1
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
2
|
Kim I, Seo J, Lee DH, Kim YH, Kim JH, Wie MB, Byun JK, Yun JH. Ulmus davidiana 60% edible ethanolic extract for prevention of pericyte apoptosis in diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1138676. [PMID: 37234799 PMCID: PMC10206296 DOI: 10.3389/fendo.2023.1138676] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic retinopathy (DR) is a disease that causes visual deficiency owing to vascular leakage or abnormal angiogenesis. Pericyte apoptosis is considered one of the main causes of vascular leakage in diabetic retina, but there are few known therapeutic agents that prevent it. Ulmus davidiana is a safe natural product that has been used in traditional medicine and is attracting attention as a potential treatment for various diseases, but its effect on pericyte loss or vascular leakage in DR is not known at all. In the present study, we investigated on the effects of 60% edible ethanolic extract of U. davidiana (U60E) and catechin 7-O-β-D-apiofuranoside (C7A), a compound of U. davidiana, on pericyte survival and endothelial permeability. U60E and C7A prevented pericyte apoptosis by inhibiting the activation of p38 and JNK induced by increased glucose and tumor necrosis factor alpha (TNF-α) levels in diabetic retina. Moreover, U60E and C7A reduced endothelial permeability by preventing pericyte apoptosis in co-cultures of pericytes and endothelial cells. These results suggest that U60E and C7A could be a potential therapeutic agent for reducing vascular leakage by preventing pericyte apoptosis in DR.
Collapse
Affiliation(s)
- Iljin Kim
- Department of Pharmacology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jieun Seo
- Faculty of Engineering, Yokohama National University, Yokohama, Japan
| | - Dong Hyun Lee
- Department of Ophthalmology, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Yo-Han Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun-Hyung Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Myung-Bok Wie
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun-Kyu Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Jang-Hyuk Yun
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| |
Collapse
|
3
|
Kim J, Kim S, Choi WJ. Non-Invasive Monitoring of Cutaneous Wound Healing in Non-Diabetic and Diabetic Model of Adult Zebrafish Using OCT Angiography. Bioengineering (Basel) 2023; 10:bioengineering10050538. [PMID: 37237607 DOI: 10.3390/bioengineering10050538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
A diabetic wound presents a severe risk of infections and other complications because of its slow healing. Evaluating the pathophysiology during wound healing is imperative for wound care, necessitating a proper diabetic wound model and assay for monitoring. The adult zebrafish is a rapid and robust model for studying human cutaneous wound healing because of its fecundity and high similarities to human wound repair. OCTA as an assay can provide three-dimensional (3D) imaging of the tissue structure and vasculature in the epidermis, enabling monitoring of the pathophysiologic alterations in the zebrafish skin wound. We present a longitudinal study for assessing the cutaneous wound healing of the diabetic adult zebrafish model using OCTA, which is of importance for the diabetes research using the alternative animal models. We used non-diabetic (n = 9) and type 1 diabetes mellitus (DM) adult zebrafish models (n = 9). The full-thickness wound was generated on the fish skin, and the wound healing was monitored with OCTA for 15 days. The OCTA results demonstrated significant differences between diabetic and non-diabetic wound healing, involving delayed tissue remodeling and impaired angiogenesis for the diabetic wound, leading to slow wound recovery. The adult zebrafish model and OCTA technique may benefit long-term metabolic disease studies using zebrafish for drug development.
Collapse
Affiliation(s)
- Jaeyoung Kim
- Research Institute for Skin Image, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
- Departments of Cancer Control Research and Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Suhyun Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Zebrafish Translational Medical Research Center, Korea University Ansan Hospital, Ansan 15355, Republic of Korea
| | - Woo June Choi
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
4
|
Pericytes in the tumor microenvironment. Cancer Lett 2023; 556:216074. [PMID: 36682706 DOI: 10.1016/j.canlet.2023.216074] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
Pericytes are a type of mural cell located between the endothelial cells of capillaries and the basement membrane, which function to regulate the capillary vasomotor and maintain normal microcirculation of local tissues and organs and have been identified as a significant component in the tumor microenvironment (TME). Pericytes have various interactions with different components of the TME, such as constituting the pre-metastatic niche, promoting the growth of cancer cells and drug resistance through paracrine activity, and inducing M2 macrophage polarization. While changes in the TME can affect the number, phenotype, and molecular markers of pericytes. For example, pericyte detachment from endothelial cells in the TME facilitates tumor cells in situ to invade the circulating blood and is beneficial to local capillary basement membrane enzymatic hydrolysis and endothelial cell proliferation and budding, which contribute to tumor angiogenesis and metastasis. In this review, we discuss the emerging role of pericytes in the TME, and tumor treatment related to pericytes. This review aimed to provide a more comprehensive understanding of the function of pericytes and the relationship between pericytes and tumors and to provide ideas for the treatment and prevention of malignant tumors.
Collapse
|
5
|
Naranjo O, Torices S, Clifford PR, Daftari MT, Osborne OM, Fattakhov N, Toborek M. Pericyte infection by HIV-1: a fatal attraction. Retrovirology 2022; 19:27. [PMID: 36476484 PMCID: PMC9730689 DOI: 10.1186/s12977-022-00614-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
While HIV-1 is primarily an infection of CD4 + T cells, there is an emerging interest towards understanding how infection of other cell types can contribute to HIV-associated comorbidities. For HIV-1 to cross from the blood stream into tissues, the virus must come in direct contact with the vascular endothelium, including pericytes that envelope vascular endothelial cells. Pericytes are multifunctional cells that have been recognized for their essential role in angiogenesis, vessel maintenance, and blood flow rate. Most importantly, recent evidence has shown that pericytes can be a target of HIV-1 infection and support an active stage of the viral life cycle, with latency also suggested by in vitro data. Pericyte infection by HIV-1 has been confirmed in the postmortem human brains and in lungs from SIV-infected macaques. Moreover, pericyte dysfunction has been implicated in a variety of pathologies ranging from ischemic stroke to diabetes, which are common comorbidities among people with HIV-1. In this review, we discuss the role of pericytes during HIV-1 infection and their contribution to the progression of HIV-associated comorbidities.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Paul R. Clifford
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Manav T. Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Olivia M. Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| |
Collapse
|
6
|
Hashemi G, Dight J, Khosrotehrani K, Sormani L. Melanoma Tumour Vascularization and Tissue-Resident Endothelial Progenitor Cells. Cancers (Basel) 2022; 14:4216. [PMID: 36077754 PMCID: PMC9454996 DOI: 10.3390/cancers14174216] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
The aggressiveness of solid cancers, such as melanoma, relies on their metastatic potential. It has become evident that this key cause of mortality is largely conferred by the tumour-associated stromal cells, especially endothelial cells. In addition to their essential role in the formation of the tumour vasculature, endothelial cells significantly contribute to the establishment of the tumour microenvironment, thus enabling the dissemination of cancer cells. Melanoma tumour vascularization occurs through diverse biological processes. Vasculogenesis is the formation of de novo blood vessels from endothelial progenitor cells (EPCs), and recent research has shown the role of EPCs in melanoma tumour vascularization. A more detailed understanding of the complex role of EPCs and how they contribute to the abnormal vessel structures in tumours is of importance. Moreover, anti-angiogenic drugs have a limited effect on melanoma tumour vascularization, and the role of these drugs on EPCs remains to be clarified. Overall, targeting cancer vasculature remains a challenge, and the role of anti-angiogenic drugs and combination therapies in melanoma, a focus of this review, is an area of extensive exploration.
Collapse
Affiliation(s)
| | | | - Kiarash Khosrotehrani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Laura Sormani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
7
|
Matsuo R, Kishibe M, Horiuchi K, Kano K, Tatsukawa T, Hayasaka T, Kabara M, Iinuma S, Eguchi R, Igawa S, Hasebe N, Ishida-Yamamoto A, Kawabe JI. Ninjurin1 deletion in neuron-glial antigen 2-positive pericytes prevents microvessel maturation and delays wound healing. JID INNOVATIONS 2022; 2:100141. [PMID: 36262667 PMCID: PMC9573932 DOI: 10.1016/j.xjidi.2022.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/20/2022] [Accepted: 05/28/2022] [Indexed: 11/24/2022] Open
Abstract
The formation of mature vasculature through angiogenesis is essential for adequate wound healing, such that blood-borne cells, nutrients, and oxygen can be delivered to the remodeling skin area. Neovessel maturation is highly dependent on the coordinated functions of vascular endothelial cells and perivascular cells, namely pericytes (PCs). However, the underlying mechanism for vascular maturation has not been completely elucidated, and its role in wound healing remains unclear. In this study, we investigated the role of Ninjurin-1 (Ninj1), a new molecule mediating vascular maturation, in wound healing using an inducible PC-specific Ninj1 deletion mouse model. Ninj1 expression increased temporarily in NG2-positive PCs in response to skin injury. When tamoxifen treatment induced a decreased Ninj1 expression in PCs, the neovessels in the regenerating wound margins were structurally and functionally immature, but the total number of microvessels was unaltered. This phenotypic change is associated with a reduction in PC-associated microvessels. Wound healing was significantly delayed in the NG2-specific Ninj1 deletion mouse model. Finally, we showed that Ninj1 is a crucial molecule that mediates vascular maturation in injured skin tissue through the interaction of vascular endothelial cells and PCs, thereby inducing adequate and prompt wound healing.
Collapse
Affiliation(s)
- Risa Matsuo
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Mari Kishibe
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Kiwamu Horiuchi
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kohei Kano
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Takamitsu Tatsukawa
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Department of Vascular Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Taiki Hayasaka
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Maki Kabara
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, Asahikawa, Japan
| | - Shin Iinuma
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Ryoji Eguchi
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Satomi Igawa
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Naoyuki Hasebe
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, Asahikawa, Japan
| | | | - Jun-ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, Asahikawa, Japan
- Correspondence: Jun-ichi Kawabe, Department of Biochemistry, Asahikawa Medical University, Asahikawa, 2-1-1 Midorigaoka-higashi, Asahikawa 078-8510, Japan.
| |
Collapse
|
8
|
Ganier C, Rognoni E, Goss G, Lynch M, Watt FM. Fibroblast Heterogeneity in Healthy and Wounded Skin. Cold Spring Harb Perspect Biol 2022; 14:a041238. [PMID: 35667795 PMCID: PMC9248828 DOI: 10.1101/cshperspect.a041238] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Fibroblasts are the main cell type in the dermis. They are responsible for the synthesis and deposition of structural proteins such as collagen and elastin, which are integrated into the extracellular matrix (ECM). Mouse and human studies using flow cytometry, cell culture, skin reconstitution, and lineage tracing experiments have shown the existence of different subpopulations of fibroblasts, including papillary fibroblasts, reticular fibroblasts, and fibroblasts comprising the dermal papilla at the base of the hair follicle. In recent years, the technological advances in single-cell sequencing have allowed researchers to study the repertoire of cells present in full-thickness skin including the dermis. Multiple groups have confirmed that distinct fibroblast populations can be identified in mouse and human dermis on the basis of differences in the transcriptional profile. Here, we discuss the current state of knowledge regarding dermal fibroblast heterogeneity in healthy mouse and human skin, highlighting the similarities and differences between mouse and human fibroblast subpopulations. We also discuss how fibroblast heterogeneity may provide insights into physiological wound healing and its dysfunction in pathological states such as hypertrophic and keloid scars.
Collapse
Affiliation(s)
- Clarisse Ganier
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Emanuel Rognoni
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Georgina Goss
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Magnus Lynch
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
- St John's Institute of Dermatology, King's College London, London SE1 9RT, United Kingdom
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| |
Collapse
|
9
|
Sequential and Dynamic Variations of IL-6, CD18, ICAM, TNF-α, and Microstructure in the Early Stage of Diabetic Retinopathy. DISEASE MARKERS 2022; 2022:1946104. [PMID: 35126785 PMCID: PMC8813280 DOI: 10.1155/2022/1946104] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022]
Abstract
Objective The purpose of this project is to make sequential and indepth observation of the variations of retinal microvascular, microstructure, and inflammatory mediators at the early stage of diabetic retinopathy (DR) in streptozotocin-induced diabetes mellitus (DM) rats. Methods DM was induced by a single intraperitoneal injection of 60 mg/kg body weight streptozotocin (STZ). The fluorescein fundus angiography, hematoxylin and eosin staining, periodic acid-Schiff staining, fluorescence imaging techniques, quantitative real-time PCR, and vascular endothelial growth factor- (VEGF-) A ELISA were performed on the 8th day, at the 4th week, 6th week, 8th week, and 10th week after DM induction, respectively. Results In this study, we observed not only the decrease of retinal ganglion cells (RGCs) and the increase of endotheliocytes to pericytes (E/P) ratio, acellular capillaries, and type IV collagen-positive strands began to occur on the 8th day after induction but the vascular permeability and new vessel buds began to appear in the diabetes group at the 8th week, while the expression of VEGF-A, VEGF mRNA, IL-6 mRNA, ICAM mRNA, and TNF-α mRNA were significantly higher in the diabetes group compared with the normal group(P < 0.01) on the 8th day after induction and maintained a high expression level throughout the 10-week observation period. However, the expression of CD18 mRNA began to increase significantly at the 4th week after induction and reached a peak at the 6th week. Conclusion Our study indicated the abnormal alterations of microvessels, microstructure, and inflammatory mediators at the early stage of DR, which confirms and supplements the previous research, and also promotes an indepth understanding and exploration of the pathophysiology and underlying pathogenesis of DR.
Collapse
|
10
|
Dissecting the Pathogenesis of Diabetic Retinopathy Based on the Biological ceRNA Network and Genome Variation Disturbance. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:9833142. [PMID: 34707685 PMCID: PMC8545528 DOI: 10.1155/2021/9833142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022]
Abstract
Background Diabetic retinopathy (DR) is the most important manifestation of diabetic microangiopathy. It is essential to explore the gene regulatory relationship and genomic variation disturbance of biological networks in DR progression. Methods In this study, we constructed a comprehensive lncRNA-mRNA ceRNA network of DR procession (CLMN) and explored its topological characteristics. Results Modular and functional analysis indicated that the organization of CLMN performed fundamental and specific functions in diabetes and DR pathology. The differential expression of hub ceRNA nodes and positive correlation reveals the highly connected ceRNA regulation and important roles in the regulating of DR pathology. A large proportion of SNPs in the TFBS, DHS, and enhancer regions of lncRNAs will affect lncRNA transcription and further cause expression variation. Some SNPs were found to disrupt the lncRNA functional elements such as miRNA target binding sites. These results indicate the complex nature of genotypic effects in the disturbing of CLMN and further contribute to gene expression variation and different disease phenotypes. Conclusion The identification of individual genomic variations and analysis of biological network disturbance by these genomic variations will help provide more personalized treatment plans and promote the development of precision medicine for DR.
Collapse
|
11
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
12
|
Sharma R, Kadife E, Myers M, Kannourakis G, Prithviraj P, Ahmed N. Determinants of resistance to VEGF-TKI and immune checkpoint inhibitors in metastatic renal cell carcinoma. J Exp Clin Cancer Res 2021; 40:186. [PMID: 34099013 PMCID: PMC8183071 DOI: 10.1186/s13046-021-01961-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/25/2021] [Indexed: 01/03/2023] Open
Abstract
Vascular endothelial growth factor tyrosine kinase inhibitors (VEGF-TKIs) have been the mainstay of treatment for patients with advanced renal cell carcinoma (RCC). Despite its early promising results in decreasing or delaying the progression of RCC in patients, VEGF-TKIs have provided modest benefits in terms of disease-free progression, as 70% of the patients who initially respond to the treatment later develop drug resistance, with 30% of the patients innately resistant to VEGF-TKIs. In the past decade, several molecular and genetic mechanisms of VEGF-TKI resistance have been reported. One of the mechanisms of VEGF-TKIs is inhibition of the classical angiogenesis pathway. However, recent studies have shown the restoration of an alternative angiogenesis pathway in modulating resistance. Further, in the last 5 years, immune checkpoint inhibitors (ICIs) have revolutionized RCC treatment. Although some patients exhibit potent responses, a non-negligible number of patients are innately resistant or develop resistance within a few months to ICI therapy. Hence, an understanding of the mechanisms of VEGF-TKI and ICI resistance will help in formulating useful knowledge about developing effective treatment strategies for patients with advanced RCC. In this article, we review recent findings on the emerging understanding of RCC pathology, VEGF-TKI and ICI resistance mechanisms, and potential avenues to overcome these resistance mechanisms through rationally designed combination therapies.
Collapse
Affiliation(s)
- Revati Sharma
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia
- Federation University Australia, Ballarat, Victoria, 3350, Australia
| | - Elif Kadife
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia
| | - Mark Myers
- Federation University Australia, Ballarat, Victoria, 3350, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia
- Federation University Australia, Ballarat, Victoria, 3350, Australia
| | | | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia.
- Federation University Australia, Ballarat, Victoria, 3350, Australia.
- The Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia.
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, 3052, Australia.
| |
Collapse
|
13
|
Looprasertkul S, Sereemaspun A, Kitkumthorn N, Sooklert K, Sarachana T, Jindatip D. Gold Nanoparticles Affect Pericyte Biology and Capillary Tube Formation. Pharmaceutics 2021; 13:pharmaceutics13050738. [PMID: 34067883 PMCID: PMC8156556 DOI: 10.3390/pharmaceutics13050738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 11/18/2022] Open
Abstract
Gold nanoparticles (AuNPs) are used for diagnostic and therapeutic purposes, especially antiangiogenesis, which are accomplished via inhibition of endothelial cell proliferation, migration, and tube formation. However, no research has been performed on the effects of AuNPs in pericytes, which play vital roles in endothelial cell functions and capillary tube formation during physiological and pathological processes. Therefore, the effects of AuNPs on the morphology and functions of pericytes need to be elucidated. This study treated human placental pericytes in monoculture with 20 nm AuNPs at a concentration of 30 ppm. Ki-67 and platelet-derived growth factor receptor-β (PDGFR-β) mRNA expression was measured using real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Cell migration was assessed by Transwell migration assay. The fine structures of pericytes were observed by transmission electron microscopy. In addition, 30 ppm AuNP-treated pericytes and intact human umbilical vein endothelial cells were cocultured on Matrigel to form three-dimensional (3D) capillary tubes. The results demonstrated that AuNPs significantly inhibited proliferation, reduced PDGFR-β mRNA expression, and decreased migration in pericytes. Ultrastructural analysis of pericytes revealed AuNPs in late endosomes, autolysosomes, and mitochondria. Remarkably, many mitochondria were swollen or damaged. Additionally, capillary tube formation was reduced. We found that numerous pericytes on 3D capillary tubes were round and did not extend their processes along the tubes, which resulted in more incomplete tube formation in the treatment group compared with the control group. In summary, AuNPs can affect pericyte proliferation, PDGFR-β mRNA expression, migration, morphology, and capillary tube formation. The findings highlight the possible application of AuNPs in pericyte-targeted therapy for antiangiogenesis.
Collapse
Affiliation(s)
- Sasikarn Looprasertkul
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Wangmai, Pathumwan, Bangkok 10330, Thailand; (S.L.); (A.S.); (K.S.)
| | - Amornpun Sereemaspun
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Wangmai, Pathumwan, Bangkok 10330, Thailand; (S.L.); (A.S.); (K.S.)
- Nanomedicine Research Unit, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Payathai Rd., Ratchathewi, Bangkok 10400, Thailand;
| | - Kanidta Sooklert
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Wangmai, Pathumwan, Bangkok 10330, Thailand; (S.L.); (A.S.); (K.S.)
- Nanomedicine Research Unit, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tewarit Sarachana
- Age-Related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Rama 1 Rd., Wangmai, Pathumwan, Bangkok 10330, Thailand;
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Depicha Jindatip
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Wangmai, Pathumwan, Bangkok 10330, Thailand; (S.L.); (A.S.); (K.S.)
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Correspondence: ; Tel.: +66-2-256-4281
| |
Collapse
|
14
|
Iesato A, Nucera C. Tumor Microenvironment-Associated Pericyte Populations May Impact Therapeutic Response in Thyroid Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:253-269. [PMID: 34664244 PMCID: PMC9839315 DOI: 10.1007/978-3-030-73119-9_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Thyroid cancer is the most common endocrine malignancy, and aggressive radioactive iodine refractory thyroid carcinomas still lack an effective treatment. A deeper understanding of tumor heterogeneity and microenvironment will be critical to establishing new therapeutic approaches. One of the important influencing factors of tumor heterogeneity is the diversity of cells in the tumor microenvironment. Among these are pericytes, which play an important role in blood vessel stability and angiogenesis, as well as tumor growth and metastasis. Pericytes also have stem cell-like properties and are a heterogeneous cell population, and their lineage, which has been challenging to define, may impact tumor resistance at different tumor stages. Pericytes are also important stroma cell types in the angiogenic microenvironment which express tyrosine-kinase (TK) pathways (e.g., PDGFR-β). Although TK inhibitors (TKI) and BRAFV600E inhibitors are currently used in the clinic for thyroid cancer, their efficacy is not durable and drug resistance often develops. Characterizing the range of distinct pericyte populations and distinguishing them from other perivascular cell types may enable the identification of their specific functions in the thyroid carcinoma vasculature. This remains an essential step in developing new therapeutic strategies. Also, assessing whether thyroid tumors hold immature and/or mature vasculature with pericyte populations coverage may be key to predicting tumor response to either targeted or anti-angiogenesis therapies. It is also critical to apply different markers in order to identify pericyte populations and characterize their cell lineage. This chapter provides an overview of pericyte ontogenesis and the lineages of diverse cell populations. We also discuss the role(s) and targeting of pericytes in thyroid carcinoma, as well as their potential impact on precision targeted therapies and drug resistance.
Collapse
Affiliation(s)
- Asumi Iesato
- Human Thyroid Cancers Preclinical and Translational Research Program, Division of Experimental Pathology, Cancer Research Institute, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA,Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Carmelo Nucera
- Human Thyroid Cancers Preclinical and Translational Research Program, Division of Experimental Pathology, Cancer Research Institute, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA,Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
15
|
Lee LL, Khakoo AY, Chintalgattu V. Cardiac pericytes function as key vasoactive cells to regulate homeostasis and disease. FEBS Open Bio 2020; 11:207-225. [PMID: 33135334 PMCID: PMC7780101 DOI: 10.1002/2211-5463.13021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/24/2020] [Accepted: 10/30/2020] [Indexed: 01/13/2023] Open
Abstract
Pericytes (PCs)—mural cells that envelop endothelial cells (ECs) of microvessels—regulate tissue‐specific vasculature development as well as maturation and maintenance of endothelial barrier integrity. However, little is known about their tissue‐specific function in the heart. Specifically, the mechanism by which cardiac PCs constrict coronary capillaries remains undetermined. To gain insights into the function of cardiac PCs at the cellular level, we isolated NG2+ PDGFRβ+ CD146+ CD34− CD31− CD45− PCs for detailed characterization. Functionally, we provide evidence that these PCs increased transepithelial electrical resistance and decreased endothelial permeability. We show for the first time that this population of PCs express contractile proteins, are stimulated by adrenergic signaling, and demonstrate stereotypical contraction and relaxation. Furthermore, we also studied for the first time, the PCs in in vitro models of disease. PCs in hypoxia activated the hypoxia‐inducible factor 1 alpha pathway, increased secretion of angiogenic factors, and caused cellular apoptosis. Supraphysiological levels of low‐density lipoprotein decreased PC proliferation and induced lipid droplet accumulation. Elevated glucose levels triggered a proinflammatory response. Taken together, our study characterizes cardiac PCs under in vitro disease conditions and supports the hypothesis that cardiac PCs are key vasoactive cells that can regulate blood flow in the heart.
Collapse
Affiliation(s)
- Linda L Lee
- Department of Cardiometabolic Disorders, Amgen Research and Discovery, Amgen Inc., South San Francisco, CA, USA
| | - Aarif Y Khakoo
- Department of Drug Development, Calico Labs, South San Francisco, CA, USA
| | - Vishnu Chintalgattu
- Department of Cardiometabolic Disorders, Amgen Research and Discovery, Amgen Inc., South San Francisco, CA, USA
| |
Collapse
|
16
|
Iendaltseva O, Orlova VV, Mummery CL, Danen EHJ, Schmidt T. Fibronectin Patches as Anchoring Points for Force Sensing and Transmission in Human Induced Pluripotent Stem Cell-Derived Pericytes. Stem Cell Reports 2020; 14:1107-1122. [PMID: 32470326 PMCID: PMC7355144 DOI: 10.1016/j.stemcr.2020.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/24/2022] Open
Abstract
Pericytes (PCs) have been reported to contribute to the mechanoregulation of the capillary diameter and blood flow in health and disease. How this is realized remains poorly understood. We designed several models representing basement membrane (BM) in between PCs and endothelial cells (ECs). These models captured a unique protein organization with micron-sized FN patches surrounded by laminin (LM) and allowed to obtain quantitative information on PC morphology and contractility. Using human induced pluripotent stem cell-derived PCs, we could address mechanical aspects of mid-capillary PC behavior in vitro. Our results showed that PCs strongly prefer FN patches over LM for adhesion formation, have an optimal stiffness for spreading in the range of EC rigidity, and react in a non-canonical way with increased traction forces and reduced spreading on other stiffness then the optimal. Our approach opens possibilities to further study PC force regulation under well-controlled conditions.
Collapse
Affiliation(s)
- Olga Iendaltseva
- Physics of Life Processes, Leiden Institute of Physics, Leiden University, Einsteinweg 55, Leiden, South Holland 2333 CC, the Netherlands; Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, Leiden, South Holland 2333 CC, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, South Holland, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, South Holland, the Netherlands
| | - Erik H J Danen
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, Leiden, South Holland 2333 CC, the Netherlands.
| | - Thomas Schmidt
- Physics of Life Processes, Leiden Institute of Physics, Leiden University, Einsteinweg 55, Leiden, South Holland 2333 CC, the Netherlands.
| |
Collapse
|
17
|
Wang X, Shi H, Zhou J, Zou Q, Zhang Q, Gou S, Chen P, Mou L, Fan N, Suo Y, Ouyang Z, Lai C, Yan Q, Lai L. Generation of rat blood vasculature and hematopoietic cells in rat-mouse chimeras by blastocyst complementation. J Genet Genomics 2020; 47:249-261. [DOI: 10.1016/j.jgg.2020.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/07/2020] [Accepted: 05/02/2020] [Indexed: 12/13/2022]
|
18
|
Zhang ZS, Zhou HN, He SS, Xue MY, Li T, Liu LM. Research advances in pericyte function and their roles in diseases. Chin J Traumatol 2020; 23:89-95. [PMID: 32192909 PMCID: PMC7156959 DOI: 10.1016/j.cjtee.2020.02.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/19/2019] [Accepted: 01/25/2020] [Indexed: 02/04/2023] Open
Abstract
Pericyte, a kind of pluripotent cell, may regulate the irrigation flow and permeability of microcirculation. Pericytes are similar to the smooth muscle cells, which express several kinds of contractile proteins and have contractility. The dysfunction of pericytes is related to many microvascular diseases, including hypoxia, hypertension, diabetic retinopathy, fibrosis, inflammation, Alzheimer's disease, multiple sclerosis, and tumor formation. For a long time, their existence and function have been neglected. The distribution, structure, biomarker, related signaling pathways as well as the roles of pericytes on vascular diseases will be introduced in this review.
Collapse
|
19
|
Alpha-Smooth Muscle Actin-Positive Perivascular Cells in Diabetic Retina and Choroid. Int J Mol Sci 2020; 21:ijms21062158. [PMID: 32245120 PMCID: PMC7139401 DOI: 10.3390/ijms21062158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
Structural alterations of pericytes in microvessels are important features of diabetic retinopathy. Although capillary pericytes had been known not to have α-smooth muscle actin (αSMA), a recent study revealed that a specific fixation method enabled the visualization of αSMA along retinal capillaries. In this study, we applied snap-fixation in wild type and streptozotocin-induced diabetic mice to evaluate the differences in vascular smooth muscle cells of the retina and the choroid. Mice eyeballs were fixed in ice-cold methanol to prevent the depolymerization of filamentous actin. Snap-fixated retina showed αSMA expression in higher-order branches along the capillaries as well as the arterioles and venules, which were not detected by paraformaldehyde fixation. In contrast, most choriocapillaris, except those close to the arterioles, were not covered with αSMA-positive perivascular mural cells. Large choroidal vessels were covered with more αSMA-positive cells in the snap-fixated eyes. Diabetes induced less coverage of αSMA-positive perivascular mural cells overall, but they reached higher-order branches of the retinal capillaries, which was prominent in the aged mice. More αSMA-positive pericytes were observed in the choroid of diabetic mice, but the αSMA-positive expression reduced with aging. This study suggests the potential role of smooth muscle cells in the pathogenesis of age-related diabetic retinopathy and choroidopathy.
Collapse
|
20
|
Ogata A, Wakamiya T, Nishihara M, Tanaka T, Mizokami T, Masuoka J, Momozaki N, Sakata S, Irie H, Abe T. Association between Pericytes in Intraplaque Neovessels and Magnetic Resonance Angiography Findings. Int J Mol Sci 2020; 21:ijms21061980. [PMID: 32183204 PMCID: PMC7139898 DOI: 10.3390/ijms21061980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 12/30/2022] Open
Abstract
(1) Background: Pericytes are involved in intraplaque neovascularization of advanced and complicated atherosclerotic lesions. However, the role of pericytes in human carotid plaques is unclear. An unstable carotid plaque that shows high-intensity signals on time-of-flight (TOF) magnetic resonance angiography (MRA) is often a cause of ischemic stroke. The aim of the present study is to examine the relationship between the pericytes in intraplaque neovessels and MRA findings. (2) Methods: A total of 46 patients with 49 carotid artery stenoses who underwent carotid endarterectomy at our hospitals were enrolled. The patients with carotid plaques that were histopathologically evaluated were retrospectively analyzed. Intraplaque hemorrhage was evaluated using glycophorin A staining, and intraplaque neovessels were evaluated using CD34 (Cluster of differentiation) stain as an endothelial cell marker or NG2 (Neuron-glial antigen 2) and CD146 stains as pericyte markers. Additionally, the relationships between the TOF-MRA findings and the carotid plaque pathologies were evaluated. (3) Results: Of the 49 stenoses, 28 had high-intensity signals (TOF-HIS group) and 21 had iso-intensity signals (TOF-IIS group) on TOF-MRA. The density of the CD34-positive neovessels was equivalent in both groups. However, the NG2- and CD146-positive neovessels had significantly higher densities in the TOF-HIS group than in the TOF-IIS group. (4) Conclusion: The presence of a high-intensity signal on TOF-MRA in carotid plaques was associated with intraplaque hemorrhage and few pericytes in intraplaque neovessels. These findings may contribute to the development of new therapeutic strategies focusing on pericytes.
Collapse
Affiliation(s)
- Atsushi Ogata
- Department of Neurosurgery, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (T.W.); (T.T.); (T.M.); (J.M.); (T.A.)
- Correspondence:
| | - Tomihiro Wakamiya
- Department of Neurosurgery, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (T.W.); (T.T.); (T.M.); (J.M.); (T.A.)
| | - Masashi Nishihara
- Department of Radiology, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (M.N.); (H.I.)
| | - Tatsuya Tanaka
- Department of Neurosurgery, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (T.W.); (T.T.); (T.M.); (J.M.); (T.A.)
- Department of Neurosurgery, Imari Arita Kyoritsu Hospital, Imari 849-4193, Japan;
| | - Taichiro Mizokami
- Department of Neurosurgery, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (T.W.); (T.T.); (T.M.); (J.M.); (T.A.)
- Department of Neurosurgery, Saga Ken Medical Center Koseikan, Saga 840-8571, Japan;
| | - Jun Masuoka
- Department of Neurosurgery, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (T.W.); (T.T.); (T.M.); (J.M.); (T.A.)
| | - Nobuaki Momozaki
- Department of Neurosurgery, Imari Arita Kyoritsu Hospital, Imari 849-4193, Japan;
| | - Shuji Sakata
- Department of Neurosurgery, Saga Ken Medical Center Koseikan, Saga 840-8571, Japan;
| | - Hiroyuki Irie
- Department of Radiology, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (M.N.); (H.I.)
| | - Tatsuya Abe
- Department of Neurosurgery, Faculty of Medicine, Saga University, Saga 840-8501, Japan; (T.W.); (T.T.); (T.M.); (J.M.); (T.A.)
| |
Collapse
|
21
|
The glucose degradation product methylglyoxal induces immature angiogenesis in patients undergoing peritoneal dialysis. Biochem Biophys Res Commun 2020; 525:767-772. [PMID: 32147098 DOI: 10.1016/j.bbrc.2020.02.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/07/2020] [Indexed: 12/20/2022]
Abstract
The accumulation of glucose degradation products (GDPs) can lead to tissue damage in patients with diabetes and those undergoing long-term peritoneal dialysis (PD). Angiogenesis is occasionally observed in the peritoneal membrane of patients undergoing PD, where it is associated with failure of ultrafiltration. To investigate the mechanism underlying the influence of angiogenesis on fluid absorption, we evaluated the effects of accumulation of the glucose degradation product methylglyoxal (MGO) on angiogenesis in vitro, and analyzed the association with angiogenesis in the peritoneal membrane. To this end, we measured the levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF)-BB in cultured endothelial and smooth muscle cells after administration of MGO. The expression of PDGF-BB mRNA and protein decreased significantly after exposure to MGO, while the expression of VEGF mRNA increased (both P < 0.01). The expression of PDGF-Rβ mRNA in cultured smooth muscle cells did not change after administration of MGO, although the expression of VEGF mRNA increased (P < 0.01). We also evaluated the associations between the number of capillary vessels, peritoneal function, and the degree of MGO deposition using peritoneum samples collected from patients undergoing PD. The number of immature capillary vessels was significantly associated with peritoneal dysfunction and the degree of MGO accumulation (both P < 0.01). In conclusion, MGO enhances the production of VEGF and suppresses the production of PDGF-BB, potentially leading to disturbance of angiogenesis in the peritoneal membrane. Accumulation of MGO in the peritoneum may cause immature angiogenesis and peritoneal dysfunction.
Collapse
|
22
|
Zuo Y, Wei J, Chen H. Utilizing Human Dermal Fibroblast Heterogeneity in Autologous Dermal Fibroblast Therapy: An Overcomplicated Strategy or a Promising Approach? Anat Rec (Hoboken) 2019; 302:2126-2131. [PMID: 31433908 DOI: 10.1002/ar.24232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/27/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022]
Abstract
Although human dermal fibroblast heterogeneity has been acknowledged for several decades and a large body of in vitro studies has been performed with zonal dermal fibroblast, current autologous dermal fibroblast therapies do not reflect human dermal fibroblast heterogeneity. To determine if the utilization of human dermal fibroblast heterogeneity in autologous dermal fibroblast therapy is more of a translational perspective that may thus be more likely to make it to the clinic, this article critically reviews the previous studies on dermal fibroblast heterogeneity performed to date. We found that in vitro studies of human dermal fibroblast heterogeneity have run nearly parallel to the in vivo study of autologous dermal fibroblast therapy. Although several human to nude mice xenotransplantation experiments have been performed in different layers of human dermal fibroblast, their clinical significance remains to be considered. We conclude that there is still a great gap between basic experiments and the clinical employment of human dermal fibroblast heterogeneity. To overcome this, it is necessary to conduct clinical trials, which might be restricted by ethical issues. Alternatively, it might be easier to conduct in vivo studies in animal models. Based on our previous study of dermal fibroblast heterogeneity in pigs, we propose the use of pigs as a good animal model for dermal fibroblast heterogeneity. Time will show whether the utilization of human dermal fibroblast heterogeneity in autologous dermal fibroblast therapy is an overcomplicated strategy or a promising approach. Anat Rec, 302:2126-2131, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Yanhai Zuo
- Department of Orthopedics, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Jianjun Wei
- Department of Orthopedics, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Chen
- Department of Orthopedics, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Iqbal M, Fan TP, Watson D, Alenezi S, Saleh K, Sahlan M. Preliminary studies: the potential anti-angiogenic activities of two Sulawesi Island (Indonesia) propolis and their chemical characterization. Heliyon 2019; 5:e01978. [PMID: 31372523 PMCID: PMC6656927 DOI: 10.1016/j.heliyon.2019.e01978] [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: 10/11/2018] [Revised: 04/23/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Several studies have previously reported propolis, or its constituents, to inhibit tumour angiogenesis. The anti-angiogenic activity of two Indonesian stingless bee propolis extracts from Sulawesi Island on vascular cells were assessed. Sample D01 was obtained from the outer side of bee hives, while D02 was from the inner side of the same hives. The extracts were profiled by using liquid chromatography coupled to high resolution mass spectrometry. The anti-angiogenic capacity was assessed on HUVECs and placenta-derived pericytes by cell viability, multi-channel wound healing, and CoCl2 based-hypoxia assays. The exact chemical composition has not been confirmed. The most abundant compounds in Indonesian sample D01 seem to be unusual since they do not immediately fall into a clear class. Two of the most abundant compounds have elemental compositions matching actinopyrones. Identification on the basis of elemental composition is not definitive but compounds in D01 are possibly due to unusually modified terpenoids. Sample D02 has abundant compounds which include four related diterpenes with differing degrees of oxygenation and some sesquiterpenes. However, again the profile is unusual. The anti-angiogenic assays demonstrated that D01 elicited a strong cytotoxic effect and a considerable anti-migratory activity on the vascular cells. Although D02 demonstrated a much weaker cytotoxic effect on the cell lines compared to D01, it elicited a substantial protective effect on the pericytes against CoCl2-induced dropout in an experiment to mimic a micro-environment commonly associated with angiogenesis and tumour growth. These results demonstrate modulatory effects of these propolis samples in vascular cells, which requires further investigation.
Collapse
Affiliation(s)
- Muhammad Iqbal
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Tai-Ping Fan
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - David Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Samya Alenezi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Khaled Saleh
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Muhamad Sahlan
- Department of Chemical Engineering, Universitas Indonesia, Depok, Indonesia
| |
Collapse
|
24
|
Tanaka T, Ogata A, Masuoka J, Mizokami T, Wakamiya T, Nakahara Y, Inoue K, Shimokawa S, Yoshioka F, Momozaki N, Sakata S, Abe T. Possible involvement of pericytes in intraplaque hemorrhage of carotid artery stenosis. J Neurosurg 2019; 130:1971-1977. [PMID: 29957113 DOI: 10.3171/2018.1.jns171942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/17/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Intraplaque hemorrhage (IPH) is most often caused by the rupture of neovessels; however, the factors of intraplaque neovessel vulnerability remain unclear. In this study, the authors focused on pericytes and aimed to investigate the relationship between IPH and pericytes. METHODS The authors retrospectively analyzed the medical records of all patients with carotid artery stenoses who had undergone carotid endarterectomy at their hospitals between August 2008 and March 2016. Patients with carotid plaques that could be evaluated histopathologically were eligible for study inclusion. Intraplaque hemorrhage was analyzed using glycophorin A staining, and patients were divided into the following 2 groups based on the extent of granular staining: high IPH (positive staining area > 10%) and low IPH (positive staining area ≤ 10%). In addition, intraplaque neovessels were immunohistochemically evaluated using antibodies to CD34 as an endothelial cell marker or antibodies to NG2 and CD146 as pericyte markers. The relationship between IPH and pathology for intraplaque neovessels was investigated. RESULTS Seventy of 126 consecutive carotid stenoses were excluded due to the lack of a specimen for histopathological evaluation; therefore, 53 patients with 56 carotid artery stenoses were eligible for study inclusion. Among the 56 stenoses, 37 lesions had high IPH and 19 had low IPH. The number of CD34-positive neovessels was equivalent between the two groups. However, the densities of NG2- and CD146-positive neovessels were significantly lower in the high IPH group than in the low IPH group (5.7 ± 0.5 vs. 17.1 ± 2.4, p < 0.0001; 6.6 ± 0.8 vs. 18.4 ± 2.5, p < 0.0001, respectively). CONCLUSIONS Plaques with high IPH are associated with fewer pericytes in the intraplaque neovessels. This finding may help in the development of novel therapeutic strategies targeting pericytes.
Collapse
Affiliation(s)
- Tatsuya Tanaka
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
- 2Department of Neurosurgery, Imari Arita Kyoritsu Hospital, Imari; and
| | - Atsushi Ogata
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Jun Masuoka
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Taichiro Mizokami
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
- 3Department of Neurosurgery, Saga Ken Medical Centre Koseikan, Saga, Japan
| | - Tomihiro Wakamiya
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Yukiko Nakahara
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Kohei Inoue
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Shoko Shimokawa
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Fumitaka Yoshioka
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| | - Nobuaki Momozaki
- 2Department of Neurosurgery, Imari Arita Kyoritsu Hospital, Imari; and
| | - Shuji Sakata
- 3Department of Neurosurgery, Saga Ken Medical Centre Koseikan, Saga, Japan
| | - Tatsuya Abe
- 1Department of Neurosurgery, Faculty of Medicine, Saga University, Saga
| |
Collapse
|
25
|
Role and Molecular Mechanisms of Pericytes in Regulation of Leukocyte Diapedesis in Inflamed Tissues. Mediators Inflamm 2019; 2019:4123605. [PMID: 31205449 PMCID: PMC6530229 DOI: 10.1155/2019/4123605] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/15/2019] [Accepted: 04/18/2019] [Indexed: 12/20/2022] Open
Abstract
Leukocyte recruitment is a hallmark of the inflammatory response. Migrating leukocytes breach the endothelium along with the vascular basement membrane and associated pericytes. While much is known about leukocyte-endothelial cell interactions, the mechanisms and role of pericytes in extravasation are poorly understood and the classical paradigm of leukocyte recruitment in the microvasculature seldom adequately discusses the involvement of pericytes. Emerging evidence shows that pericytes are essential players in the regulation of leukocyte extravasation in addition to their functions in blood vessel formation and blood-brain barrier maintenance. Junctions between venular endothelial cells are closely aligned with extracellular matrix protein low expression regions (LERs) in the basement membrane, which in turn are aligned with gaps between pericytes. This forms preferential paths for leukocyte extravasation. Breaching of the layer formed by pericytes and the basement membrane entails remodelling of LERs, leukocyte-pericyte adhesion, crawling of leukocytes on pericyte processes, and enlargement of gaps between pericytes to form channels for migrating leukocytes. Furthermore, inflamed arteriolar and capillary pericytes induce chemotactic migration of leukocytes that exit postcapillary venules, and through direct pericyte-leukocyte contact, they induce efficient interstitial migration to enhance the immunosurveillance capacity of leukocytes. Given their role as regulators of leukocyte extravasation, proper pericyte function is imperative in inflammatory disease contexts such as diabetic retinopathy and sepsis. This review summarizes research on the molecular mechanisms by which pericytes mediate leukocyte diapedesis in inflamed tissues.
Collapse
|
26
|
Fernández-Hernando C, Suárez Y. MicroRNAs in endothelial cell homeostasis and vascular disease. Curr Opin Hematol 2019; 25:227-236. [PMID: 29547400 DOI: 10.1097/moh.0000000000000424] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Since the first discovery of microRNAs (miRNAs) in 1993, the involvement of miRNAs in different aspects of vascular disease has emerged as an important research field. In this review, we summarize the fundamental roles of miRNAs in controlling endothelial cell functions and their implication with several aspects of vascular dysfunction. RECENT FINDINGS MiRNAs have been found to be critical modulators of endothelial homeostasis. The dysregulation of miRNAs has been linked to endothelial dysfunction and the development and progression of vascular disease which and open new opportunities of using miRNAs as potential therapeutic targets for vascular disease. SUMMARY Further determination of miRNA regulatory circuits and defining miRNAs-specific target genes remains key to future miRNA-based therapeutic applications toward vascular disease prevention. Many new and unanticipated roles of miRNAs in the control of endothelial functions will assist clinicians and researchers in developing potential therapeutic applications.
Collapse
Affiliation(s)
- Carlos Fernández-Hernando
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
27
|
Abstract
Breast cancer is a heterogeneous disease driven not only by evolutionally diverse cancer cell themselves but also by highly dynamic microenvironment. At the center of the tumor microenvironment, tumor vasculature plays multiple roles from supporting tumor growth to providing a route for metastasis to the distant organ sites. Blood vessels in breast cancer present with perfusion defects associated with vessel dilation, tortuosity, and poor perivascular coverage (Li et al., Ultrasound Med 32:1145-1155, 2013; Eberhard et al., Cancer Res 60:1388-1393, 2000; Cooke et al., Cancer Cell 21:66-81, 2012). Such abnormal vascular system is partly due to the morphological and molecular alteration of pericytes that is accompanied by a significant heterogeneity within the populations (Kim et al., JCI Insight 1:e90733, 2016). While pericytes are implicated for their controversial roles in breast cancer metastasis (Cooke et al., Cancer Cell 21:66-81, 2012; Gerhardt and Semb, J Mol Med (Berl) 86:135-144, 2008; Keskin et al., Cell Rep 10:1066-1081, 2015; Meng et al., Future Oncol 11:169-179, 2015; Xian et al., J Clin Invest 116:642-651, 2006), the impact of their heterogeneity on breast cancer progression, metastasis, intratumoral immunity, and response to chemotherapy are largely unknown. Due to the complexity of angiogenic programs of breast cancer, the anti-angiogenic or anti-vascular treatment has been mostly unsuccessful (Tolaney et al., Proc Natl Acad Sci U S A 112:14325-14330, 2015; Mackey et al., Cancer Treat Rev 38:673-688, 2012; Sledge, J Clin Oncol 33:133-135, 2015) and requires much in-depth knowledge on different components of tumor microenvironment and how these stromal cells are interacting and communicating to each other. Therefore, understanding pericyte heterogeneity and their differential functional contribution will shed light on new potential approaches to treat breast cancer.
Collapse
Affiliation(s)
- Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
28
|
Davidoff MS. The Pluripotent Microvascular Pericytes Are the Adult Stem Cells Even in the Testis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:235-267. [PMID: 30937872 DOI: 10.1007/978-3-030-11093-2_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pericytes of the testis are part of the omnipresent population of pericytes in the vertebrate body and are the only true pluripotent adult stem cells able to produce structures typical for the tree primitive germ layers: ectoderm, mesoderm, and endoderm. They originate very early in the embryogenesis from the pluripotent epiblast. The pericytes become disseminated through the whole vertebrate organism by the growing and differentiating blood vessels where they remain in specialized periendothelial vascular niches as resting pluripotent adult stem cells for tissue generation, maintenance, repair, and regeneration. The pericytes are also the ancestors of the perivascular multipotent stromal cells (MSCs). The variable appearance of the pericytes and their progeny reflects the plasticity under the influence of their own epigenetic and the local environmental factors of the host organ. In the testis the pericytes are the ancestors of the neuroendocrine Leydig cells. After activation the pericytes start to proliferate, migrate, and build transit-amplifying cells that transdifferentiate into multipotent stromal cells. These represent progenitors for a number of different cell types in an organ. Finally, it becomes evident that the pericytes are a brilliant achievement of the biological nature aiming to supply every organ with an omnipresent population of pluripotent adult stem cells. Their fascinating features are prerequisites for future therapy concepts supporting cell systems of organs.
Collapse
Affiliation(s)
- Michail S Davidoff
- University Medical Center Hamburg-Eppendorf, Hamburg Museum of Medical History, Hamburg, Germany.
| |
Collapse
|
29
|
Lee S, Ko J, Park D, Lee SR, Chung M, Lee Y, Jeon NL. Microfluidic-based vascularized microphysiological systems. LAB ON A CHIP 2018; 18:2686-2709. [PMID: 30110034 DOI: 10.1039/c8lc00285a] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Microphysiological systems have emerged in the last decade to provide an alternative to in vivo models in basic science and pharmaceutical research. In the field of vascular biology, in particular, there has been a lack of a suitable in vitro model exhibiting a three-dimensional structure and the physiological function of vasculature integrated with organ-on-a-chip models. The rapid development of organ-on-a-chip technology is well positioned to fulfill unmet needs. Recently, functional integration of vasculature with diverse microphysiological systems has been increasing. This recent trend corresponds to emerging research interest in how the vascular system contributes to various physiological and pathological conditions. This innovative platform has undergone significant development, but adoption of this technology by end-users and researchers in biology is still a work in progress. Therefore, it is critical to focus on simplification and standardization to promote the distribution and acceptance of this technology by the end-users. In this review, we will introduce the latest developments in vascularized microphysiological systems and summarize their outlook in basic research and drug screening applications.
Collapse
Affiliation(s)
- Somin Lee
- Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
30
|
Kolinko Y, Kralickova M, Tonar Z. The impact of pericytes on the brain and approaches for their morphological analysis. J Chem Neuroanat 2018; 91:35-45. [DOI: 10.1016/j.jchemneu.2018.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/10/2018] [Accepted: 04/15/2018] [Indexed: 12/15/2022]
|
31
|
Ahn J, Cho CS, Cho SW, Kang JH, Kim SY, Min DH, Song JM, Park TE, Jeon NL. Investigation on vascular cytotoxicity and extravascular transport of cationic polymer nanoparticles using perfusable 3D microvessel model. Acta Biomater 2018; 76:154-163. [PMID: 29807185 DOI: 10.1016/j.actbio.2018.05.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/16/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022]
Abstract
Vascular networks are the first sites exposed to cationic polymer nanoparticles (NPs) administered intravenously, and thus function as a barrier for NPs reaching the target organ. While cationic polymer NPs have been intensively studied as non-viral delivery systems, their biological effects in human microvessels have been poorly investigated due to a lack of appropriate in vitro systems. Here, we employed a three-dimensional microvessel on a chip, which accurately models in vivo conditions. An open and perfused microvessel surrounded by pericytes was shown to reproduce the important features of living vasculature, including barrier function and biomarkers. Using this microvessel chip, we observed contraction of the microvascular lumen induced by perfused polyethylenimine (PEI)/DNA NPs. We demonstrated that the oxidative stress present when microvessels were exposed to PEI NPs led to rearrangement of microtubules resulting in microvessel contraction. Furthermore, the transcytotic behavior of PEI NPs was analyzed in the microvessel by monitoring the escape of PEI NPs from the microvascular lumen into the perivascular region, which was not possible in two-dimensional culture systems. With our new understanding of the different behaviors of cationic polymer NPs depending on their transcytotic route, we suggest that caveolae-mediated transcytosis is a powerful route for efficient extravascular transport. STATEMENT OF SIGNIFICANCE Microvascular networks are not only biological system constituting largest surface area in the body and but also first site exposed to nanoparticle in vivo. While cationic polymer NPs have been intensively studied as non-viral delivery systems, its biological effects in human microvessel have been poorly investigated due to lack of appropriate in vitro systems. Here, we microengineered an open and perfused 3D pericyte incorporated microvessel model which possesses same morphological characteristic of in vivo. Using the microengineered model, this study represents the first report of transcytotic behavior of NPs in 3D microvessel, and its effect on extravasation efficiency. Our study lays the groundwork for the integration of innovative technologies to examine blood vessel-nanoparticle interaction, which a critical but ill-defined phenomenon.
Collapse
Affiliation(s)
- Jungho Ahn
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, South Korea; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA 30332, USA
| | - Chong-Su Cho
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Seong Woo Cho
- Ulsan National Institute of Science and Technology, Ulsan 44914, South Korea
| | - Joo H Kang
- Ulsan National Institute of Science and Technology, Ulsan 44914, South Korea
| | - Sung-Yon Kim
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Joon Myong Song
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Tae-Eun Park
- Ulsan National Institute of Science and Technology, Ulsan 44914, South Korea.
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
32
|
Cathery W, Faulkner A, Maselli D, Madeddu P. Concise Review: The Regenerative Journey of Pericytes Toward Clinical Translation. Stem Cells 2018; 36:1295-1310. [PMID: 29732653 PMCID: PMC6175115 DOI: 10.1002/stem.2846] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/15/2018] [Accepted: 04/19/2018] [Indexed: 12/27/2022]
Abstract
Coronary artery disease (CAD) is the single leading cause of death worldwide. Advances in treatment and management have significantly improved patient outcomes. On the other hand, although mortality rates have decreased, more people are left with sequelae that require additional treatment and hospitalization. Moreover, patients with severe nonrevascularizable CAD remain with only the option of heart transplantation, which is limited by the shortage of suitable donors. In recent years, cell-based regenerative therapy has emerged as a possible alternative treatment, with several regenerative medicinal products already in the clinical phase of development and others emerging as competitive preclinical solutions. Recent evidence indicates that pericytes, the mural cells of blood microvessels, represent a promising therapeutic candidate. Pericytes are abundant in the human body, play an active role in angiogenesis, vessel stabilization and blood flow regulation, and possess the capacity to differentiate into multiple cells of the mesenchymal lineage. Moreover, early studies suggest a robustness to hypoxic insult, making them uniquely equipped to withstand the ischemic microenvironment. This review summarizes the rationale behind pericyte-based cell therapy and the progress that has been made toward its clinical application. We present the different sources of pericytes and the case for harvesting them from tissue leftovers of cardiovascular surgery. We also discuss the healing potential of pericytes in preclinical animal models of myocardial ischemia (MI) and current practices to upgrade the production protocol for translation to the clinic. Standardization of these procedures is of utmost importance, as lack of uniformity in cell manufacturing may influence clinical outcome. Stem Cells 2018;36:1295-1310.
Collapse
Affiliation(s)
- William Cathery
- Experimental Cardiovascular Medicine, University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Ashton Faulkner
- Experimental Cardiovascular Medicine, University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Davide Maselli
- School of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom & IRCCS Multimedica, Milan, Italy
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine, University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, United Kingdom
| |
Collapse
|
33
|
Microvascular Mural Cell Organotypic Heterogeneity and Functional Plasticity. Trends Cell Biol 2018; 28:302-316. [DOI: 10.1016/j.tcb.2017.12.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 01/28/2023]
|
34
|
Leszczynska A, Murphy JM. Vascular Calcification: Is it rather a Stem/Progenitor Cells Driven Phenomenon? Front Bioeng Biotechnol 2018; 6:10. [PMID: 29479528 PMCID: PMC5811524 DOI: 10.3389/fbioe.2018.00010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Vascular calcification (VC) has witnessed a surge of interest. Vasculature is virtually an omnipresent organ and has a notably high capacity for repair throughout embryonic and adult life. Of the vascular diseases, atherosclerosis is a leading cause of morbidity and mortality on account of ectopic cartilage and bone formation. Despite the identification of a number of risk factors, all the current theories explaining pathogenesis of VC in atherosclerosis are far from complete. The most widely accepted response to injury theory and smooth muscle transdifferentiation to explain the VC observed in atherosclerosis is being challenged. Recent focus on circulating and resident progenitor cells in the vasculature and their role in atherogenesis and VC has been the driving force behind this review. This review discusses intrinsic cellular players contributing to fate determination of cells and tissues to form ectopic cartilage and bone formation.
Collapse
Affiliation(s)
- Aleksandra Leszczynska
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - J Mary Murphy
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
35
|
Lynch MD, Watt FM. Fibroblast heterogeneity: implications for human disease. J Clin Invest 2018; 128:26-35. [PMID: 29293096 DOI: 10.1172/jci93555] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fibroblasts synthesize the extracellular matrix of connective tissue and play an essential role in maintaining the structural integrity of most tissues. Researchers have long suspected that fibroblasts exhibit functional specialization according to their organ of origin, body site, and spatial location. In recent years, a number of approaches have revealed the existence of fibroblast subtypes in mice. Here, we discuss fibroblast heterogeneity with a focus on the mammalian dermis, which has proven an accessible and tractable system for the dissection of these relationships. We begin by considering differences in fibroblast identity according to anatomical site of origin. Subsequently, we discuss new results relating to the existence of multiple fibroblast subtypes within the mouse dermis. We consider the developmental origin of fibroblasts and how this influences heterogeneity and lineage restriction. We discuss the mechanisms by which fibroblast heterogeneity arises, including intrinsic specification by transcriptional regulatory networks and epigenetic factors in combination with extrinsic effects of the spatial context within tissue. Finally, we discuss how fibroblast heterogeneity may provide insights into pathological states including wound healing, fibrotic diseases, and aging. Our evolving understanding suggests that ex vivo expansion or in vivo inhibition of specific fibroblast subtypes may have important therapeutic applications.
Collapse
Affiliation(s)
- Magnus D Lynch
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, Great Maze Pond, London, United Kingdom.,St John's Institute of Dermatology, King's College London, London, United Kingdom
| | - Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, Great Maze Pond, London, United Kingdom
| |
Collapse
|
36
|
Menon NV, Tay HM, Wee SN, Li KHH, Hou HW. Micro-engineered perfusable 3D vasculatures for cardiovascular diseases. LAB ON A CHIP 2017; 17:2960-2968. [PMID: 28740980 DOI: 10.1039/c7lc00607a] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Vessel geometries in microengineered in vitro vascular models are important to recapitulate a pathophysiological microenvironment for the study of flow-induced endothelial dysfunction and inflammation in cardiovascular diseases. Herein, we present a simple and novel extracellular matrix (ECM) hydrogel patterning method to create perfusable vascularized microchannels of different geometries based on the concept of capillary burst valve (CBV). No surface modification is necessary and the method is suitable for different ECM types including collagen, matrigel and fibrin. We first created collagen-patterned, endothelialized microchannels to study barrier permeability and neutrophil transendothelial migration, followed by the development of a biomimetic 3D endothelial-smooth muscle cell (EC-SMC) vascular model. We observed a significant decrease in barrier permeability in the co-culture model during inflammation, which indicates the importance of perivascular cells in ECM remodeling. Finally, we engineered collagen-patterned constricted vascular microchannels to mimic stenosis in atherosclerosis. Whole blood was perfused (1-10 dyne cm-2) into the microdevices and distinct platelet and leukocyte adherence patterns were observed due to increased shear stresses at the constriction, and an additional convective flow through the collagen. Taken together, the developed hydrogel patterning technique enables the formation of unique pathophysiological architectures in organ-on-chip microsystems for real-time study of hemodynamics and cellular interactions in cardiovascular diseases.
Collapse
Affiliation(s)
- Nishanth Venugopal Menon
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Block N3, Singapore 639798
| | | | | | | | | |
Collapse
|
37
|
Hosono J, Morikawa S, Ezaki T, Kawamata T, Okada Y. Pericytes promote abnormal tumor angiogenesis in a rat RG2 glioma model. Brain Tumor Pathol 2017. [PMID: 28646266 DOI: 10.1007/s10014-017-0291-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In glioma angiogenesis, tumor vessels cause morphological and functional abnormalities associated with malignancy and tumor progression. We hypothesized that certain structural changes or scantiness of functional pericytes may be involved in the formation of dysfunctional blood vessels in gliomas. In this study, we performed morphological examinations to elucidate the possible involvement of pericytes in brain tumor vessel abnormalities using a rat RG2 glioma model. After implantation of RG2 glioma cells in the syngeneic rat brain, gliomas were formed as early as day 7. In immunohistochemical examinations, desmin-positive pericytes, characterized by morphological abnormalities, were abundantly found on leaky vessels, as assessed by extravasation of lectin and high-molecular-weight dextrans. Interestingly, desmin-positive pericytes seemed to be characteristic of gliomas in rats. These pericytes were also found to express heat-shock protein 47, which plays an important role in the formation of the basement membrane, suggesting that RG2 pericytes promoted angiogenesis by producing basement membrane as a scaffold for newly forming blood vessels and caused functional abnormalities. We concluded that RG2 pericytes may be responsible for abnormal tumor angiogenesis lacking the functional ability to maintain the blood-brain barrier.
Collapse
Affiliation(s)
- Junji Hosono
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan.,Department of Anatomy and Developmental Biology, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yoshikazu Okada
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
38
|
Gonçalves MR, Johnson SP, Ramasawmy R, Lythgoe MF, Pedley RB, Walker-Samuel S. The effect of imatinib therapy on tumour cycling hypoxia, tissue oxygenation and vascular reactivity. Wellcome Open Res 2017. [DOI: 10.12688/wellcomeopenres.11715.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background: Several biomedical imaging techniques have recently been developed to probe hypoxia in tumours, including oxygen-enhanced (OE) and blood oxygen level-dependent (BOLD) magnetic resonance imaging (MRI). These techniques have strong potential for measuring both chronic and transient (cycling) changes in hypoxia, and to assess response to vascular-targeting therapies in the clinic. Methods: In this study, we investigated the use of BOLD and OE-MRI to assess changes in cycling hypoxia, tissue oxygenation and vascular reactivity to hyperoxic gas challenges, in mouse models of colorectal therapy, following treatment with the PDGF-receptor inhibitor, imatinib mesylate (Glivec). Results: Whilst no changes were observed in imaging biomarkers of cycling hypoxia (from BOLD) or chronic hypoxia (from OE-MRI), the BOLD response to carbogen-breathing became significantly more positive in some tumour regions and more negative in other regions, thereby increasing overall heterogeneity. Conclusions: Imatinib did not affect the magnitude of cycling hypoxia or OE-MRI signal, but increased the heterogeneity of the spatial distribution of BOLD MRI changes in response to gas challenges.
Collapse
|
39
|
Zheng Z, Liu Y, Yang Y, Tang J, Cheng B. Topical 1% propranolol cream promotes cutaneous wound healing in spontaneously diabetic mice. Wound Repair Regen 2017; 25:389-397. [PMID: 28494521 DOI: 10.1111/wrr.12546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 05/04/2017] [Indexed: 12/11/2022]
Abstract
Diabetic foot ulcers (DFUs) are a constant threat to diabetic patients and can lead to amputations and even death. Intralesional administration of propranolol in diabetic wounds has not been reported previously. This study aimed to investigate the efficacy of propranolol cream in diabetic wounds. Fifty-six spontaneously diabetic mice were divided into the propranolol group and the control group. After preparing full-thickness wounds on the back of the mice, 1% propranolol cream was topically applied to wounds in the experimental group and 0% propranolol cream in controls. The wound sizes were measured and calculated against the original area. The wounds were analyzed up to 21 days after injury. At all evaluation time-points, the wound size (%) in the propranolol group was significantly smaller than in the controls. Epidermal growth factor (EGF) protein expression increased in the experimental vs. CONTROL GROUP Vascular endothelial growth factor (VEGF) expression was significantly lower in the experimental vs. control group whereas NG2 proteoglycan was increased throughout the study. However, matrix metallopeptidase (MMP)-9 expression was at first significantly higher in the experimental vs. control group then the MMP-9 protein level in the control group increased and surpassed that in the experimental group. In conclusion, intralesional administration of 1% propranolol cream promotes reepithelialization and regulates abnormal angiogenesis in diabetic wounds. Propranolol cream may become a new drug for the treatment of DFUs.
Collapse
Affiliation(s)
- Zhifang Zheng
- The Graduate School of Southern Medical University, Guangzhou, China.,Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yishu Liu
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China.,The Graduate School of Third Military Medical University, Chongqing, China
| | - Yu Yang
- The Graduate School of Southern Medical University, Guangzhou, China.,Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Jianbing Tang
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Biao Cheng
- The Graduate School of Southern Medical University, Guangzhou, China.,Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China.,The Graduate School of Third Military Medical University, Chongqing, China.,Center of Wound Treatment, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China.,The Key Laboratory of Trauma Treatment & Tissue Repair of Tropical Area, PLA, Guangzhou, China
| |
Collapse
|
40
|
Kim E, Na S, An B, Yang SR, Kim WJ, Ha KS, Han ET, Park WS, Lee CM, Lee JY, Lee SJ, Hong SH. Paracrine influence of human perivascular cells on the proliferation of adenocarcinoma alveolar epithelial cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:161-168. [PMID: 28280409 PMCID: PMC5343049 DOI: 10.4196/kjpp.2017.21.2.161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/20/2016] [Accepted: 01/09/2017] [Indexed: 12/16/2022]
Abstract
Understanding the crosstalk mechanisms between perivascular cells (PVCs) and cancer cells might be beneficial in preventing cancer development and metastasis. In this study, we investigated the paracrine influence of PVCs derived from human umbilical cords on the proliferation of lung adenocarcinoma epithelial cells (A549) and erythroleukemia cells (TF-1α and K562) in vitro using Transwell® co-culture systems. PVCs promoted the proliferation of A549 cells without inducing morphological changes, but had no effect on the proliferation of TF-1α and K562 cells. To identify the factors secreted from PVCs, conditioned media harvested from PVC cultures were analyzed by antibody arrays. We identified a set of cytokines, including persephin (PSPN), a neurotrophic factor, and a key regulator of oral squamous cell carcinoma progression. Supplementation with PSPN significantly increased the proliferation of A549 cells. These results suggested that PVCs produced a differential effect on the proliferation of cancer cells in a cell-type dependent manner. Further, secretome analyses of PVCs and the elucidation of the molecular mechanisms could facilitate the discovery of therapeutic target(s) for lung cancer.
Collapse
Affiliation(s)
- Eunbi Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Sunghun Na
- Department of Obstetrics & Gynecology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Borim An
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Department of Medicine, Alpert Medical School, Brown University, Providence, Rhode Island 02912, US
| | - Ji Yoon Lee
- Department of Biomedical Laboratory Science, College of Health Sciences, Sanji University, Wonju 26339, Korea
| | - Seung-Joon Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
41
|
Jaffe GJ, Ciulla TA, Ciardella AP, Devin F, Dugel PU, Eandi CM, Masonson H, Monés J, Pearlman JA, Quaranta-El Maftouhi M, Ricci F, Westby K, Patel SC. Dual Antagonism of PDGF and VEGF in Neovascular Age-Related Macular Degeneration. Ophthalmology 2017; 124:224-234. [DOI: 10.1016/j.ophtha.2016.10.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 10/06/2016] [Accepted: 10/08/2016] [Indexed: 01/20/2023] Open
|
42
|
Pfister F, Hussain H, Belharazem D, Busch S, Simon-Keller K, Becker D, Pfister E, Rieker R, Ströbel P, Marx A. Vascular architecture as a diagnostic marker for differentiation of World Health Organization thymoma subtypes and thymic carcinoma. Histopathology 2017; 70:693-703. [DOI: 10.1111/his.13114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 10/26/2016] [Indexed: 01/27/2023]
Affiliation(s)
- Frederick Pfister
- Department of Nephropathology; Institute of Pathology; Universitätsklinikum Erlangen; Friedrich-Alexander-University Erlangen-Nürnberg; Erlangen Germany
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Hussam Hussain
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Djeda Belharazem
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Svenja Busch
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Katja Simon-Keller
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Dominic Becker
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Eva Pfister
- Department of Nephropathology; Institute of Pathology; Universitätsklinikum Erlangen; Friedrich-Alexander-University Erlangen-Nürnberg; Erlangen Germany
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Ralf Rieker
- Institute of Pathology; Universitätsklinikum Erlangen; Friedrich-Alexander-University Erlangen-Nürnberg; Erlangen Germany
| | - Philipp Ströbel
- Institute of Pathology; University Medicine Göttingen; Göttingen Germany
| | - Alexander Marx
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| |
Collapse
|
43
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
44
|
ÇELEBİ SALTIK B, GÖKÇINAR YAĞCI B. Expansion of human umbilical cord blood hematopoieticprogenitors with cord vein pericytes. Turk J Biol 2017. [DOI: 10.3906/biy-1510-49] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
45
|
Zheng Z, Wan Y, Liu Y, Yang Y, Tang J, Huang W, Cheng B. Sympathetic Denervation Accelerates Wound Contraction but Inhibits Reepithelialization and Pericyte Proliferation in Diabetic Mice. J Diabetes Res 2017; 2017:7614685. [PMID: 29147666 PMCID: PMC5632918 DOI: 10.1155/2017/7614685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/28/2017] [Indexed: 01/13/2023] Open
Abstract
Previous studies focused on the effects of sympathetic denervation with 6-hydroxydopamine (6-OHDA) on nondiabetic wounds, but the effects of 6-OHDA on diabetic wounds have not been previously reported. In this study, treated mice received intraperitoneal 6-OHDA, and control mice received intraperitoneal injections of normal saline. Full-thickness wounds were established on the backs of mice. The wounds were sectioned (four mice per group) for analysis at 2, 5, 7, 10, 14, 17, and 21 days after injury. The wound areas in the control group were larger than those in the treatment group. Histological scores for epidermal and dermal regeneration were reduced in the 6-OHDA-treated group on day 21. The mast cells (MCs) in each field decreased after sympathectomy on days 17 and 21. The expression levels of norepinephrine, epidermal growth factor (EGF), interleukin-1 beta, NG2 proteoglycan, and desmin in the treatment group were less than those in the control group. In conclusion, 6-OHDA delays reepithelialization during wound healing in diabetic mice by decreasing EGF, but increases wound contraction by reducing IL-1β levels and the number of MCs. Besides, 6-OHDA led to reduced pericyte proliferation in diabetic wounds, which might explain the vascular dysfunction after sympathetic nerve loss in diabetic wounds.
Collapse
Affiliation(s)
- Zhifang Zheng
- The Graduate School of Southern Medical University, Guangzhou, China
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
- Department of Anatomy, School of Basic Medicine Sciences, Southern Medical University, Guangzhou, China
| | - Yu Wan
- The Graduate School of Southern Medical University, Guangzhou, China
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yishu Liu
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
- The Graduate School of Third Military Medical University, Chongqing, China
| | - Yu Yang
- The Graduate School of Southern Medical University, Guangzhou, China
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Jianbing Tang
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Wenhua Huang
- The Graduate School of Southern Medical University, Guangzhou, China
- Department of Anatomy, School of Basic Medicine Sciences, Southern Medical University, Guangzhou, China
| | - Biao Cheng
- The Graduate School of Southern Medical University, Guangzhou, China
- Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
- Department of Anatomy, School of Basic Medicine Sciences, Southern Medical University, Guangzhou, China
- The Graduate School of Third Military Medical University, Chongqing, China
- Center of Wound Treatment, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
- The Key Laboratory of Trauma Treatment & Tissue Repair of Tropical Area, PLA, Guangzhou, China
| |
Collapse
|
46
|
Embryonic Stem Cell-like Population in Dupuytren's Disease. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e1064. [PMID: 27975007 PMCID: PMC5142473 DOI: 10.1097/gox.0000000000001064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/10/2016] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Recent research has identified mesenchymal stem cells (MSCs) within Dupuytren’s disease (DD) tissue and they have been proposed to give rise to the myofibroblasts, implicated in the progression of this condition. The aim of this study was to identify and characterize the primitive population that might be upstream of the MSC population, within DD. Methods: Formalin-fixed paraffin-embedded 4-µm-thick sections of DD cords and nodules obtained from 6 patients underwent 3,3-diaminobenzidine and immunofluorescent immunohistochemical staining for embryonic stem cell (ESC) markers OCT4, NANOG, SOX2, pSTAT3, and SALL4 and endothelial markers CD34 and ERG. NanoString gene expression analysis was performed to determine the transcriptional activation of these markers. Results: Immunohistochemical staining demonstrated the expression of ESC markers OCT4, NANOG, SOX2, pSTAT3, and SALL4 on the endothelium of the microvessels expressing CD34 and ERG, particularly those surrounding the DD nodules. NanoString analysis confirmed the transcriptional activation of OCT4, NANOG, STAT3, and SALL4, but not SOX2. Conclusion: This article demonstrates the novel finding of an ESC-like population expressing ESC markers OCT4, NANOG, SOX2, pSTAT3, and SALL4, localized to the endothelium of the microvessels within DD tissue, suggesting a potential therapeutic target for this condition.
Collapse
|
47
|
TGFβ induces BIGH3 expression and human retinal pericyte apoptosis: a novel pathway of diabetic retinopathy. Eye (Lond) 2016; 30:1639-1647. [PMID: 27564721 DOI: 10.1038/eye.2016.179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/14/2016] [Indexed: 02/05/2023] Open
Abstract
PurposeOne of the earliest hallmarks of diabetic retinopathy is the loss of retinal pericytes. However, the mechanisms that promote pericyte dropout are unknown. In the present study, we propose a novel pathway in which pericyte apoptosis is mediated by macrophages, TGFβ and pro-apoptotic BIGH3 (TGFβ-induced Gene Human Clone 3) protein.Patients and methodsTo elucidate this pathway, we assayed human retinal pericyte (HRP) apoptosis by TUNEL assay, BIGH3 mRNA expression by qPCR, and BIGH3 protein expression by western blot analysis. HRP were treated with BIGH3 protein, TGFβ1 and TGFβ2 and inhibition assays were carried out by blocking with antibodies against BIGH3. The distribution of BIGH3 and CD68+ macrophages were compared in a post-mortem donor eye with 7-year history of Type II diabetes and histopathogically confirmed non-proliferative diabetic retinopathy (NPDR).ResultsTGFβ induced a significant increase in BIGH3 mRNA and protein expression, and HRP apoptosis. BIGH3 treatment showed HRP undergo apoptosis in a dose-dependent manner. At 5 μg/ml, BIGH3 induced 3.5-times more apoptosis in HRP than in retinal endothelial cells. TGFβ induced apoptosis was inhibited by blocking with antibodies against BIGH3. In an example of NPDR, BIGH3 accumulated within the walls of the inner retina arterioles. Macrophage infiltrates were frequently associated with these vessels and the inner nuclear layer.ConclusionTogether with our previously published results on macrophage-induced retinal endothelial cell apoptosis, the present study supports a novel inflammatory pathway mediated by macrophages and the BIGH3 protein leading to HRP apoptosis. As shown in human post-mortem globes, these observations are clinically relevant, suggesting a new mechanism underlying pericyte dropout during NPDR.
Collapse
|
48
|
Vezzani B, Pierantozzi E, Sorrentino V. Not All Pericytes Are Born Equal: Pericytes from Human Adult Tissues Present Different Differentiation Properties. Stem Cells Dev 2016; 25:1549-1558. [PMID: 27549576 DOI: 10.1089/scd.2016.0177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pericytes (PCs) have been recognized for a long time only as structural cells of the blood vessels. The identification of tight contacts with endothelial cells and the ability to interact with surrounding cells through paracrine signaling revealed additional functions of PCs in maintaining the homeostasis of the perivascular environment. PCs got the front page, in the late 1990s, after the identification and characterization of a new embryonic cell population, the mesoangioblasts, from which PCs present in the adult organism are thought to derive. From these studies, it was clear that PCs were also endowed with multipotent mesodermal abilities. Furthermore, their ability to cross the vascular wall and to reconstitute skeletal muscle tissue after systemic injection opened the way to a number of studies aimed to develop therapeutic protocols for a cell therapy of muscular dystrophy. This has resulted in a major effort to characterize pericytic cell populations from skeletal muscle and other adult tissues. Additional studies also addressed their relationship with other cells of the perivascular compartment and with mesenchymal stem cells. These data have provided initial evidence that PCs from different adult tissues might be endowed with distinctive differentiation abilities. This would suggest that the multipotent mesenchymal ability of PCs might be restrained within different tissues, likely depending on the specific cell renewal and repair requirements of each tissue. This review presents current knowledge on human PCs and highlights recent data on the differentiation properties of PCs isolated from different adult tissues.
Collapse
Affiliation(s)
- Bianca Vezzani
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| |
Collapse
|
49
|
Ceserani V, Ferri A, Berenzi A, Benetti A, Ciusani E, Pascucci L, Bazzucchi C, Coccè V, Bonomi A, Pessina A, Ghezzi E, Zeira O, Ceccarelli P, Versari S, Tremolada C, Alessandri G. Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue and its derived mesenchymal stromal cells. Vasc Cell 2016; 8:3. [PMID: 27547374 PMCID: PMC4991117 DOI: 10.1186/s13221-016-0037-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for advanced cell-based therapies. They are routinely obtained enzymatically from fat lipoaspirate (LP) as SVF, and may undergo prolonged ex vivo expansion, with significant senescence and decline in multipotency. Besides, these techniques have complex regulatory issues, thus incurring in the compelling requirements of GMP guidelines. Hence, availability of a minimally manipulated, autologous adipose tissue would have remarkable biomedical and clinical relevance. For this reason, a new device, named Lipogems® (LG), has been developed. This ready-to-use adipose tissue cell derivate has been shown to have in vivo efficacy upon transplantation for ischemic and inflammatory diseases. To broaden our knowledge, we here investigated the angiogenic and anti-inflammatory properties of LG and its derived MSC (LG-MSCs) population. METHODS Human LG samples and their LG-MSCs were analyzed by immunohistochemistry for pericyte, endothelial and mesenchymal stromal cell marker expression. Angiogenesis was investigated testing the conditioned media (CM) of LG (LG-CM) and LG-MSCs (LG-MSCs-CM) on cultured endothelial cells (HUVECs), evaluating proliferation, cord formation, and the expression of the adhesion molecules (AM) VCAM-1 and ICAM-1. The macrophage cell line U937 was used to evaluate the anti-inflammatory properties, such as migration, adhesion on HUVECs, and release of RANTES and MCP-1. RESULTS Our results indicate that LG contained a very high number of mesenchymal cells expressing NG2 and CD146 (both pericyte markers) together with an abundant microvascular endothelial cell (mEC) population. Substantially, both LG-CM and LG-MSC-CM increased cord formation, inhibited endothelial ICAM-1 and VCAM-1 expression following TNFα stimulation, and slightly improved HUVEC proliferation. The addition of LG-CM and LG-MSC-CM strongly inhibited U937 migration upon stimulation with the chemokine MCP-1, reduced their adhesion on HUVECs and significantly suppressed the release of RANTES and MCP-1. CONCLUSIONS Our data indicate that LG micro-fragmented adipose tissue retains either per se, or in its embedded MSCs content, the capacity to induce vascular stabilization and to inhibit several macrophage functions involved in inflammation.
Collapse
Affiliation(s)
- Valentina Ceserani
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Via Celoria 11, 20131 Milan, Italy
| | - Anna Ferri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Via Celoria 11, 20131 Milan, Italy
| | - Angiola Berenzi
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Anna Benetti
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, Fondazione IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Cinzia Bazzucchi
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Arianna Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Erica Ghezzi
- San Michele Veterinary Hospital, Tavezzano con Villavesco, Lodi, Italy
| | - Offer Zeira
- San Michele Veterinary Hospital, Tavezzano con Villavesco, Lodi, Italy
| | - Piero Ceccarelli
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | | | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Via Celoria 11, 20131 Milan, Italy
| |
Collapse
|
50
|
|