1
|
Tan J, Li X, Dou N. Insulin Resistance Triggers Atherosclerosis: Caveolin 1 Cooperates with PKCzeta to Block Insulin Signaling in Vascular Endothelial Cells. Cardiovasc Drugs Ther 2024; 38:885-893. [PMID: 37289375 PMCID: PMC11438709 DOI: 10.1007/s10557-023-07477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To date, therapies for endothelial dysfunction have primarily focused on ameliorating identified atherosclerosis (AS) risk factors rather than explicitly addressing endothelium-based mechanism. An in-depth exploration of the pathological mechanisms of endothelial injury was performed herein. METHODS Aortic caveolin 1 (Cav1) knockdown was achieved in mice using lentivirus, and AS was induced using a high-fat diet. Mouse body weight, blood glucose, insulin, lipid parameters, aortic plaque, endothelial injury, vascular nitric oxide synthase (eNOS), injury marker, and oxidative stress were examined. The effect of Cav1 knockdown on the content of PKCzeta and PI3K/Akt/eNOS pathway-related protein levels, as well as PKCzeta binding to Akt, was studied. ZIP, a PKCzeta inhibitor, was utilized to treat HUVECs in vitro, and the effect of ZIP on cell viability, inflammatory response, oxidative stress, and Akt activation was evaluated. RESULTS Cav1 knockdown had no significant effect on body weight or blood glucose in mice over an 8-week period, whereas drastically reduced insulin, lipid parameters, endothelial damage, E-selectin, and oxidative stress and elevated eNOS levels. Moreover, Cav1 knockdown triggered decreased PKCzeta enrichment and the activation of the PI3K/Akt/eNOS pathway. PKCzeta has a positive effect on cells without being coupled by Cav1, and ZIP had no marked influence on PKCzeta-Akt binding following Cav1/PKCzeta coupling. CONCLUSION Cav1/PKCzeta coupling antagonizes the activation of PI3K on Akt, leading to eNOS dysfunction, insulin resistance, and endothelial cell damage.
Collapse
Affiliation(s)
- Jingjing Tan
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
| | - Xiaoguang Li
- Department of Thyroid Breast and Vascular Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai, 200081, China
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Ning Dou
- Department of Thyroid Breast and Vascular Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai, 200081, China.
| |
Collapse
|
2
|
Jian Y, Li Y, Zhang Y, Tang M, Deng M, Liu C, Cheng M, Xiao S, Deng C, Wei Z. Lymphangiogenesis: novel strategies to promote cutaneous wound healing. BURNS & TRAUMA 2024; 12:tkae040. [PMID: 39328366 PMCID: PMC11427083 DOI: 10.1093/burnst/tkae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 09/28/2024]
Abstract
The cutaneous lymphatic system regulates tissue inflammation, fluid balance and immunological responses. Lymphangiogenesis or lymphatic dysfunction may lead to lymphedema, immune deficiency, chronic inflammation etc. Tissue regeneration and healing depend on angiogenesis and lymphangiogenesis during wound healing. Tissue oedema and chronic inflammation can slow wound healing due to impaired lymphangiogenesis or lymphatic dysfunction. For example, impaired lymphangiogenesis or lymphatic dysfunction has been detected in nonhealing wounds such as diabetic ulcers, venous ulcers and bedsores. This review summarizes the structure and function of the cutaneous lymphatic vessel system and lymphangiogenesis in wounds. Furthermore, we review wound lymphangiogenesis processes and remodelling, especially the influence of the inflammatory phase. Finally, we outline how to control lymphangiogenesis to promote wound healing, assess the possibility of targeting lymphangiogenesis as a novel treatment strategy for chronic wounds and provide an analysis of the possible problems that need to be addressed.
Collapse
Affiliation(s)
- Yang Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanqi Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanji Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingyuan Tang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingfu Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Chenxiaoxiao Liu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Maolin Cheng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Zairong Wei
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| |
Collapse
|
3
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
4
|
The Lymphatic Endothelium in the Context of Radioimmuno-Oncology. Cancers (Basel) 2022; 15:cancers15010021. [PMID: 36612017 PMCID: PMC9817924 DOI: 10.3390/cancers15010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The study of lymphatic tumor vasculature has been gaining interest in the context of cancer immunotherapy. These vessels constitute conduits for immune cells' transit toward the lymph nodes, and they endow tumors with routes to metastasize to the lymph nodes and, from them, toward distant sites. In addition, this vasculature participates in the modulation of the immune response directly through the interaction with tumor-infiltrating leukocytes and indirectly through the secretion of cytokines and chemokines that attract leukocytes and tumor cells. Radiotherapy constitutes the therapeutic option for more than 50% of solid tumors. Besides impacting transformed cells, RT affects stromal cells such as endothelial and immune cells. Mature lymphatic endothelial cells are resistant to RT, but we do not know to what extent RT may affect tumor-aberrant lymphatics. RT compromises lymphatic integrity and functionality, and it is a risk factor to the onset of lymphedema, a condition characterized by deficient lymphatic drainage and compromised tissue homeostasis. This review aims to provide evidence of RT's effects on tumor vessels, particularly on lymphatic endothelial cell physiology and immune properties. We will also explore the therapeutic options available so far to modulate signaling through lymphatic endothelial cell receptors and their repercussions on tumor immune cells in the context of cancer. There is a need for careful consideration of the RT dosage to come to terms with the participation of the lymphatic vasculature in anti-tumor response. Here, we provide new approaches to enhance the contribution of the lymphatic endothelium to radioimmuno-oncology.
Collapse
|
5
|
Buntinx F, Lebeau A, Gillot L, Baudin L, Ndong Penda R, Morfoisse F, Lallemand F, Vottero G, Nizet C, Nizet JL, Blacher S, Noel A. Single and combined impacts of irradiation and surgery on lymphatic vasculature and fibrosis associated to secondary lymphedema. Front Pharmacol 2022; 13:1016138. [PMID: 36330083 PMCID: PMC9622766 DOI: 10.3389/fphar.2022.1016138] [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: 08/10/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Lymphedema (LD) refers to a condition of lymphatic dysfunction associated with excessive fluid accumulation, fibroadipose tissue deposition and swelling. In industrialized countries, LD development mainly results from a local disruption of the lymphatic network by an infection or cancer-related surgery (secondary LD). In the absence of efficient therapy, animal models are needed to decipher the cellular and molecular mechanisms underlying LD and test putative drugs. In this study, we optimized and characterized a murine model of LD that combines an irradiation of the mice hind limb and a radical surgery (lymph node resection associated to lymphatic vessel ligation). We investigated the respective roles of irradiation and surgery in LD formation by comparing their impacts, alone or in combination (with different intervention sequences), on eight different features of the pathology: swelling (paw thickness), indocyanine green (ICG) clearance, lymphatic vasculature remodeling, epidermal and dermal thickening, adipocyte accumulation, inflammatory cell infiltration and collagen deposition. This study supports the importance of radiation prior to surgery to experimentally induce a rapid, severe and sustained tissue remodeling harboring the different hallmarks of LD. We provide the first experimental evidence for an excessive deposition of periostin (POSTN) and tenascin-C (TNC) in LD. Through a computerized method of digital image quantification, we established the spatial map of lymphatic expansion, as well as collagen, POSTN and TNC deposition in papillary and reticular dermis of lymphedematous skins. This mouse model is available to study the patho-physiology of LD and test potential therapeutic targets.
Collapse
Affiliation(s)
- F. Buntinx
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - A. Lebeau
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - L. Gillot
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - L. Baudin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - R. Ndong Penda
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - F. Morfoisse
- U1297-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - F. Lallemand
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
- Department of Radiotherapy-Oncology, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - G. Vottero
- Department of Plastic and Reconstructive Surgery, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - C. Nizet
- Department of Plastic and Reconstructive Surgery, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - J. L. Nizet
- Department of Plastic and Reconstructive Surgery, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - S. Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - A. Noel
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
- *Correspondence: A. Noel,
| |
Collapse
|
6
|
Zadory M, Lopez E, Babity S, Gravel SP, Brambilla D. Current knowledge on the tissue distribution of mRNA nanocarriers for therapeutic protein expression. Biomater Sci 2022; 10:6077-6115. [PMID: 36097955 DOI: 10.1039/d2bm00859a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exogenously delivered mRNA-based drugs are emerging as a new class of therapeutics with the potential to treat several diseases. Over the last decade, advancements in the design of non-viral delivery tools have enabled mRNA to be evaluated for several therapeutic purposes including protein replacement therapies, gene editing, and vaccines. However, in vivo delivery of mRNA to targeted organs and cells remains a critical challenge. Evaluation of the biodistribution of mRNA vehicles is of utmost importance for the development of effective pharmaceutical candidates. In this review, we discuss the recent advances in the design of nanoparticles loaded with mRNA and extrapolate the key factors influencing their biodistribution following administration. Finally, we highlight the latest developments in the preclinical and clinical translation of mRNA therapeutics for protein supplementation therapy.
Collapse
Affiliation(s)
- Matthias Zadory
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Elliot Lopez
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Samuel Babity
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Simon-Pierre Gravel
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Davide Brambilla
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| |
Collapse
|
7
|
Bekisz S, Baudin L, Buntinx F, Noël A, Geris L. In Vitro, In Vivo, and In Silico Models of Lymphangiogenesis in Solid Malignancies. Cancers (Basel) 2022; 14:1525. [PMID: 35326676 PMCID: PMC8946816 DOI: 10.3390/cancers14061525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Lymphangiogenesis (LA) is the formation of new lymphatic vessels by lymphatic endothelial cells (LECs) sprouting from pre-existing lymphatic vessels. It is increasingly recognized as being involved in many diseases, such as in cancer and secondary lymphedema, which most often results from cancer treatments. For some cancers, excessive LA is associated with cancer progression and metastatic dissemination to the lymph nodes (LNs) through lymphatic vessels. The study of LA through in vitro, in vivo, and, more recently, in silico models is of paramount importance in providing novel insights and identifying the key molecular actors in the biological dysregulation of this process under pathological conditions. In this review, the different biological (in vitro and in vivo) models of LA, especially in a cancer context, are explained and discussed, highlighting their principal modeled features as well as their advantages and drawbacks. Imaging techniques of the lymphatics, complementary or even essential to in vivo models, are also clarified and allow the establishment of the link with computational approaches. In silico models are introduced, theoretically described, and illustrated with examples specific to the lymphatic system and the LA. Together, these models constitute a toolbox allowing the LA research to be brought to the next level.
Collapse
Affiliation(s)
- Sophie Bekisz
- Biomechanics Research Unit, GIGA In silico Medicine, ULiège, 4000 Liège, Belgium;
| | - Louis Baudin
- Laboratory of Biology of Tumor and Development, GIGA Cancer, ULiège, 4000 Liège, Belgium; (L.B.); (F.B.); (A.N.)
| | - Florence Buntinx
- Laboratory of Biology of Tumor and Development, GIGA Cancer, ULiège, 4000 Liège, Belgium; (L.B.); (F.B.); (A.N.)
| | - Agnès Noël
- Laboratory of Biology of Tumor and Development, GIGA Cancer, ULiège, 4000 Liège, Belgium; (L.B.); (F.B.); (A.N.)
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In silico Medicine, ULiège, 4000 Liège, Belgium;
- Biomechanics Section, KU Leuven, 3000 Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
8
|
Molecular mechanisms of cyclic phosphatidic acid-induced lymphangiogenic actions in vitro. Microvasc Res 2021; 139:104273. [PMID: 34699844 DOI: 10.1016/j.mvr.2021.104273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/21/2022]
Abstract
The lymphatic system plays important roles in various physiological and pathological phenomena. As a bioactive phospholipid, lysophosphatidic acid (LPA) has been reported to function as a lymphangiogenic factor as well as some growth factors, yet the involvement of phospholipids including LPA and its derivatives in lymphangiogenesis is not fully understood. In the present study, we have developed an in-vitro lymphangiogenesis model (termed a collagen sandwich model) by utilizing type-I collagen, which exists around the lymphatic endothelial cells of lymphatic capillaries in vivo. The collagen sandwich model has revealed that cyclic phosphatidic acid (cPA), and not LPA, augmented the tube formation of human dermal lymphatic endothelial cells (HDLECs). Both cPA and LPA increased the migration of HDLECs cultured on the collagen. As the gene expression of LPA receptor 6 (LPA6) was predominantly expressed in HDLECs, a siRNA experiment against LPA6 attenuated the cPA-mediated tube formation. A synthetic LPA1/3 inhibitor, Ki16425, suppressed the cPA-augmented tube formation and migration of the HDLECs, and the LPA-induced migration. The activity of Rho-associated protein kinase (ROCK) located at the downstream of the LPA receptors was augmented in both the cPA- and LPA-treated cells. A potent ROCK inhibitor, Y-27632, suppressed the cPA-dependent tube formation but not the migration of the HDLECs. Furthermore, cPA, but not LPA, augmented the gene expression of VE-cadherin and β-catenin in the HDLECs. These results provide novel evidence that cPA facilitates the capillary-like morphogenesis and the migration of HDLECs through LPA6/ROCK and LPA1/3 signaling pathways in concomitance with the augmentation of VE-cadherin and β-catenin expression. Thus, cPA is likely to be a potent lymphangiogenic factor for the initial lymphatics adjacent to type I collagen under physiological conditions.
Collapse
|
9
|
Sex-Based Differences in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:499-533. [PMID: 34664253 DOI: 10.1007/978-3-030-73119-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Cancers are heterogeneous multifactorial diseases consisting of a major public health issue worldwide. Sex disparities are evidenced in cancer incidence, mortality, expression of prognosis factor, response to treatment, and survival. For both sexes, an interplay of intrinsic and environmental factors influences cancer cells and tumor microenvironment (TME) components. The TME cumulates both supportive and communicative functions, contributing to cancer development, progression, and metastasis dissemination. The frontline topics of this chapter are focused on the contribution of sex, via steroid hormones, such as estrogens and androgens, on the following components of the TME: cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), blood and lymphatic endothelial cells, and immunity/inflammatory system.
Collapse
|
10
|
Nikolenko VN, Oganesyan MV, Vovkogon AD, Nikitina AT, Sozonova EA, Kudryashova VA, Rizaeva NA, Cabezas R, Avila-Rodriguez M, Neganova ME, Mikhaleva LM, Bachurin SO, Somasundaram SG, Kirkland CE, Tarasov VV, Aliev G. Current Understanding of Central Nervous System Drainage Systems: Implications in the Context of Neurodegenerative Diseases. Curr Neuropharmacol 2021; 18:1054-1063. [PMID: 31729299 PMCID: PMC7709156 DOI: 10.2174/1570159x17666191113103850] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/15/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022] Open
Abstract
Until recently, it was thought that there were no lymphatic vessels in the central nervous system (CNS). Therefore, all metabolic processes were assumed to take place only in the circulation of the cerebrospinal fluid (CSF) and through the blood-brain barrier's (BBB), which regulate ion transport and ensure the functioning of the CNS. However, recent findings yield a new perspective: There is an exchange of CSF with interstitial fluid (ISF), which is drained to the paravenous space and reaches lymphatic nodes at the end. This circulation is known as the glymphatic system. The glymphatic system is an extensive network of meningeal lymphatic vessels (MLV) in the basal area of the skull that provides another path for waste products from CNS to reach the bloodstream. MLV develop postnatally, initially appearing around the foramina in the basal part of the skull and the spinal cord, thereafter sprouting along the skull's blood vessels and spinal nerves in various areas of the meninges. VEGF-C protein (vascular endothelial growth factor), expressed mainly by vascular smooth cells, plays an important role in the development of the MLV. The regenerative potential and plasticity of MLV and the novel discoveries related to CNS drainage offer potential for the treatment of neurodegenerative diseases such as dementia, hydrocephalus, stroke, multiple sclerosis, and Alzheimer disease (AD). Herein, we present an overview of the structure and function of the glymphatic system and MLV, and their potential involvement in the pathology and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Vladimir N Nikolenko
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia,Department of Normal and Topographic Anatomy, Federal State Budget Educational Institution of Higher Education M.V. Lomonosov Moscow State University, Leninskie Gory, 1, Moscow, 119991, Russia
| | - Marine V Oganesyan
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Angela D Vovkogon
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Arina T Nikitina
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Ekaterina A Sozonova
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Valentina A Kudryashova
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Negoria A Rizaeva
- Department of Human Anatomy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Ricardo Cabezas
- Department of Biochemistry and Nutrition, Science Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Marco Avila-Rodriguez
- Health Sciences Faculty, Clinic Sciences Department, University of Tolima, 730006 Ibague, Colombia
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia
| | - Liudmila M Mikhaleva
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russian Federation
| | - Sergey O Bachurin
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia
| | | | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, Salem, WV, USA
| | - Vadim V Tarasov
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Gjumrakch Aliev
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia,Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russian Federation,Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia,GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX, 78229, USA
| |
Collapse
|
11
|
Wasson EM, Dubbin K, Moya ML. Go with the flow: modeling unique biological flows in engineered in vitro platforms. LAB ON A CHIP 2021; 21:2095-2120. [PMID: 34008661 DOI: 10.1039/d1lc00014d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Interest in recapitulating in vivo phenomena in vitro using organ-on-a-chip technology has grown rapidly and with it, attention to the types of fluid flow experienced in the body has followed suit. These platforms offer distinct advantages over in vivo models with regards to human relevance, cost, and control of inputs (e.g., controlled manipulation of biomechanical cues from fluid perfusion). Given the critical role biophysical forces play in several tissues and organs, it is therefore imperative that engineered in vitro platforms capture the complex, unique flow profiles experienced in the body that are intimately tied with organ function. In this review, we outline the complex and unique flow regimes experienced by three different organ systems: blood vasculature, lymphatic vasculature, and the intestinal system. We highlight current state-of-the-art platforms that strive to replicate physiological flows within engineered tissues while introducing potential limitations in current approaches.
Collapse
Affiliation(s)
- Elisa M Wasson
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Karen Dubbin
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Monica L Moya
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| |
Collapse
|
12
|
Klaourakis K, Vieira JM, Riley PR. The evolving cardiac lymphatic vasculature in development, repair and regeneration. Nat Rev Cardiol 2021; 18:368-379. [PMID: 33462421 PMCID: PMC7812989 DOI: 10.1038/s41569-020-00489-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 02/08/2023]
Abstract
The lymphatic vasculature has an essential role in maintaining normal fluid balance in tissues and modulating the inflammatory response to injury or pathogens. Disruption of normal development or function of lymphatic vessels can have severe consequences. In the heart, reduced lymphatic function can lead to myocardial oedema and persistent inflammation. Macrophages, which are phagocytic cells of the innate immune system, contribute to cardiac development and to fibrotic repair and regeneration of cardiac tissue after myocardial infarction. In this Review, we discuss the cardiac lymphatic vasculature with a focus on developments over the past 5 years arising from the study of mammalian and zebrafish model organisms. In addition, we examine the interplay between the cardiac lymphatics and macrophages during fibrotic repair and regeneration after myocardial infarction. Finally, we discuss the therapeutic potential of targeting the cardiac lymphatic network to regulate immune cell content and alleviate inflammation in patients with ischaemic heart disease.
Collapse
Affiliation(s)
- Konstantinos Klaourakis
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- British Heart Foundation-Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford, UK
| | - Joaquim M Vieira
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
- British Heart Foundation-Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford, UK.
| | - Paul R Riley
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
- British Heart Foundation-Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Lee S, Kang H, Park D, Yu J, Koh SK, Cho D, Kim D, Kang K, Jeon NL. Modeling 3D Human Tumor Lymphatic Vessel Network Using High‐Throughput Platform. Adv Biol (Weinh) 2021. [DOI: 10.1002/adbi.202000195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Somin Lee
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Habin Kang
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - James Yu
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Seung Kwon Koh
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Duck Cho
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
- Department of Laboratory Medicine and Genetics Samsung Medical Center Sungkyunkwan University School of Medicine 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Da‐Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Kyung‐Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of Advanced Machinery and Design Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of BioEngineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| |
Collapse
|
14
|
Permana AD, Nainu F, Moffatt K, Larrañeta E, Donnelly RF. Recent advances in combination of microneedles and nanomedicines for lymphatic targeted drug delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1690. [PMID: 33401339 DOI: 10.1002/wnan.1690] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Numerous diseases have been reported to affect the lymphatic system. As such, several strategies have been developed to deliver chemotherapeutics to this specific network of tissues and associated organs. Nanotechnology has been exploited as one of the main approaches to improve the lymphatic uptake of drugs. Different nanoparticle approaches utilized for both active and passive targeting of the lymphatic system are discussed here. Specifically, due to the rich abundance of lymphatic capillaries in the dermis, particular attention is given to this route of administration, as intradermal administration could potentially result in higher lymphatic uptake compared to other routes of administration. Recently, progress in microneedle research has attracted particular attention as an alternative for the use of conventional hypodermic injections. The benefits of microneedles, when compared to intradermal injection, are subsequently highlighted. Importantly, microneedles exhibit particular benefit in relation to therapeutic targeting of the lymphatic system, especially when combined with nanoparticles, which are further discussed. However, despite the apparent benefits provided by this combination approach, further comprehensive preclinical and clinical studies are now necessary to realize the potential extent of this dual-delivery platform, further taking into consideration eventual usability and acceptability in the intended patient end-users. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Kurtis Moffatt
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | | | | |
Collapse
|
15
|
Cui Y, Chen G, Yang Z. Mitochondrial superoxide mediates PM 2.5-induced cytotoxicity in human pulmonary lymphatic endothelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114423. [PMID: 32222623 DOI: 10.1016/j.envpol.2020.114423] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/29/2020] [Accepted: 03/19/2020] [Indexed: 06/10/2023]
Abstract
Exposure to airborne fine particulate matter (PM2.5) is associated with a variety of respiratory health effects and contributes to premature mortality. Lymphatic vessels are instrumental in facilitating the transport of toxic materials away from the lung to maintain alveolar clearance and have been shown to play important roles in lung injury and repair. Despite intense research efforts in delineating the effects of PM2.5 on blood vascular endothelial cells, the impacts of PM2.5 on lymphatic endothelial cells (LECs), a specialized subset of endothelial cells that comprise lymphatic vessels, remain enigmatic. Here, we conducted MTT assay and show that treatment of human pulmonary LECs with PM2.5 suppresses cell viability in a time- and dose-dependent manner. We subsequently performed Annexin V/propidium iodide labeling and demonstrate that PM2.5 induces LECs apoptosis and necrosis. Furthermore, we found that manganese superoxide dismutase (SOD2) expression and mitochondrial SOD activity were profoundly reduced following PM2.5 exposure. Mechanistically, we provide compelling evidence that PM2.5 reduces SOD2 expression through activation of Akt pathway, which leads to a disruption of mitochondrial redox homeostasis characterized by increased accumulation of mitochondrial superoxide. Conversely, mitochondria-targeted SOD mimetic (MitoTEMPO) corrects the disturbed oxidative milieu in PM2.5-treated LECs. Additionally, MitoTEMPO ameliorates the deleterious impacts of PM2.5 on mitochondrial DNA integrity and preserves the viability of LECs. Taken together, these novel data support a critical role for mitochondrial superoxide in the pathogenesis of PM2.5-induced LECs injury and identity mitochondrial-targeted antioxidants as promising therapeutic options to treat environmental lung diseases. Our findings are limited to experimental studies with primary LECs, and future investigations in animal models are warranted to shed light on the precise pathophysiology of lymphatic system in response to PM exposure.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, People's Republic of China.
| | - Guang Chen
- Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Zeran Yang
- Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| |
Collapse
|
16
|
Luque‐González MA, Reis RL, Kundu SC, Caballero D. Human Microcirculation‐on‐Chip Models in Cancer Research: Key Integration of Lymphatic and Blood Vasculatures. ACTA ACUST UNITED AC 2020; 4:e2000045. [DOI: 10.1002/adbi.202000045] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/27/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Maria Angélica Luque‐González
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineICVS/3B’s—PT Government Associate Laboratory AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Braga/Guimarães Portugal
| | - Rui Luis Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineICVS/3B’s—PT Government Associate Laboratory AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Braga/Guimarães Portugal
| | - Subhas Chandra Kundu
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineICVS/3B’s—PT Government Associate Laboratory AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Braga/Guimarães Portugal
| | - David Caballero
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineICVS/3B’s—PT Government Associate Laboratory AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Braga/Guimarães Portugal
| |
Collapse
|
17
|
Connolly S, Newport D, McGourty K. The mechanical responses of advecting cells in confined flow. BIOMICROFLUIDICS 2020; 14:031501. [PMID: 32454924 PMCID: PMC7200165 DOI: 10.1063/5.0005154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/21/2020] [Indexed: 05/03/2023]
Abstract
Fluid dynamics have long influenced cells in suspension. Red blood cells and white blood cells are advected through biological microchannels in both the cardiovascular and lymphatic systems and, as a result, are subject to a wide variety of complex fluidic forces as they pass through. In vivo, microfluidic forces influence different biological processes such as the spreading of infection, cancer metastasis, and cell viability, highlighting the importance of fluid dynamics in the blood and lymphatic vessels. This suggests that in vitro devices carrying cell suspensions may influence the viability and functionality of cells. Lab-on-a-chip, flow cytometry, and cell therapies involve cell suspensions flowing through microchannels of approximately 100-800 μ m. This review begins by examining the current fundamental theories and techniques behind the fluidic forces and inertial focusing acting on cells in suspension, before exploring studies that have investigated how these fluidic forces affect the reactions of suspended cells. In light of these studies' findings, both in vivo and in vitro fluidic cell microenvironments shall also be discussed before concluding with recommendations for the field.
Collapse
Affiliation(s)
- S Connolly
- School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - D Newport
- School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | | |
Collapse
|
18
|
Systemic Sclerosis Serum Significantly Impairs the Multi-Step Lymphangiogenic Process: in Vitro Evidence. Int J Mol Sci 2019; 20:ijms20246189. [PMID: 31817940 PMCID: PMC6940874 DOI: 10.3390/ijms20246189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/22/2019] [Accepted: 12/06/2019] [Indexed: 01/29/2023] Open
Abstract
In systemic sclerosis (SSc), the possible involvement of lymphatic microcirculation and lymphangiogenesis has traditionally been overshadowed by the greater emphasis placed on dysfunctional blood vascular system and angiogenesis. In the present in vitro study, we explore for the first time whether the SSc microenvironment may interfere with lymphangiogenesis, a complex, multi-step process in which lymphatic microvascular endothelial cells (LMVECs) sprout, migrate, and proliferate to generate new lymphatic capillaries. Normal human adult dermal LMVECs from three donors were treated with serum from SSc patients (n = 8), serum from healthy individuals (n = 8), or recombinant human vascular endothelial growth factor (VEGF)-C as a positive control for lymphangiogenesis. Cell proliferation, Boyden chamber Matrigel chemoinvasion, wound healing capacity, and lymphatic capillary morphogenesis on Geltrex were assayed. VEGF-C serum levels were measured by enzyme-linked immunosorbent assay. Gene and protein expression levels of the lymphangiogenic orchestrators VEGF receptor-3 (VEGFR-3)/Flt-4 and neuropilin-2 (NRP-2) were determined by real-time PCR and Western blotting, respectively. Conditioning with SSc serum significantly inhibited LMVEC proliferation, Matrigel invasion, and wound healing capacity with respect to healthy serum. The ability of LMVECs to form lymphatic tubes on Geltrex was also severely compromised in the presence of SSc serum. VEGF-C levels were comparable in SSc and healthy sera. Treatment with SSc serum resulted in a significant downregulation of both VEGFR-3/Flt-4 and NRP-2 mRNA and protein levels. In SSc, the pathologic environment severely hampers every lymphangiogenesis step, likely through the reduction of pro-lymphangiogenic VEGFR-3/NRP-2 co-receptor signaling. The impairment of the lymphangiogenic process opens a new scenario underlying SSc vascular pathophysiology, which is worth investigating further.
Collapse
|
19
|
Blei F. Update October 2019. Lymphat Res Biol 2019. [DOI: 10.1089/lrb.2019.29072.fb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|