBS and asymptomatic populations. Gene ontology enrichment analysis revealed the serotonergic pathway as most prevalent among the differentially expressed genes. Further analysis via real-time polymerase chain reaction suggested that IBS patient-derived RNA exhibited lower levels of tryptophan hydroxylase-1 expression, the enzyme that catalyzes the rate-limiting step in serotonin biosynthesis. Finally, mean values for 25(OH)D were lower in IBS patients relative to non-IBS controls. CONCLUSIONS Values for serum 25(OH)D concentrations exhibited a trend towards lower vitamin D levels within the IBS cohort. In addition, the expression of select IBS genetic biomarkers, including tryptophan hydroxylase 1, was modulated by vitamin D. Strikingly, the direction of gene regulation elicited by vitamin D in colonic cells is "opposite" to the gene expression profile observed in IBS patients, suggesting that vitamin D may help "reverse" the pathological direction of biomarker gene expression in IBS. Thus, our results intimate that IBS pathogenesis and pathophysiology may involve dysregulated serotonin production and/or vitamin D insufficiency.
Collapse
Affiliation(s)
- Christopher M Dussik
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Maryam Hockley
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Aleksandra Grozić
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Ichiro Kaneko
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ,
USA
| | - Lin Zhang
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Marya S Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Jin Park
- School of Life Sciences, Biodesign Institute, Arizona State University, Tempe, AZ,
USA
| | - Jie Wang
- School of Life Sciences, Biodesign Institute, Arizona State University, Tempe, AZ,
USA
| | - Cheryl A Nickerson
- School of Life Sciences, Biodesign Institute, Arizona State University, Tempe, AZ,
USA
| | - Steven H Yale
- Department of Medicine, North Florida Regional Medical Center, Gainesville, FL,
USA
| | | | - Amy E Foxx-Orenstein
- Department of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Scottsdale, AZ,
USA
| | - Connie M Borror
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Todd R Sandrin
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
| | - Peter W Jurutka
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ,
USA
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ,
USA
| |
Collapse
|
23
|
Lymphangiogenesis is a feature of acute GVHD, and VEGFR-3 inhibition protects against experimental GVHD. Blood 2017; 129:1865-1875. [PMID: 28096093 DOI: 10.1182/blood-2016-08-734210] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/09/2017] [Indexed: 01/01/2023] Open
Abstract
Lymph vessels play a crucial role in immune reactions in health and disease. In oncology the inhibition of lymphangiogenesis is an established therapeutic concept for reducing metastatic spreading of tumor cells. During allogeneic tissue transplantation, the inhibition of lymphangiogenesis has been successfully used to attenuate graft rejection. Despite its critical importance for tumor growth, alloimmune responses, and inflammation, the role of lymphangiogenesis has not been investigated during allogeneic hematopoietic stem cell transplantation (allo-HSCT). We found that acute graft-versus-host disease (aGVHD) is associated with lymphangiogenesis in murine allo-HSCT models as well as in patient intestinal biopsies. Inhibition of aGVHD-associated lymphangiogenesis by monoclonal antibodies against vascular endothelial growth factor receptor 3 (VEGFR-3) ameliorated aGVHD and improved survival in murine models. The administration of anti-VEGFR-3 antibodies did not interfere with hematopoietic engraftment and improved immune reconstitution in allo-HSCT recipients with aGVHD. Anti-VEGFR-3 therapy had no significant impact on growth of malignant lymphoma after allo-HSCT. We conclude that aGVHD is associated with lymphangiogenesis in intestinal lesions and in lymph nodes. Our data show that anti-VEGFR-3 treatment ameliorates lethal aGVHD and identifies the lymphatic vasculature as a novel therapeutic target in the setting of allo-HSCT.
Collapse
|
24
|
Hosono K, Isonaka R, Kawakami T, Narumiya S, Majima M. Signaling of Prostaglandin E Receptors, EP3 and EP4 Facilitates Wound Healing and Lymphangiogenesis with Enhanced Recruitment of M2 Macrophages in Mice. PLoS One 2016; 11:e0162532. [PMID: 27711210 PMCID: PMC5053515 DOI: 10.1371/journal.pone.0162532] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 08/24/2016] [Indexed: 01/09/2023] Open
Abstract
Lymphangiogenesis plays an important role in homeostasis, metabolism, and immunity, and also occurs during wound-healing. Here, we examined the roles of prostaglandin E2 (PGE2) receptor (EP) signaling in enhancement of lymphangiogenesis in wound healing processes. The hole-punch was made in the ears of male C57BL/6 mice using a metal ear punch. Healing process and lymphangiogenesis together with macrophage recruitment were analyzed in EP knockout mice. Lymphangiogenesis was up-regulated in the granulation tissues at the margins of punched-hole wounds in mouse ears, and this increase was accompanied by increased expression levels of COX-2 and microsomal prostaglandin E synthase-1. Administration of celecoxib, a COX-2 inhibitor, suppressed lymphangiogenesis in the granulation tissues and reduced the induction of the pro-lymphangiogenic factors, vascular endothelial growth factor (VEGF) -C and VEGF-D. Topical applications of selective EP receptor agonists enhanced the expressions of lymphatic vessel endothelial hyaluronan receptor-1 and VEGF receptor-3. The wound-healing processes and recruitment of CD11b-positive macrophages, which produced VEGF-C and VEGF-D, were suppressed under COX-2 inhibition. Mice lacking either EP3 or EP4 exhibited reduced wound-healing, lymphangiogenesis and recruitment of M2 macrophages, compared with wild type mice. Proliferation of cultured human lymphatic endothelial cells was not detected under PGE2 stimulation. Lymphangiogenesis and recruitment of M2 macrophages that produced VEGF-C/D were suppressed in mice treated with a COX-2 inhibitor or lacking either EP3 or EP4 during wound healing. COX-2 and EP3/EP4 signaling may be novel targets to control lymphangiogenesis in vivo.
Collapse
MESH Headings
- Animals
- CD11b Antigen/metabolism
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Ear/physiology
- Gene Knockout Techniques
- Lymphangiogenesis/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Prostaglandin-E Synthases/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/deficiency
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/deficiency
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction/drug effects
- Up-Regulation/drug effects
- Vascular Endothelial Growth Factor C/biosynthesis
- Vascular Endothelial Growth Factor D/biosynthesis
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Risa Isonaka
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tadashi Kawakami
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shuh Narumiya
- Center for Innovation in Immunoregulation Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Majima
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
25
|
Zhang P, He D, Chen Z, Pan Q, Du F, Zang X, Wang Y, Tang C, Li H, Lu H, Yao X, Jin J, Ma X. Chemotherapy enhances tumor vascularization via Notch signaling-mediated formation of tumor-derived endothelium in breast cancer. Biochem Pharmacol 2016; 118:18-30. [DOI: 10.1016/j.bcp.2016.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/08/2016] [Indexed: 01/28/2023]
|
26
|
Amosco MD, Villar VAM, Naniong JMA, David-Bustamante LMG, Jose PA, Palmes-Saloma CP. VEGF-A and VEGFR1 SNPs associate with preeclampsia in a Philippine population. Clin Exp Hypertens 2016; 38:578-585. [PMID: 27668980 DOI: 10.3109/10641963.2016.1174252] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The vascular endothelial growth factor (VEGF) family is important for establishing normal pregnancy, and related single nucleotide polymorphisms (SNPs) are implicated in abnormal placentation and preeclampsia. We evaluated the association between preeclampsia and several VEGF SNPs among Filipinos, an ethnically distinct group with high prevalence of preeclampsia. The genotypes and allelic variants were determined in a case-control study (191 controls and 165 preeclampsia patients) through SNP analysis of VEGF-A (rs2010963, rs3025039) and VEGF-C (rs7664413) and their corresponding receptors VEGFR1 (rs722503, rs12584067, rs7335588) and VEGFR3 (rs307826) from venous blood DNA. VEGF-A rs3025039 C allele has been shown to associate with preeclampsia (odds ratio of 1.648 (1.03-2.62)), while the T allele bestowed an additive effect for the maintenance of normal, uncomplicated pregnancy and against the development of preeclampsia (odds ratio of 0.62 (0.39-0.98)). VEGFR1 rs722503 is associated with preeclampsia occurring at or after the age of 40 years. The results showed that genetic variability of VEGF-A and VEGFR1 are important in the etiology of preeclampsia among Filipinos.
Collapse
Affiliation(s)
- Melissa D Amosco
- a National Institute of Molecular Biology and Biotechnology, National Science Complex, College of Science, University of the Philippines , Diliman , Quezon City , Philippines.,b Department of Obstetrics and Gynecology , Philippine General Hospital - University of the Philippines , Manila , Philippines
| | - Van Anthony M Villar
- c Division of Nephrology, Department of Medicine , University of Maryland School of Medicine , Baltimore , Maryland , USA.,d Division of Renal Diseases and Hypertension, Department of Medicine , The George Washington University School of Medicine and Health Sciences , Washington , DC , USA
| | - Justin Michael A Naniong
- a National Institute of Molecular Biology and Biotechnology, National Science Complex, College of Science, University of the Philippines , Diliman , Quezon City , Philippines
| | - Lara Marie G David-Bustamante
- b Department of Obstetrics and Gynecology , Philippine General Hospital - University of the Philippines , Manila , Philippines
| | - Pedro A Jose
- c Division of Nephrology, Department of Medicine , University of Maryland School of Medicine , Baltimore , Maryland , USA.,d Division of Renal Diseases and Hypertension, Department of Medicine , The George Washington University School of Medicine and Health Sciences , Washington , DC , USA.,e Department of Physiology , University of Maryland School of Medicine , Baltimore , Maryland , USA.,f Department of Physiology , The George Washington University School of Medicine and Health Sciences , Washington , DC , USA
| | - Cynthia P Palmes-Saloma
- a National Institute of Molecular Biology and Biotechnology, National Science Complex, College of Science, University of the Philippines , Diliman , Quezon City , Philippines
| |
Collapse
|
27
|
Akcal A, Sirvan SS, Karsidag S, Görgülü T, Akcal MA, Ozagari A, Tatlidede S. Combination of ischemic preconditioning and postconditioning can minimise skin flap loss: experimental study. J Plast Surg Hand Surg 2016; 50:233-8. [PMID: 27072670 DOI: 10.3109/2000656x.2016.1154468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Ischaemic preconditioning and postconditioning, which consist of one or a series of short ischaemic events. This study aimed to determine the efficiency of post-conditioning a flap in the minimisation of flap loss after a preconditioned skin flap. METHODS The rats were divided into five groups: sham group, control group, pre-con group, post-con group, and pre + post-con group. On postoperative days 3 and 7, the entire flaps along with the margins of necrosis were traced onto transparent sheets. The areas of intact skin and tissue were recorded. RESULTS The flap necrosis area and percentage of necrosis were calculated for each animal. The necrotic area percentage of the control group was found to be significantly higher than those of the other groups on Days 3 and 7 (p = 0.01 and p = 0.03, respectively). The necrotic area percentage of the pre-con group was significantly higher than the pre + post-con group on Day 7 (p = 0.01). VEGFR-3 expression was observed at a rate of more than 50% in the post-con group. The presence of a protective effect in the late period was separately investigated by immunohistochemical staining of VEGFR-3 in the proliferating vessels. The necrotic areas was reduced in the flaps of the pre-con, post-con, and pre + post-con groups and the combined preconditioning and postconditioning group has reduced necrotic area compared to preconditioning of the skin flap. CONCLUSION The protective effect was observed on day 7 for combined ischaemic preconditioning and postconditioning. The presence of a protective effect in the late period was separately investigated by immunohistochemical staining of VEGFR-3 in the proliferating vessels.
Collapse
Affiliation(s)
- Arzu Akcal
- a Medical Faculty, Department of Plastic and Reconstructive Surgery , Akdeniz University , Antalya , Turkey
| | - Selami Serhat Sirvan
- b Department of Plastic and Reconstructive Surgery , Sisli Etfal Research and Training Hospital , Istanbul , Turkey
| | - Semra Karsidag
- b Department of Plastic and Reconstructive Surgery , Sisli Etfal Research and Training Hospital , Istanbul , Turkey
| | - Tahsin Görgülü
- c Medical Faculty, Department of Plastic and Reconstructive Surgery , Bulent Ecevit University , Zonguldak , Turkey
| | - Mehmet Akif Akcal
- d Deparment of Orthopedic and Traumatology , Ataturk State Hospital , Antalya , Turkey
| | - Aysim Ozagari
- e Deparment of Pathology , Sisli Etfal Research and Training Hospital , Istanbul , Turkey
| | - Soner Tatlidede
- b Department of Plastic and Reconstructive Surgery , Sisli Etfal Research and Training Hospital , Istanbul , Turkey
| |
Collapse
|
28
|
Coon BG, Baeyens N, Han J, Budatha M, Ross TD, Fang JS, Yun S, Thomas JL, Schwartz MA. Intramembrane binding of VE-cadherin to VEGFR2 and VEGFR3 assembles the endothelial mechanosensory complex. ACTA ACUST UNITED AC 2015; 208:975-86. [PMID: 25800053 PMCID: PMC4384728 DOI: 10.1083/jcb.201408103] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
VE-cadherin plays a critical role in endothelial shear stress mechanotransduction by interacting with VEGFRs through their transmembrane domains. Endothelial responses to fluid shear stress are essential for vascular development and physiology, and determine the formation of atherosclerotic plaques at regions of disturbed flow. Previous work identified VE-cadherin as an essential component, along with PECAM-1 and VEGFR2, of a complex that mediates flow signaling. However, VE-cadherin’s precise role is poorly understood. We now show that the transmembrane domain of VE-cadherin mediates an essential adapter function by binding directly to the transmembrane domain of VEGFR2, as well as VEGFR3, which we now identify as another component of the junctional mechanosensory complex. VEGFR2 and VEGFR3 signal redundantly downstream of VE-cadherin. Furthermore, VEGFR3 expression is observed in the aortic endothelium, where it contributes to flow responses in vivo. In summary, this study identifies a novel adapter function for VE-cadherin mediated by transmembrane domain association with VEGFRs.
Collapse
Affiliation(s)
- Brian G Coon
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Nicolas Baeyens
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Jinah Han
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Madhusudhan Budatha
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Tyler D Ross
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Jennifer S Fang
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Sanguk Yun
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Jeon-Leon Thomas
- Université Pierre and Marie Curie-Paris 6, 75005 Paris, France Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique U-1127/UMR-7225, 75654 Paris, France Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France Department of Cell Biology, Department of Biomedical Engineering, and Department of Neurology, Yale University, New Haven, CT 06520
| | - Martin A Schwartz
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06510 Department of Cell Biology, Department of Biomedical Engineering, and Department of Neurology, Yale University, New Haven, CT 06520 Department of Cell Biology, Department of Biomedical Engineering, and Department of Neurology, Yale University, New Haven, CT 06520
| |
Collapse
|
29
|
Gerald D, Adini I, Shechter S, Perruzzi C, Varnau J, Hopkins B, Kazerounian S, Kurschat P, Blachon S, Khedkar S, Bagchi M, Sherris D, Prendergast GC, Klagsbrun M, Stuhlmann H, Rigby AC, Nagy JA, Benjamin LE. RhoB controls coordination of adult angiogenesis and lymphangiogenesis following injury by regulating VEZF1-mediated transcription. Nat Commun 2014; 4:2824. [PMID: 24280686 PMCID: PMC3868161 DOI: 10.1038/ncomms3824] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 10/25/2013] [Indexed: 12/30/2022] Open
Abstract
Mechanisms governing the distinct temporal dynamics that characterize post-natal angiogenesis and lymphangiogenesis elicited by cutaneous wounds and inflammation remain unclear. RhoB, a stress-induced small GTPase, modulates cellular responses to growth factors, genotoxic stress and neoplastic transformation. Here we show, using RhoB null mice, that loss of RhoB decreases pathological angiogenesis in the ischaemic retina and reduces angiogenesis in response to cutaneous wounding, but enhances lymphangiogenesis following both dermal wounding and inflammatory challenge. We link these unique and opposing roles of RhoB in blood versus lymphatic vasculatures to the RhoB-mediated differential regulation of sprouting and proliferation in primary human blood versus lymphatic endothelial cells. We demonstrate that nuclear RhoB-GTP controls expression of distinct gene sets in each endothelial lineage by regulating VEZF1-mediated transcription. Finally, we identify a small-molecule inhibitor of VEZF1-DNA interaction that recapitulates RhoB loss in ischaemic retinopathy. Our findings establish the first intra-endothelial molecular pathway governing the phased response of angiogenesis and lymphangiogenesis following injury.
Collapse
Affiliation(s)
- Damien Gerald
- 1] Center for Vascular Biology Research, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA [2] ImClone Systems (a wholly owned subsidiary of Eli Lilly and Company), New York, New York 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sheybani A, Almony A, Blinder KJ, Shah GK. Neovascular age-related macular degeneration and anti-VEGF nonresponders. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.09.67] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Rutkowski JM, Ihm JE, Lee ST, Kilarski WW, Greenwood VI, Pasquier MC, Quazzola A, Trono D, Hubbell JA, Swartz MA. VEGFR-3 neutralization inhibits ovarian lymphangiogenesis, follicle maturation, and murine pregnancy. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1596-1607. [PMID: 24036251 DOI: 10.1016/j.ajpath.2013.07.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 07/15/2013] [Accepted: 07/31/2013] [Indexed: 11/18/2022]
Abstract
Lymphatic vessels surround follicles within the ovary, but their roles in folliculogenesis and pregnancy, as well as the necessity of lymphangiogenesis in follicle maturation and health, are undefined. We used systemic delivery of mF4-31C1, a specific antagonist vascular endothelial growth factor receptor 3 (VEGFR-3) antibody to block lymphangiogenesis in mice. VEGFR-3 neutralization for 2 weeks before mating blocked ovarian lymphangiogenesis at all stages of follicle maturation, most notably around corpora lutea, without significantly affecting follicular blood angiogenesis. The numbers of oocytes ovulated, fertilized, and implanted in the uterus were normal in these mice; however, pregnancies were unsuccessful because of retarded fetal growth and miscarriage. Fewer patent secondary follicles were isolated from treated ovaries, and isolated blastocysts exhibited reduced cell densities. Embryos from VEGFR-3-neutralized dams developed normally when transferred to untreated surrogates. Conversely, normal embryos transferred into mF4-31C1-treated dams led to the same fetal deficiencies observed with in situ gestation. Although no significant changes were measured in uterine blood or lymphatic vascular densities, VEGFR-3 neutralization reduced serum and ovarian estradiol concentrations during gestation. VEGFR-3-mediated lymphangiogenesis thus appears to modulate the folliculogenic microenvironment and may be necessary for maintenance of hormone levels during pregnancy; both of these are novel roles for the lymphatic vasculature.
Collapse
Affiliation(s)
- Joseph M Rutkowski
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Jong Eun Ihm
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Seung Tae Lee
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Witold W Kilarski
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Veronique I Greenwood
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Miriella C Pasquier
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Alexandra Quazzola
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Didier Trono
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Jeffrey A Hubbell
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Melody A Swartz
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
32
|
Choi I, Lee S, Hong YK. The new era of the lymphatic system: no longer secondary to the blood vascular system. Cold Spring Harb Perspect Med 2013; 2:a006445. [PMID: 22474611 DOI: 10.1101/cshperspect.a006445] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The blood and lymphatic systems are the two major circulatory systems in our body. Although the blood system has been studied extensively, the lymphatic system has received much less scientific and medical attention because of its elusive morphology and mysterious pathophysiology. However, a series of landmark discoveries made in the past decade has begun to change the previous misconception of the lymphatic system to be secondary to the more essential blood vascular system. In this article, we review the current understanding of the development and pathology of the lymphatic system. We hope to convince readers that the lymphatic system is no less essential than the blood circulatory system for human health and well-being.
Collapse
Affiliation(s)
- Inho Choi
- Department of Surgery, Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | |
Collapse
|
33
|
Brake M, Lim CS, Shepherd AC, Shalhoub J, Davies AH. Pathogenesis and etiology of recurrent varicose veins. J Vasc Surg 2013; 57:860-8. [DOI: 10.1016/j.jvs.2012.10.102] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 10/12/2012] [Accepted: 10/22/2012] [Indexed: 11/30/2022]
|
34
|
Qi CX, Zhang XD, Yuan J, Yang JZ, Sun Y, Wang T, Ye H, Ling SQ. Relationship between angiogenesis and lymphangiogenesis in recurrent pterygium. Int J Ophthalmol 2013; 5:655-60. [PMID: 23275896 DOI: 10.3980/j.issn.2222-3959.2012.06.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 11/05/2012] [Indexed: 11/02/2022] Open
Abstract
AIM To examine the relationship between angiogenesis and lymphangigenesis in recurrent pterygia. METHODS Tissues from 34 excised recurrent pterygia (including 12 Grade 1, 10 Grade 2, and 12 Grade 3) were involved in the study and tissues from 7 nasal epibulbar conjunctivae segments were used as controls. Sections from each pterygium were immunostained with CD(31) and LYVE-1 monoclonal antibodies to evaluate lymphatic microvessel density (LMVD) and blood microvessel density (BMVD), and the relationship between LMVD and BMVD in the pterygium was examined. RESULTS There was a large number of CD(31) ((+))LYVE-1((-)) blood vessels but only a few CD(31) ((+))LYVE-1((+)) lymphatic vessels in grades 1 and 2 pterygium. However, lymphatic vessels were dramatically increased in grade 3 pterygium. LMVD correlated closely with BMVD in all pterygia, including grades 1, 2 and 3 peterygium patients (all P values <0.01). Although both the density of blood and lymphatic vessels increased in recurrent pterygia, lymphatic vessels developed much faster than blood vessels, especially in grade 3 pterygia. CONCLUSION There is a significant but not parallel relationship between angiogenesis and lymphangiogenesis in recurrent pterygium. The outgrowth of blood and lymphatic vessels provide evidence that immunological mechanism may play a role in the development and recurrence of pterygium.
Collapse
Affiliation(s)
- Chao-Xiu Qi
- Department of Ophthalmology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Siemerink MJ, Klaassen I, Van Noorden CJF, Schlingemann RO. Endothelial tip cells in ocular angiogenesis: potential target for anti-angiogenesis therapy. J Histochem Cytochem 2012; 61:101-15. [PMID: 23092791 PMCID: PMC3636692 DOI: 10.1369/0022155412467635] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Endothelial tip cells are leading cells at the tips of vascular sprouts coordinating multiple processes during angiogenesis. In the developing retina, tip cells play a tightly controlled, timely role in angiogenesis. In contrast, excessive numbers of tip cells are a characteristic of the chaotic pathological blood vessels in proliferative retinopathies. Tip cells control adjacent endothelial cells in a hierarchical manner to form the stalk of the sprouting vessel, using, among others, the VEGF-DLL-Notch signaling pathway, and recruit pericytes. Tip cells are guided toward avascular areas by signals from the local extracellular matrix that are released by cells from the neuroretina such as astrocytes. Recently, tip cells were identified in endothelial cell cultures, enabling identification of novel molecular markers and mechanisms involved in tip cell biology. These mechanisms are relevant for understanding proliferative retinopathies. Agents that primarily target tip cells can block pathological angiogenesis in the retina efficiently and safely without adverse effects. A striking example is platelet-derived growth factor, which was recently shown to be an efficacious additional target in the treatment of retinal neovascularization. Here we discuss these and other tip cell-based strategies with respect to their potential to treat patients with ocular diseases dominated by neovascularization.
Collapse
Affiliation(s)
- Martin J Siemerink
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
36
|
Fukuhara J, Kase S, Ohashi T, Ando R, Dong Z, Noda K, Ohguchi T, Kanda A, Ishida S. Expression of vascular endothelial growth factor C in human pterygium. Histochem Cell Biol 2012; 139:381-9. [PMID: 22910845 DOI: 10.1007/s00418-012-1019-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2012] [Indexed: 01/06/2023]
Abstract
Vascular endothelial growth factor C (VEGF-C) and its receptor VEGFR-3 mediate lymphangiogenesis. In this study, we analyzed the expression of VEGF-C and VEGFR-3 as well as lymphatic vessels in the pterygium and normal conjunctiva of humans. Fifteen primary nasal pterygia and three normal bulbar conjunctivas, surgically removed, were examined in this study. The lymphatic vessel density (LVD) and blood vessel density were determined by the immunolabeling of D2-40 and CD31, markers for lymphatic and blood vessels, respectively. VEGF-C and VEGFR-3 expression in pterygial and conjunctival tissue proteins was detected by Western blotting and were evaluated using immunohistochemistry. The LVD was significantly higher in the pterygium than normal conjunctiva (p < 0.05). Western blot demonstrated high-level expression of VEGF-C and VEGFR-3 in the pterygium compared with normal conjunctiva. VEGF-C immunoreactivity was detected in the cytoplasm of pterygial and normal conjunctival epithelial cells. The number of VEGF-C-immunopositive cells in pterygial epithelial cells was significantly higher than in normal conjunctival cells (p < 0.05). VEGFR-3 immunoreactivity was localized in the D2-40-positive lymphatic endothelial cells. The present findings suggest the potential role of VEGF-C in the pathogenesis and development of a pterygium through lymphangiogenesis and the VEGF-C/VEGFR-3 pathway as a novel therapeutic target for the human pterygium.
Collapse
Affiliation(s)
- Junichi Fukuhara
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Hokkaido University Graduate School of Medicine, Nishi 7, Kita 15, Sapporo 060-8638, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gu JW, Rizzo P, Pannuti A, Golde T, Osborne B, Miele L. Notch signals in the endothelium and cancer "stem-like" cells: opportunities for cancer therapy. Vasc Cell 2012; 4:7. [PMID: 22487493 PMCID: PMC3348040 DOI: 10.1186/2045-824x-4-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 04/09/2012] [Indexed: 12/14/2022] Open
Abstract
Anti-angiogenesis agents and the identification of cancer stem-like cells (CSC) are opening new avenues for targeted cancer therapy. Recent evidence indicates that angiogenesis regulatory pathways and developmental pathways that control CSC fate are intimately connected, and that endothelial cells are a key component of the CSC niche. Numerous anti-angiogenic therapies developed so far target the VEGF pathway. However, VEGF-targeted therapy is hindered by clinical resistance and side effects, and new approaches are needed. One such approach may be direct targeting of tumor endothelial cell fate determination. Interfering with tumor endothelial cells growth and survival could inhibit not only angiogenesis but also the self-replication of CSC, which relies on signals from surrounding endothelial cells in the tumor microenvironment. The Notch pathway is central to controlling cell fate both during angiogenesis and in CSC from several tumors. A number of investigational Notch inhibitors are being developed. Understanding how Notch interacts with other factors that control endothelial cell functions and angiogenesis in cancers could pave the way to innovative therapeutic strategies that simultaneously target angiogenesis and CSC.
Collapse
Affiliation(s)
- Jian-Wei Gu
- University of Mississippi Cancer Institute, Jackson, MS, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Scala G, Maruccio L. Angiogenesis of buffalo choroid plexuses: structural and immunocytochemical study. Microsc Res Tech 2012; 75:1104-12. [PMID: 22434550 DOI: 10.1002/jemt.22037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/14/2012] [Indexed: 01/05/2023]
Abstract
Mammalian choroid plexuses (CPs) are vascularized structures involved in numerous exchange processes that supply nutrients and hormones to the brain, and that remove deleterious compounds and metabolites from the brain. Studies in the adult Mediterranean buffalo have investigated the morphology of CPs using histochemical and immunohistochemical techniques. To date, however, there have been no studies conducted on ruminants regarding this removal process which serves to repair functional vascular damage in the CPs. Each of these vascular repair processes is a very complex and none of these has not yet been completely understood. Then, the aim of the present study is to investigate the morphological processes during angiogenesis in the CPs of healthy adult buffaloes, utilizing transmission electron microscopy (TEM), scanning electron microscopy (SEM), and immunogold-labeling SEM analysis (biomarkers: angiopoietin-2 [Ang-2], vascular endothelial growth factor receptor-3 [VEGFR-3], and CD133). At TEM, the inner surface of the blood capillaries sometimes showed pillar-like cells, which in contact with endothelial cells formed prominences, which in turn formed neo-blood capillaries. With immunogold-labeling SEM analysis, the CP blood capillaries showed Ang-2 and VEGF-3, respectively, in positive particles and spheroid formations. In addition, the external surface of the blood capillaries showed spheroid formations that originated from the neo-vascular capillaries whose terminals formed a capillary network, positive to CD133. On the basis of these results, the following hypothesis can be made, namely, that these CPs are vascular structures which play a fundamental role in maintaining brain homeostasis and self-repairing of functional vascular damage, independently of the presence of rete mirabile in this species.
Collapse
Affiliation(s)
- Gaetano Scala
- Department of Biological Structures, Functions and Technologies, University of Naples Federico II, Naples, Italy.
| | | |
Collapse
|
39
|
CD34 marks angiogenic tip cells in human vascular endothelial cell cultures. Angiogenesis 2012; 15:151-63. [PMID: 22249946 PMCID: PMC3274677 DOI: 10.1007/s10456-011-9251-z] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 12/20/2011] [Indexed: 12/21/2022]
Abstract
The functional shift of quiescent endothelial cells into tip cells that migrate and stalk cells that proliferate is a key event during sprouting angiogenesis. We previously showed that the sialomucin CD34 is expressed in a small subset of cultured endothelial cells and that these cells extend filopodia: a hallmark of tip cells in vivo. In the present study, we characterized endothelial cells expressing CD34 in endothelial monolayers in vitro. We found that CD34-positive human umbilical vein endothelial cells show low proliferation activity and increased mRNA expression of all known tip cell markers, as compared to CD34-negative cells. Genome-wide mRNA profiling analysis of CD34-positive endothelial cells demonstrated enrichment for biological functions related to angiogenesis and migration, whereas CD34-negative cells were enriched for functions related to proliferation. In addition, we found an increase or decrease of CD34-positive cells in vitro upon exposure to stimuli that enhance or limit the number of tip cells in vivo, respectively. Our findings suggest cells with virtually all known properties of tip cells are present in vascular endothelial cell cultures and that they can be isolated based on expression of CD34. This novel strategy may open alternative avenues for future studies of molecular processes and functions in tip cells in angiogenesis.
Collapse
|
40
|
Scott CA, Eckstrum KS, Bany BM. Localization of C-Fos-Induced Growth Factor ( Figf) mRNA Expression in the Mouse Uterus during Implantation. REPRODUCTIVE SYSTEM & SEXUAL DISORDERS : CURRENT RESEARCH 2012; Suppl 1:003. [PMID: 26918199 PMCID: PMC4764100 DOI: 10.4172/2161-038x.s1-003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The purpose of this study was to characterize the localization of Figf mRNA in the mouse uterus during embryo implantation. Strong Figf mRNA hybridization signals were seen in the primary decidual zone just after the onset of implantation from Days 4.5–6.5. On Day 7.5, this expression continued around the concept us, but in addition we observed high expression of Figf mRNA in the endothelial cells that line the forming vascular sinusoids in the lateral me some trial decidua. Interestingly, on Days 8.5 this high expression continued in the endothelial cells of sinusoids in the lateral me some trial decidual tissue but not in the decidual cells surrounding the concept us. As implantation and placental development finished, Figf mRNA expression remained in the endothelial cells of the sinusoids and spiral arterioles of the decidua basalis. Interestingly, Flt4 mRNA was localized to the endothelial cells lining the sinusoids that form during implantation. Since the endothelial cells of the me some trial sinusoids exhibit a high level of proliferation, we speculate that FIGF-FLT4 signaling may play a role in their formation and function during implantation. This work will provide a basis for further research on the potential role of FIGF-FLT4 signaling in endometrial angiogenesis during implantation in mice.
Collapse
Affiliation(s)
- Charles A Scott
- Departments of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Kirsten S Eckstrum
- Departments of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Brent M Bany
- Departments of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA; Departments of Obstetrics & Gynecology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| |
Collapse
|
41
|
Sikkema AH, de Bont ESJM, Molema G, Dimberg A, Zwiers PJ, Diks SH, Hoving EW, Kamps WA, Peppelenbosch MP, den Dunnen WFA. Vascular endothelial growth factor receptor 2 (VEGFR-2) signalling activity in paediatric pilocytic astrocytoma is restricted to tumour endothelial cells. Neuropathol Appl Neurobiol 2011; 37:538-48. [PMID: 21208252 DOI: 10.1111/j.1365-2990.2011.01160.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS Tumours depend on angiogenesis for enhanced tumour cell survival and progression. Vascular endothelial growth factor receptor (VEGFR) signalling plays a major part in this process. Previously, we evaluated tyrosine kinase activity in paediatric brain tumour tissue lysates using a peptide microarray containing 144 different tyrosine kinase peptide substrates. When applied to paediatric pilocytic astrocytoma tissue, this analysis revealed extensive phosphorylation of VEGFR-derived peptides. The aim of the current study was to validate this result and determine the presence of VEGFR-2 activity in paediatric pilocytic astrocytoma as the main VEGFR in terms of mitogenic signalling. In addition, the localization of VEGFR1-3 mRNA expression was assessed. METHODS VEGFR-2 phosphorylation was determined by adopting a proximity ligation assay approach. Enrichment of endothelial markers and VEGFRs in tumour endothelium was determined by quantitative polymerase chain reaction (qPCR) analysis of laser-microdissected blood vessels. RESULTS Proximity ligation assays on tumour cryosections showed the presence of phosphorylation of VEGFR-2, which primarily localized to vascular endothelium. qPCR analysis of endothelial markers and VEGFRs showed a 13.6-fold average enrichment of VEGFR-2 expression in the laser-microdissected endothelium compared to whole tumour. Also the expression of VEGFR-1 and -3 was highly enriched in the endothelium fraction with an average fold-enrichment of 16.5 and 50.8 respectively. CONCLUSIONS Phosphorylated VEGFR-2 is detected on endothelial cells in paediatric pilocytic astrocytoma. Furthermore, endothelial cells are the main source of VEGFR1-3 mRNA expression. This suggests a crucial role for VEGF/VEGFR-induced angiogenesis in the progression and maintenance of these tumours.
Collapse
Affiliation(s)
- A H Sikkema
- Paediatric Oncology Division, Beatrix Children's Hospital, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Differential mRNA and tissue expression of lymphangiogenic growth factors (VEGF-C and -D) and their receptor (VEGFR-3) during tail regeneration in a gecko. J Comp Physiol B 2011; 182:109-26. [DOI: 10.1007/s00360-011-0604-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 06/28/2011] [Accepted: 07/04/2011] [Indexed: 10/17/2022]
|
43
|
Wang K, Yang R, Xu J, Zhang Y, Zhu L, Lin J, Huang B. DEVELOPMENT OF AN ALPHASCREEN-BASED HIGH-THROUGHPUT SCREENING ASSAY FOR INHIBITORS OF HUMAN VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR-3. J Immunoassay Immunochem 2011; 32:219-32. [DOI: 10.1080/15321819.2011.559296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
44
|
Park YH, Roh SY, Lee YC. Effect of sorafenib on experimental choroidal neovascularization in the rat. Clin Exp Ophthalmol 2010; 38:718-26. [PMID: 20497436 DOI: 10.1111/j.1442-9071.2010.02328.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE This study was designed to evaluate the effect of sorafenib administration on laser-induced experimental choroidal neovascularization (CNV) in rats. METHODS A total of 36 rats were divided into three groups. Sorafenib, an oral, multitargeted receptor tyrosine kinase inhibitor, was administered at a dose of 10 mg/kg per day (n = 12) or 30 mg/kg per day (n = 12) for 1 day before the laser induction of CNV. Rats continued to receive the drug for 14 days. Fluorescein angiograms were analysed for CNV dye leakage and the thickness of CNV was assessed by histology. The levels of vascular endothelial growth factor, platelet-derived growth factor, monocyte chemoattractant protein-1 and intercellular adhesion molecule-1 mRNA were measured by the use of real-time quantitative reverse-transcription polymerase chain reaction. RESULTS Sorafenib-treated rats had significantly less fluorescence leakage as compared with vehicle-treated rats (P < 0.05), The CNV thickness in sorafenib-treated rats was significantly reduced as compared with vehicle-treated rats in a dose-dependent manner (P = 0.00163 for 10 mg/kg and P < 0.00001 for 30 mg/kg). After sorafenib (30 mg/kg) administration, expression of the vascular endothelial growth factor and intercellular adhesion molecule-1 genes was significantly decreased (P < 0.05 and P = 0.00802). CONCLUSION These results suggest that sorafenib may be potentially beneficial for the treatment of CNV in human; further studies on this subject are warranted.
Collapse
Affiliation(s)
- Young-Hoon Park
- Department of Ophthalmology and Visual Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | |
Collapse
|
45
|
Abstract
Endothelial cells are found in most organs and tissues in our body. Despite their apparent morphological and functional similarities, endothelial cells exhibit remarkable heterogeneity and plasticity. In a strict sense, no two endothelial cells are identical in terms of their biological, immunological, functional, metabolic, morphological, and anatomical aspects. Their heterogeneity and plasticity are now known to be dependent upon and conferred by their microenvironments, arteriovenous-lymphatic cell identity, organ-specific vascular beds, fluid dynamics, vessel sizes, anatomical locations, physiological and pathological states, and more. Although abundant evidence is available to demonstrate endothelial heterogeneity in the blood vascular system, studies of heterogeneity and plasticity of lymphatic endothelial cells are limited because of the short history of lymphatic research. Nonetheless, a growing body of exciting work has begun to discover that lymphatic endothelial cells are as heterogeneous as blood vascular endothelial cells. In this article, we discuss the heterogeneity and plasticity of lymphatic endothelial cells.
Collapse
Affiliation(s)
- Sunju Lee
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
46
|
Ongstad EL, Bouta EM, Roberts JE, Uzarski JS, Gibbs SE, Sabel MS, Cimmino VM, Roberts MA, Goldman J. Lymphangiogenesis-independent resolution of experimental edema. Am J Physiol Heart Circ Physiol 2010; 299:H46-54. [PMID: 20207821 DOI: 10.1152/ajpheart.00008.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor (VEGF)-C is necessary for lymphangiogenesis, and excess VEGF-C has been shown to be ameliorative for edema produced by lymphatic obstruction in experimental models. However, it has recently been shown that edema can resolve in the mouse tail even in the complete absence of capillary lymphangiogenesis when distal lymph fluid crosses the regenerating wound site interstitially. This finding has raised questions about the action of VEGF-C/VEGF receptor (VEGFR) signaling during the resolution of experimental edema. Here, the roles of VEGFR-2 and VEGFR-3 signaling in edema resolution were explored. It was found that edema resolved following neutralization of either VEGFR-2 or VEGFR-3 in the mouse tail skin, which inhibited lymphangiogenesis. Neutralization of either VEGFR-2 or VEGFR-3 reduced angiogenesis at the site of obstruction at day 10 (9.2 +/- 1.2% and 11.5 +/- 1.0% blood capillary coverage, respectively) relative to controls (14.3 +/- 1.5% blood capillary coverage). Combined VEGFR-2/-3 neutralization more strongly inhibited angiogenesis (6.9 +/- 1.5% blood capillary coverage), leading to a reduced wound repair of the lymphatic obstruction and extended edema in the tail skin. In contrast, improved tissue repair of the obstruction site increased edema resolution. Macrophages in the swollen tissue were excluded as contributing factors in the VEGFR-dependent extended edema. These results support a role for VEGFR-2/-3-combined signaling in the resolution of experimental edema that is lymphangiogenesis independent.
Collapse
Affiliation(s)
- Emily L Ongstad
- Biomedical Engineering Department, Michigan Technological University, Houghton, MI 49931, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sorensen EW, Gerber SA, Frelinger JG, Lord EM. IL-12 suppresses vascular endothelial growth factor receptor 3 expression on tumor vessels by two distinct IFN-gamma-dependent mechanisms. THE JOURNAL OF IMMUNOLOGY 2010; 184:1858-66. [PMID: 20061409 DOI: 10.4049/jimmunol.0903210] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-12 has been shown to be effective in enhancing antitumor responses. However, how IL-12 exerts its antiangiogenic effect is largely unknown. In this study, we elucidate this mechanism using B16 transfected to express IL-12 (B16/IL-12), a system that provides constant, local production of IL-12 within the tumor microenvironment. Intratumoral IL-12 resulted in a significant delay in tumor growth and phenotypic changes in the vasculature. Vessels found within B16 tumors are chaotic and poorly formed and express vascular endothelial growth factor receptor 3 (VEGFR3), a growth factor receptor not expressed on normal adult vessels. However, the vessels within B16/IL-12 tumors have a more normal morphology and do not express VEGFR3. We have shown that IFN-gamma is required for IL-12 to suppress the aberrant expression of VEGFR3. Indeed, the presence of intratumoral IL-12 stimulates the immune system resulting in more IFN-gamma-producing tumor-infiltrating lymphocytes per tumor when compared with parental B16 tumors, which may have a marked effect on control of tumor growth. Interestingly, within B16/IL-12 tumors, T cells are necessary to suppress VEGFR3 expression on tumor vessels. Finally, using IFN-gamma receptor knockout mice in a bone marrow chimera system, we show that the IFN-gamma produced within the tumor suppresses VEGFR3 expression in two ways: 1) acting directly on tumor vessel endothelial cells, and 2) acting on the tumor-infiltrating lymphocytes to indirectly alter endothelial cells' VEGFR3 expression. Our data indicate a mechanism in which tumor-infiltrating immune cells regulate tumor vessel phenotype.
Collapse
Affiliation(s)
- Elizabeth W Sorensen
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
48
|
Deng J, Liang H, Sun D, Pan Y, Wang B, Guo Y. Vascular endothelial growth factor-D is correlated with hepatic metastasis from gastric cancer after radical gastrectomy. Surgery 2009; 146:896-905. [PMID: 19744460 DOI: 10.1016/j.surg.2009.04.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 04/30/2009] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vascular endothelial growth factor-D (VEGF-D), the activating ligand for vascular endothelial growth factor receptor-3 (VEGFR-3), has been identified as an important endogenic tumor-associated lymphangiogenic factors which can promote lymphatic metastasis from primary tumor. Lymphatic metastasis, which is the most common metastatic method of gastric cancer (GC), is still deemed a potentially relative indicator of distant metastasis from GC. We hypothesize that VEGF-D is associated with hepatic metastasis (HM) from GC after radical gastrectomy by promoting lymphatic metastasis and/or hematogenous metastasis. METHODS This study included 150 GC patients who underwent radical gastrectomy (75 cases presented with HM and 75 cases presented without any recurrences). VEGF-D, VEGF-C, VEGF-A, VEGFR-2, VEGFR-3, and microvessel density (MVD) were evaluated by immunohistochemistry. Besides, clinicopathologic variables and prognosis of GC patients were analyzed statistically. RESULTS Significant associations were seen between Lauren classification and HM, VEGF-D and HM, VEGFR-3 and HM, MVD and HM, and between nodal status and HM. VEGF-D remained an independent factor for HM from GC after radical gastrectomy. Further significant association was evaluated between high-grade VEGF-D expression and short disease-free survival of HM from GC after radical gastrectomy. CONCLUSION VEGF-D, which can induce lymphangiogenesis specifically in many kinds of malignant diseases, is an important factor for predicting HM from GC following radical gastrectomy.
Collapse
Affiliation(s)
- Jingyu Deng
- Department of Gastric Cancer Surgery, City Key Laboratory of Tianjin Cancer Center, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | |
Collapse
|
49
|
Chung ES, Chauhan SK, Jin Y, Nakao S, Hafezi-Moghadam A, van Rooijen N, Zhang Q, Chen L, Dana R. Contribution of macrophages to angiogenesis induced by vascular endothelial growth factor receptor-3-specific ligands. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1984-92. [PMID: 19808642 DOI: 10.2353/ajpath.2009.080515] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Vascular endothelial growth factor receptor (VEGFR)-2 is a major stimulator of hemangiogenesis (HA), whereas VEGFR-3 stimulates lymphangiogenesis (LA). Contrary to this understanding, we demonstrate that implantation of pellets containing VEGFR-3-specific ligands (VEGF-C156S and recombinant murine VEGF-D) into the corneal stroma induce not only LA but also robust HA characterized by blood vessels that are positive for VEGFR-3 expression. The implantation of pellets containing VEGFR-3-specific ligands also leads to the recruitment of VEGF-A-secreting macrophages. Depletion of these infiltrating macrophages using clodronate-liposome administration shows a significant reduction in HA as well as LA. Blockade of either VEGFR-2 or VEGFR-3 signaling reduces both HA and LA; however, the percent reduction of HA is greater in the VEGFR-2 blockade group. In addition, in the VEGFR-3 blockade group, the percent reduction of HA is significantly greater with VEGFR-3-specific ligands than that by VEGF-A or VEGF-C. Collectively, our data suggest that VEGFR-3-specific signaling can induce new blood vessels, to which macrophages contribute a major role, and signify its potential as an additional therapeutic target to the existing VEGF-A/VEGFR-2 signaling-based antiangiogenesis strategies.
Collapse
Affiliation(s)
- Eui-Sang Chung
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Shimamura K, Nakatani T, Ueda A, Sugama J, Okuwa M. Relationship between lymphangiogenesis and exudates during the wound-healing process of mouse skin full-thickness wound. Wound Repair Regen 2009; 17:598-605. [PMID: 19614925 DOI: 10.1111/j.1524-475x.2009.00512.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We considered the relationship among exudate, wound area, angiogenesis, lymphangiogenesis, and reepithelialization during wound healing. Full-thickness wounds were made on the dorsum of mice. The weight of exudate absorbed into the dressing as well as the wound area was determined daily. Sections of the wounds were stained with anti-LYVE-1 and CD31 antibodies. Indian ink was injected into the wound for observing the movement of the exudate on days 3, 5, and 7 after wounding. New epithelium completely covered the wound on day 11. The quantity of exudate peaked on day 1, and then rapidly decreased until it was undetectable on day 11. Most of the Indian ink injected into the wound was retained within the wound and did not flow into the surrounding tissue. New blood vessels showed a uniform distribution in the granulation tissue on day 5. New lymphatics appeared in the granulation tissue approximately 2 days later than the blood vessels and they were distributed toward the center of the granulation tissue on day 11. Thus, reduction of exudate from the wound appears to be related to blood vessels, not lymphatics. However, increasing lymphatics may play a role in the late phase of the wound-healing process.
Collapse
Affiliation(s)
- Kimi Shimamura
- Department of Clinical Nursing, Graduate Course of Nursing Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, Kanazawa 9200942, Japan
| | | | | | | | | |
Collapse
|