1
|
Westcott GP, Emont MP, Gulko A, Zhou Z, Kim C, Varma G, Tsai LL, O'Donnell E, Loureiro ZY, Liang W, Jacobs C, Tsai LT, Padera TP, Singhal D, Rosen ED. Single-nuclear transcriptomics of lymphedema-associated adipose reveals a pro-lymphangiogenic stromal cell population. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638907. [PMID: 40027673 PMCID: PMC11870541 DOI: 10.1101/2025.02.18.638907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Chronic lymphedema is a progressive, disfiguring disease that results from dysfunction of the lymphatic vasculature, causing distal accumulation of interstitial fluid, localized development of tissue edema, and expansion of subcutaneous adipose tissue (SAT). As the molecular mechanisms governing SAT remodeling in this disease are unclear, we performed single-nucleus RNA sequencing on paired control and affected SAT biopsies from patients with unilateral lymphedema. Lymphedema samples were characterized by expansion of SAA + adipocytes, pro-adipogenic stem cells, and proliferation of lymphatic capillaries. A GRIA1 + lymphedema-enriched stromal cell population expressing VEGFC , ADAMTS3 , and CCBE1 was identified, suggesting an enhanced axis of communication between adipose stem and progenitor cells (ASPCs) and lymphatic endothelial cells. Furthermore, lymphedema ASPC-conditioned media promoted lymphatic endothelial tube elongation in vitro . These findings indicate a critical role for ASPCs in regulating adipocyte differentiation and lymphatic vascular remodeling in lymphedema, and provide a valuable resource for better understanding this disease.
Collapse
|
2
|
Yang Y, Wu X, Zhao Y, Zhang D, Zhang L, Cai X, Ji J, Jing Z, Boström KI, Yao Y. Arterial-Lymphatic-Like Endothelial Cells Appear in Hereditary Hemorrhagic Telangiectasia 2 and Contribute to Vascular Leakage and Arteriovenous Malformations. Circulation 2025; 151:299-317. [PMID: 39429196 PMCID: PMC11789604 DOI: 10.1161/circulationaha.124.070925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Arteriovenous malformations (AVMs) are characteristic of hereditary hemorrhagic telangiectasia. Loss-of-function mutations in the activin receptor-like kinase 1 (Alk1) are linked to hemorrhagic telangiectasia type 2. METHODS Endothelial-specific deletion of Alk1, endothelial lineage tracing, transcriptomics of single-cell analysis, and electron microscopy were performed to examine the vascular phenotype and characteristics of ALK1-deficient endothelial cells (ECs) after EC-specific Alk1 deletion. Ischemia assays were used to examine the cell capacity for vascular malformation. Connectivity Map with transcriptomic analysis was applied to identify chemical compounds. Specific methods for arteriovenous malformations, such as micro-computed tomography, with other molecular and cell biological tools were also performed. RESULTS We performed endothelial-specific deletion of Alk1 in mice and found severe arteriovenous malformations and vascular leakage. The transcriptomics of single-cell analysis revealed a new distinctive cell cluster formed after Alk1 deletion where the cells coexpressed arterial and lymphatic endothelial markers. The analysis projected that these cells potentially originated from arterial ECs after Alk1 deletion. This new population was referred to as arterial-lymphatic-like ECs according to its cellular markers, and its appearance was validated in the pulmonary small arteries after Alk1 deletion. Transplantation of these cells caused vascular malformations. Endothelial lineage tracing confirmed that these new arterial-lymphatic-like ECs were derived from ALK1 depleted ECs, potentially arterial ECs. We discovered that SOX17 (SRY-box transcription factor 17) induction was responsible for the derivation of these arterial-lymphatic-like ECs. We showed that direct binding of MDM2 (mouse double minute 2) was required for Sox17 to execute this activity. Inhibition of MDM2 reduced the arteriovenous malformations in the mouse model. CONCLUSIONS Together, our studies revealed the mechanistic underpinnings of ALK1 signaling in regulating the endothelial phenotype and provided possibilities for new therapeutic strategies in hemorrhagic telangiectasia type 2.
Collapse
MESH Headings
- Animals
- Arteriovenous Malformations/pathology
- Arteriovenous Malformations/genetics
- Arteriovenous Malformations/metabolism
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/pathology
- Telangiectasia, Hereditary Hemorrhagic/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Mice
- Activin Receptors, Type II/genetics
- Activin Receptors, Type II/metabolism
- Activin Receptors, Type II/deficiency
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/deficiency
- Disease Models, Animal
- SOXF Transcription Factors/metabolism
- SOXF Transcription Factors/genetics
- Humans
- Mice, Knockout
- Arteries/pathology
- Arteries/metabolism
- Lymphatic Vessels/pathology
- Lymphatic Vessels/metabolism
Collapse
Affiliation(s)
- Yang Yang
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Yan Zhao
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Daoqin Zhang
- Department of Pediatrics, Stanford University, CA (D.Z.)
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Jaden Ji
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Zheng Jing
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| | - Kristina I. Boström
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
- Molecular Biology Institute (K.I.B.), University of California, Los Angeles
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine (Y. Yang, X.W., Y.Z., L.Z., X.C., J.J., Z.J., K.I.B., Y. Yao), University of California, Los Angeles
| |
Collapse
|
3
|
Doni A, Sironi M, Del Prete A, Pasqualini F, Valentino S, Cuccovillo I, Parente R, Calvi M, Tosoni A, Vago G, Nebuloni M, Garlanda C, Vecchi A, Bottazzi B, Mantovani A. PTX3 is expressed in terminal lymphatics and shapes their organization and function. Front Immunol 2024; 15:1426869. [PMID: 39640269 PMCID: PMC11617523 DOI: 10.3389/fimmu.2024.1426869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction The lymphatic system is a multifaceted regulator of tissue homeostasis and an integral part of immune responses. Previous studies had shown that subsets of lymphatic endothelial cells (LEC) express PTX3, an essential component of humoral innate immunity and tissue homeostasis. Methods In the present study using whole-mount imaging and image-based morphometric quantifications, Ptx3-targeted mice and in vivo functional analysis, we investigated the involvement of PTX3 in shaping and function of the lymphatic vasculature. Results We found that PTX3 is localized in the extracellular matrix (ECM) surrounding human and murine lymphatic vessels (LV). In murine tissues, PTX3 was localized in the ECM close to LV terminals and sprouting. Ptx3-deficient mice showed LV abnormalities in the colon submucosa and diaphragm, including a disorganized pattern and hyperplasia of initial LV capillaries associated with altered distribution of tight junction-associated molecules. Mice with LEC-restricted PTX3 gene inactivation showed morphological and organization abnormalities similar to those observed in Ptx3-deficient animals. Ptx3-deficient mice showed defective fluid drainage from footpads and defective dendritic cell (DC) trafficking. Discussion Thus, PTX3 is strategically localized in the ECM of specialized LV, playing an essential role in their structural organization and immunological function.
Collapse
Affiliation(s)
- Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Marina Sironi
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Annalisa Del Prete
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Fabio Pasqualini
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Sonia Valentino
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Ivan Cuccovillo
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Raffaella Parente
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Michela Calvi
- Clinical and Experimental Immunology Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Antonella Tosoni
- Pathology Unit, L. Sacco Hospital, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Gianluca Vago
- Pathology Unit, L. Sacco Hospital, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Manuela Nebuloni
- Pathology Unit, L. Sacco Hospital, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Cecilia Garlanda
- Experimental Immunopathology Lab, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Annunciata Vecchi
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Barbara Bottazzi
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alberto Mantovani
- Cellular and Humoral Innate Immunity Lab, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
4
|
Limbu S, McCloskey KE. An Endothelial Cell Is Not Simply an Endothelial Cell. Stem Cells Dev 2024; 33:517-527. [PMID: 39030822 PMCID: PMC11564855 DOI: 10.1089/scd.2024.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 07/22/2024] Open
Abstract
Endothelial cells (ECs) are a multifaceted component of the vascular system with roles in immunity, maintaining tissue fluid balance, and vascular tone. Dysregulation or dysfunction of ECs can have far-reaching implications, leading pathologies ranging from cardiovascular diseases, such as hypertension and atherosclerosis, ischemia, chronic kidney disease, blood-brain barrier integrity, dementia, and tumor metastasis. Recent advancements in regenerative medicine have highlighted the potential of stem cell-derived ECs, particularly from induced pluripotent stem cells, to treat ischemic tissues, as well as models of vascular integrity. This review summarizes what is known in the generation of ECs with an emphasis on tissue-specific ECs and EC subphenotypes important in the development of targeted cell-based therapies for patient treatment.
Collapse
Affiliation(s)
- Shiwani Limbu
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
| | - Kara E. McCloskey
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
- Materials Science and Engineering Department, University of California, Merced, USA
| |
Collapse
|
5
|
Huang J, Liao C, Yang J, Zhang L. The role of vascular and lymphatic networks in bone and joint homeostasis and pathology. Front Endocrinol (Lausanne) 2024; 15:1465816. [PMID: 39324127 PMCID: PMC11422228 DOI: 10.3389/fendo.2024.1465816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
The vascular and lymphatic systems are integral to maintaining skeletal homeostasis and responding to pathological conditions in bone and joint tissues. This review explores the interplay between blood vessels and lymphatic vessels in bones and joints, focusing on their roles in homeostasis, regeneration, and disease progression. Type H blood vessels, characterized by high expression of CD31 and endomucin, are crucial for coupling angiogenesis with osteogenesis, thus supporting bone homeostasis and repair. These vessels facilitate nutrient delivery and waste removal, and their dysfunction can lead to conditions such as ischemia and arthritis. Recent discoveries have highlighted the presence and significance of lymphatic vessels within bone tissue, challenging the traditional view that bones are devoid of lymphatics. Lymphatic vessels contribute to interstitial fluid regulation, immune cell trafficking, and tissue repair through lymphangiocrine signaling. The pathological alterations in these networks are closely linked to inflammatory joint diseases, emphasizing the need for further research into their co-regulatory mechanisms. This comprehensive review summarizes the current understanding of the structural and functional aspects of vascular and lymphatic networks in bone and joint tissues, their roles in homeostasis, and the implications of their dysfunction in disease. By elucidating the dynamic interactions between these systems, we aim to enhance the understanding of their contributions to skeletal health and disease, potentially informing the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jingxiong Huang
- Center of Stomatology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| | - Chengcheng Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Guizhou, Zunyi, China
| | - Jian Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liang Zhang
- Center of Stomatology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
7
|
Huang X, Jie S, Li W, Liu C. CHRDL2 activates the PI3K/AKT pathway to ameliorate glucocorticoid-induced damages to bone microvascular endothelial cells (BMECs). Heliyon 2024; 10:e33867. [PMID: 39050472 PMCID: PMC11268171 DOI: 10.1016/j.heliyon.2024.e33867] [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: 05/26/2023] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024] Open
Abstract
Steroid-induced avascular necrosis of the femoral head (ANFH) is characterized by the death of bone tissues, leading to the impairment of normal reparative processes within micro-fractures in the femoral head. Glucocorticoid (GCs)-induced bone microvascular endothelial cell (BMEC) damage has been reported to contribute to ANFH development. In this study, differentially expressed genes (DEGs) between necrosis of the femoral head (NFH) and normal samples were analyzed based on two sets of online expression profiles, GSE74089 and GSE26316. Chordin-like 2 (CHRDL2) was found to be dramatically downregulated in NFH samples. In GCs-stimulated BMECs, cellular damages were observed alongside CHRDL2 down-regulation. GCs-caused cell viability suppression, cell apoptosis promotion, tubule formation suppression, and cell migration suppression were partially abolished by CHRDL2 overexpression but amplified by CHRDL2 knockdown; consistent trends were observed in GCs-caused alterations in the protein levels of VEGFA, VEGFR2, and BMP-9 levels, and the ratios of Bax/Bcl-2 and cleaved-caspase3/Caspase3. GC stimulation significantly inhibited PI3K and Akt phosphorylation in BMECs, whereas the inhibitor effects of GCs on PI3K and Akt phosphorylation were partially attenuated by CHRDL2 overexpression but further amplified by CHRDL2 knockdown. Moreover, CHRDL2 overexpression caused improvement in GCs-induced damages to BMECs that were partially eliminated by PI3K inhibitor LY294002. In conclusion, CHRDL2 is down-regulated in NFH samples and GCs-stimulated BMECs. CHRDL2 overexpression could improve GCs-caused BMEC apoptosis and dysfunctions, possibly via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xianzhe Huang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuo Jie
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Wenzhao Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chan Liu
- International Medical Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|
8
|
Veloso A, Bleuart A, Conrard L, Orban T, Bruyr J, Cabochette P, Germano RFV, Schevenels G, Bernard A, Zindy E, Demeyer S, Vanhollebeke B, Dequiedt F, Martin M. The cytoskeleton adaptor protein Sorbs1 controls the development of lymphatic and venous vessels in zebrafish. BMC Biol 2024; 22:51. [PMID: 38414014 PMCID: PMC10900589 DOI: 10.1186/s12915-024-01850-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Lymphangiogenesis, the formation of lymphatic vessels, is tightly linked to the development of the venous vasculature, both at the cellular and molecular levels. Here, we identify a novel role for Sorbs1, the founding member of the SoHo family of cytoskeleton adaptor proteins, in vascular and lymphatic development in the zebrafish. RESULTS We show that Sorbs1 is required for secondary sprouting and emergence of several vascular structures specifically derived from the axial vein. Most notably, formation of the precursor parachordal lymphatic structures is affected in sorbs1 mutant embryos, severely impacting the establishment of the trunk lymphatic vessel network. Interestingly, we show that Sorbs1 interacts with the BMP pathway and could function outside of Vegfc signaling. Mechanistically, Sorbs1 controls FAK/Src signaling and subsequently impacts on the cytoskeleton processes regulated by Rac1 and RhoA GTPases. Inactivation of Sorbs1 altered cell-extracellular matrix (ECM) contacts rearrangement and cytoskeleton dynamics, leading to specific defects in endothelial cell migratory and adhesive properties. CONCLUSIONS Overall, using in vitro and in vivo assays, we identify Sorbs1 as an important regulator of venous and lymphatic angiogenesis independently of the Vegfc signaling axis. These results provide a better understanding of the complexity found within context-specific vascular and lymphatic development.
Collapse
Affiliation(s)
- Alexandra Veloso
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Anouk Bleuart
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Louise Conrard
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Tanguy Orban
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Jonathan Bruyr
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Pauline Cabochette
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
- Present Address: Laboratory of Developmental Genetics, ULB Neuroscience Institute, Université Libre de Bruxelles, B-6041, Gosselies, Belgium
| | - Raoul F V Germano
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Giel Schevenels
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Alice Bernard
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, University of Liège (ULiège), Liège, Belgium
| | - Egor Zindy
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Sofie Demeyer
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Benoit Vanhollebeke
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium
| | - Franck Dequiedt
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium
| | - Maud Martin
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège (ULiège), Liège, Belgium.
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, University of Liège (ULiège), Liège, Belgium.
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium.
- Department of Molecular Biology, Laboratory of Neurovascular Signaling, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), B-6041, Gosselies, Belgium.
- WEL Research Institute (WELBIO Department), Avenue Pasteur, 6, 1300, Wavre, Belgium.
| |
Collapse
|
9
|
Shang P, Zheng R, Wu K, Yuan C, Pan S. New Insights on Mechanisms and Therapeutic Targets of Cerebral Edema. Curr Neuropharmacol 2024; 22:2330-2352. [PMID: 38808718 PMCID: PMC11451312 DOI: 10.2174/1570159x22666240528160237] [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: 11/30/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 05/30/2024] Open
Abstract
Cerebral Edema (CE) is the final common pathway of brain death. In severe neurological disease, neuronal cell damage first contributes to tissue edema, and then Increased Intracranial Pressure (ICP) occurs, which results in diminishing cerebral perfusion pressure. In turn, anoxic brain injury brought on by decreased cerebral perfusion pressure eventually results in neuronal cell impairment, creating a vicious cycle. Traditionally, CE is understood to be tightly linked to elevated ICP, which ultimately generates cerebral hernia and is therefore regarded as a risk factor for mortality. Intracranial hypertension and brain edema are two serious neurological disorders that are commonly treated with mannitol. However, mannitol usage should be monitored since inappropriate utilization of the substance could conversely have negative effects on CE patients. CE is thought to be related to bloodbrain barrier dysfunction. Nonetheless, a fluid clearance mechanism called the glial-lymphatic or glymphatic system was updated. This pathway facilitates the transport of cerebrospinal fluid (CSF) into the brain along arterial perivascular spaces and later into the brain interstitium. After removing solutes from the neuropil into meningeal and cervical lymphatic drainage arteries, the route then directs flows into the venous perivascular and perineuronal regions. Remarkably, the dual function of the glymphatic system was observed to protect the brain from further exacerbated damage. From our point of view, future studies ought to concentrate on the management of CE based on numerous targets of the updated glymphatic system. Further clinical trials are encouraged to apply these agents to the clinic as soon as possible.
Collapse
Affiliation(s)
- Pei Shang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Neurology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Ruoyi Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kou Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chao Yuan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Fotie J, Matherne CM, Wroblewski JE. Silicon switch: Carbon-silicon Bioisosteric replacement as a strategy to modulate the selectivity, physicochemical, and drug-like properties in anticancer pharmacophores. Chem Biol Drug Des 2023; 102:235-254. [PMID: 37029092 DOI: 10.1111/cbdd.14239] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/09/2023]
Abstract
Bioisosterism is one of the leading strategies in medicinal chemistry for the design and modification of drugs, consisting in replacing an atom or a substituent with a different atom or a group with similar chemical properties and an inherent biocompatibility. The objective of such an exercise is to produce a diversity of molecules with similar behavior while enhancing the desire biological and pharmacological properties, without inducing significant changes to the chemical framework. In drug discovery and development, the optimization of the absorption, distribution, metabolism, elimination, and toxicity (ADMETox) profile is of paramount importance. Silicon appears to be the right choice as a carbon isostere because they possess very similar intrinsic properties. However, the replacement of a carbon by a silicon atom in pharmaceuticals has proven to result in improved efficacy and selectivity, while enhancing physicochemical properties and bioavailability. The current review discusses how silicon has been strategically introduced to modulate drug-like properties of anticancer agents, from a molecular design strategy, biological activity, computational modeling, and structure-activity relationships perspectives.
Collapse
Affiliation(s)
- Jean Fotie
- Department of Chemistry and Physics, Southeastern Louisiana University, Hammond, Louisiana, USA
| | - Caitlyn M Matherne
- Department of Chemistry and Physics, Southeastern Louisiana University, Hammond, Louisiana, USA
| | - Jordan E Wroblewski
- Department of Chemistry and Physics, Southeastern Louisiana University, Hammond, Louisiana, USA
| |
Collapse
|
11
|
Alkhathami AG, Abdullah MR, Ahmed M, Hassan Ahmed H, Alwash SW, Muhammed Mahdi Z, Alsaikhan F, Dera AA. Bone morphogenetic protein (BMP)9 in cancer development: mechanistic, diagnostic, and therapeutic approaches? J Drug Target 2023:1-11. [PMID: 37461888 DOI: 10.1080/1061186x.2023.2236330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Bone morphogenetic protein (BMP)-9 is considered a member of the transforming growth factor (TGF)β superfamily. It was first found as an inducer of bone and cartilage formation and then discovered that this factor mediates several physiologic functions and hemostasis. Besides physiological conditions, BMP9 has also been elucidated that it is involved in several pathological situations, especially cancer. In various cancers, dysregulation of BMP9 has raised the issue that BMP9 might play a conflicting role in tumour development. BMP9 binding to its receptors (BMPRs), including ALKs and BMPRII, induces canonical SMAD-dependent and non-canonical PI3K/AKT and MAPK signalling pathways in tumour cells. BMP9, via inducing apoptosis, inhibiting tumour-promoting cell signalling pathways, suppressing epithelial-mesenchymal transition (EMT) process, blocking angiogenesis, and preventing cross-talk in the tumour microenvironment, mainly exerts tumour-suppressive functions. In contrast, BMP9 triggers tumour-supportive signalling pathways, promotes EMT, and enhances angiogenesis, suggesting that BMP9 is also involved in tumour development. It has been demonstrated that modulating BMP9 expression and functions might be a promising approach to cancer treatment. It has also been indicated that evaluating BMP9 expression in cancers might be a biomarker for predicting cancer prognosis. Overall, BMP9 would provide a promising target in cancer management.
Collapse
Affiliation(s)
- Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Muhjaha Ahmed
- Medical Technical college, Al-Farahidi University, Iraq
| | | | - Sarab W Alwash
- Medical Laboratory Techniques Department, Al-Mustaqbal University College, Babylon, Iraq Hillah
| | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Ayed A Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
12
|
Liu M, Sui L, Fang Z, Jiang WG, Ye L. Aberrant expression of bone morphogenetic proteins in the disease progression and metastasis of breast cancer. Front Oncol 2023; 13:1166955. [PMID: 37333824 PMCID: PMC10272747 DOI: 10.3389/fonc.2023.1166955] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Background Bone morphogenetic proteins (BMPs) play crucial roles in the tumorigenesis and metastasis of cancers. Controversy remains about the exact implications of BMPs and their antagonists in breast cancer (BC), due to their diverse and complex biological functions and signalling. A comprehensive study of the whole family and their signalling in breast cancer is provoked. Methods Aberrant expression of BMP, BMP receptors and antagonists in primary tumours in breast cancer were analysed by using TCGA-BRCA and E-MTAB-6703 cohorts. Related biomarkers including ER, HER, proliferation, invasion, angiogenesis, lymphangiogenesis and bone metastasis were involved to identify the relationship with BMPs in breast cancer. Results The present study showed BMP8B was significantly increased in breast tumours, while BMP6 and ACVRL1 were decreased in breast cancer tissues. The expressions of BMP2, BMP6, TGFBR1 and GREM1 were significantly correlated with BC patients' poor overall survival. Aberrant expression of BMPs, together with BMP receptors, were explored in different subtypes of breast cancer according to ER, PR and HER2 status. Furthermore, higher levels of BMP2, BMP6 and GDF5 were revealed in triple negative breast cancer (TNBC) whilst BMP4, GDF15, ACVR1B, ACVR2B and BMPR1B were relatively higher in Luminal type BC. ACVR1B and BMPR1B were positively correlated with ERα but were inversely correlated with ERβ. High expression of GDF15, BMP4 and ACVR1B were associated with poorer overall survival in HER2 positive BC. BMPs also play dual roles in tumour growth and metastasis of BC. Conclusion A shift pattern of BMPs was showed in different subtypes of breast cancer suggesting a subtype specific involvement. It provokes more research to shed light on the exact role of these BMPs and receptors in the disease progression and distant metastasis through a regulation of proliferation, invasion and EMT.
Collapse
Affiliation(s)
- Ming Liu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
- Department of Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, China
| | - Laijian Sui
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ziqian Fang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Wen G. Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
13
|
Lan YL, Wang H, Chen A, Zhang J. Update on the current knowledge of lymphatic drainage system and its emerging roles in glioma management. Immunology 2023; 168:233-247. [PMID: 35719015 DOI: 10.1111/imm.13517] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/22/2022] [Indexed: 01/17/2023] Open
Abstract
The draining of brain interstitial fluid (ISF) to cerebrospinal fluid (CSF) and the subsequent draining of CSF to meningeal lymphatics is well-known. Nonetheless, its role in the development of glioma is a remarkable finding that has to be extensively understood. The glymphatic system (GS) collects CSF from the subarachnoid space and brain ISF through aquaporin-4 (AQP4) water channels. The glial limiting membrane and the perivascular astrocyte-end-feet membrane both have elevated levels of AQP4. CSF is thought to drain through the nerve sheaths of the olfactory and other cranial nerves as well as spinal meningeal lymphatics via dorsal or basal lymphatic vessels. Meningeal lymphatic vessels (MLVs) exist below the skull in the dorsal and basal regions. In this view, MLVs offer a pathway to drain macromolecules and traffic immunological cells from the CNS into cervical lymph nodes (CLNs), and thus can be used as a candidate curing strategy against glioma and other associated complications, such as neuro-inflammation. Taken together, the lymphatic drainage system could provide a route or approach for drug targeting of glioma and other neurological conditions. Nevertheless, its pathophysiological role in glioma remains elusive, which needs extensive research. The current review aims to explore the lymphatic drainage system, its role in glioma progression, and possible therapeutic techniques that target MLVs in the CNS.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongjin Wang
- Department of Neurology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Aiqin Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Choi H, Kim BG, Kim YH, Lee SJ, Lee YJ, Oh SP. BMP10 functions independently from BMP9 for the development of a proper arteriovenous network. Angiogenesis 2023; 26:167-186. [PMID: 36348215 PMCID: PMC9908740 DOI: 10.1007/s10456-022-09859-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic vascular disorder characterized by the presence of arteriovenous malformation (AVM) in multiple organs. HHT is caused by mutations in genes encoding major constituents for transforming growth factor-β (TGF-β) family signaling: endoglin (ENG), activin receptor-like kinase 1 (ALK1), and SMAD4. The identity of physiological ligands for this ENG-ALK1 signaling pertinent to AVM formation has yet to be clearly determined. To investigate whether bone morphogenetic protein 9 (BMP9), BMP10, or both are physiological ligands of ENG-ALK1 signaling involved in arteriovenous network formation, we generated a novel Bmp10 conditional knockout mouse strain. We examined whether global Bmp10-inducible knockout (iKO) mice develop AVMs at neonatal and adult stages in comparison with control, Bmp9-KO, and Bmp9/10-double KO (dKO) mice. Bmp10-iKO and Bmp9/10-dKO mice showed AVMs in developing retina, postnatal brain, and adult wounded skin, while Bmp9-KO did not display any noticeable vascular defects. Bmp10 deficiency resulted in increased proliferation and size of endothelial cells in AVM vessels. The impaired neurovascular integrity in the brain and retina of Bmp10-iKO and Bmp9/10-dKO mice was detected. Bmp9/10-dKO mice exhibited the lethality and vascular malformation similar to Bmp10-iKO mice, but their phenotypes were more pronounced. Administration of BMP10 protein, but not BMP9 protein, prevented retinal AVM in Bmp9/10-dKO and endothelial-specific Eng-iKO mice. These data indicate that BMP10 is indispensable for the development of a proper arteriovenous network, whereas BMP9 has limited compensatory functions for the loss of BMP10. We suggest that BMP10 is the most relevant physiological ligand of the ENG-ALK1 signaling pathway pertinent to HHT pathogenesis.
Collapse
Affiliation(s)
- Hyunwoo Choi
- Barrow Aneurysm & AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Road, Phoenix, AZ, 85013, USA
| | - Bo-Gyeong Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Gaetbeol-Ro, Yeonsu-Gu, 21999, Incheon, Republic of Korea
| | - Yong Hwan Kim
- Barrow Aneurysm & AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Road, Phoenix, AZ, 85013, USA
| | - Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Young Jae Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Gaetbeol-Ro, Yeonsu-Gu, 21999, Incheon, Republic of Korea.
- Department of Biochemistry, Gachon University College of Medicine, Incheon, Republic of Korea.
| | - S Paul Oh
- Barrow Aneurysm & AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Road, Phoenix, AZ, 85013, USA.
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
15
|
Patnam M, Dommaraju SR, Masood F, Herbst P, Chang JH, Hu WY, Rosenblatt MI, Azar DT. Lymphangiogenesis Guidance Mechanisms and Therapeutic Implications in Pathological States of the Cornea. Cells 2023; 12:319. [PMID: 36672254 PMCID: PMC9856498 DOI: 10.3390/cells12020319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/22/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Corneal lymphangiogenesis is one component of the neovascularization observed in several inflammatory pathologies of the cornea including dry eye disease and corneal graft rejection. Following injury, corneal (lymph)angiogenic privilege is impaired, allowing ingrowth of blood and lymphatic vessels into the previously avascular cornea. While the mechanisms underlying pathological corneal hemangiogenesis have been well described, knowledge of the lymphangiogenesis guidance mechanisms in the cornea is relatively scarce. Various signaling pathways are involved in lymphangiogenesis guidance in general, each influencing one or multiple stages of lymphatic vessel development. Most endogenous factors that guide corneal lymphatic vessel growth or regression act via the vascular endothelial growth factor C signaling pathway, a central regulator of lymphangiogenesis. Several exogenous factors have recently been repurposed and shown to regulate corneal lymphangiogenesis, uncovering unique signaling pathways not previously known to influence lymphatic vessel guidance. A strong understanding of the relevant lymphangiogenesis guidance mechanisms can facilitate the development of targeted anti-lymphangiogenic therapeutics for corneal pathologies. In this review, we examine the current knowledge of lymphatic guidance cues, their regulation of inflammatory states in the cornea, and recently discovered anti-lymphangiogenic therapeutic modalities.
Collapse
Affiliation(s)
- Mehul Patnam
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunil R. Dommaraju
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Faisal Masood
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark I. Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
16
|
Itoh F, Watabe T. TGF-β signaling in lymphatic vascular vessel formation and maintenance. Front Physiol 2022; 13:1081376. [PMID: 36589453 PMCID: PMC9799095 DOI: 10.3389/fphys.2022.1081376] [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: 10/27/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor (TGF)-β and its family members, including bone morphogenetic proteins (BMPs), nodal proteins, and activins, are implicated in the development and maintenance of various organs. Here, we review its role in the lymphatic vascular system (the secondary vascular system in vertebrates), which plays a crucial role in various physiological and pathological processes, participating in the maintenance of the normal tissue fluid balance, immune cell trafficking, and fatty acid absorption in the gut. The lymphatic system is associated with pathogenesis in multiple diseases, including lymphedema, inflammatory diseases, and tumor metastasis. Lymphatic vessels are composed of lymphatic endothelial cells, which differentiate from blood vascular endothelial cells (BECs). Although TGF-β family signaling is essential for maintaining blood vessel function, little is known about the role of TGF-β in lymphatic homeostasis. Recently, we reported that endothelial-specific depletion of TGF-β signaling affects lymphatic function. These reports suggest that TGF-β signaling in lymphatic endothelial cells maintains the structure of lymphatic vessels and lymphatic homeostasis, and promotes tumor lymphatic metastasis. Suppression of TGF-β signaling in lymphatic endothelial cells may therefore be effective in inhibiting cancer metastasis. We highlight recent advances in understanding the roles of TGF-β signaling in the formation and maintenance of the lymphatic system.
Collapse
Affiliation(s)
- Fumiko Itoh
- Laboratory of Stem Cells Regulations, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan,*Correspondence: Fumiko Itoh, ; Tetsuro Watabe,
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,*Correspondence: Fumiko Itoh, ; Tetsuro Watabe,
| |
Collapse
|
17
|
Ehata S, Miyazono K. Bone Morphogenetic Protein Signaling in Cancer; Some Topics in the Recent 10 Years. Front Cell Dev Biol 2022; 10:883523. [PMID: 35693928 PMCID: PMC9174896 DOI: 10.3389/fcell.2022.883523] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/09/2022] [Indexed: 12/19/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-β (TGF-β) family, are multifunctional cytokines. BMPs have a broad range of functions, and abnormalities in BMP signaling pathways are involved in cancer progression. BMPs activate the proliferation of certain cancer cells. Malignant phenotypes of cancer cells, such as increased motility, invasiveness, and stemness, are enhanced by BMPs. Simultaneously, BMPs act on various cellular components and regulate angiogenesis in the tumor microenvironment. Thus, BMPs function as pro-tumorigenic factors in various types of cancer. However, similar to TGF-β, which shows both positive and negative effects on tumorigenesis, BMPs also act as tumor suppressors in other types of cancers. In this article, we review important findings published in the recent decade and summarize the pro-oncogenic functions of BMPs and their underlying mechanisms. The current status of BMP-targeted therapies for cancers is also discussed.
Collapse
Affiliation(s)
- Shogo Ehata
- Department of Pathology, School of Medicine, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Shogo Ehata,
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Kulikauskas MR, X S, Bautch VL. The versatility and paradox of BMP signaling in endothelial cell behaviors and blood vessel function. Cell Mol Life Sci 2022; 79:77. [PMID: 35044529 PMCID: PMC8770421 DOI: 10.1007/s00018-021-04033-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022]
Abstract
Blood vessels expand via sprouting angiogenesis, and this process involves numerous endothelial cell behaviors, such as collective migration, proliferation, cell–cell junction rearrangements, and anastomosis and lumen formation. Subsequently, blood vessels remodel to form a hierarchical network that circulates blood and delivers oxygen and nutrients to tissue. During this time, endothelial cells become quiescent and form a barrier between blood and tissues that regulates transport of liquids and solutes. Bone morphogenetic protein (BMP) signaling regulates both proangiogenic and homeostatic endothelial cell behaviors as blood vessels form and mature. Almost 30 years ago, human pedigrees linked BMP signaling to diseases associated with blood vessel hemorrhage and shunts, and recent work greatly expanded our knowledge of the players and the effects of vascular BMP signaling. Despite these gains, there remain paradoxes and questions, especially with respect to how and where the different and opposing BMP signaling outputs are regulated. This review examines endothelial cell BMP signaling in vitro and in vivo and discusses the paradox of BMP signals that both destabilize and stabilize endothelial cell behaviors.
Collapse
Affiliation(s)
- Molly R Kulikauskas
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shaka X
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
19
|
Jiang QQ, Liu BB, Xu KS. New insights into BMP9 signaling in liver diseases. Mol Cell Biochem 2021; 476:3591-3600. [PMID: 34019202 DOI: 10.1007/s11010-021-04182-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/12/2021] [Indexed: 02/08/2023]
Abstract
Bone morphogenetic protein 9 (BMP9) is a recently discovered cytokine mainly secreted by the liver and is a member of the transforming growth factor β (TGF-β) superfamily. In recent years, an increasing number of studies have shown that BMP9 is associated with liver diseases, including nonalcoholic fatty liver disease (NAFLD), liver fibrosis and hepatocellular carcinoma (HCC), and BMP9 signaling may play dual roles in liver diseases. In this review, we mainly summarized and discussed the roles and potential mechanisms of BMP9 signaling in NAFLD, liver fibrosis and HCC. Specifically, this article will provide a better understanding of BMP9 signaling and new clues for the treatment of liver diseases.
Collapse
Affiliation(s)
- Qian-Qian Jiang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bei-Bei Liu
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke-Shu Xu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
20
|
Chen S, Shao L, Ma L. Cerebral Edema Formation After Stroke: Emphasis on Blood-Brain Barrier and the Lymphatic Drainage System of the Brain. Front Cell Neurosci 2021; 15:716825. [PMID: 34483842 PMCID: PMC8415457 DOI: 10.3389/fncel.2021.716825] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Brain edema is a severe stroke complication that is associated with prolonged hospitalization and poor outcomes. Swollen tissues in the brain compromise cerebral perfusion and may also result in transtentorial herniation. As a physical and biochemical barrier between the peripheral circulation and the central nervous system (CNS), the blood–brain barrier (BBB) plays a vital role in maintaining the stable microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the dysfunction of the BBB results in increased paracellular permeability, directly contributing to the extravasation of blood components into the brain and causing cerebral vasogenic edema. Recent studies have led to the discovery of the glymphatic system and meningeal lymphatic vessels, which provide a channel for cerebrospinal fluid (CSF) to enter the brain and drain to nearby lymph nodes and communicate with the peripheral immune system, modulating immune surveillance and brain responses. A deeper understanding of the function of the cerebral lymphatic system calls into question the known mechanisms of cerebral edema after stroke. In this review, we first discuss how BBB disruption after stroke can cause or contribute to cerebral edema from the perspective of molecular and cellular pathophysiology. Finally, we discuss how the cerebral lymphatic system participates in the formation of cerebral edema after stroke and summarize the pathophysiological process of cerebral edema formation after stroke from the two directions of the BBB and cerebral lymphatic system.
Collapse
Affiliation(s)
- Sichao Chen
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linqian Shao
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Ma
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Desroches-Castan A, Tillet E, Bouvard C, Bailly S. BMP9 and BMP10: two close vascular quiescence partners that stand out. Dev Dyn 2021; 251:178-197. [PMID: 34240497 DOI: 10.1002/dvdy.395] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are dimeric transforming growth factor ß (TGFß) family cytokines that were first described in bone and cartilage formation but have since been shown to be involved in many pleiotropic functions. In human, there are 15 BMP ligands, which initiate their cellular signaling by forming a complex with two copies of type I receptors and two copies of type II receptors, both of which are transmembrane receptors with an intracellular serine/threonine kinase domain. Within this receptor family, ALK1 (Activin receptor-Like Kinase 1), which is a type I receptor mainly expressed on endothelial cells, and BMPRII (BMP Receptor type II), a type II receptor also highly expressed on endothelial cells, have been directly linked to two rare vascular diseases: hereditary haemorrhagic telangiectasia (HHT), and pulmonary arterial hypertension (PAH), respectively. BMP9 (gene name GDF2) and BMP10, two close members of the BMP family, are the only known ligands for the ALK1 receptor. This specificity gives them a unique role in physiological and pathological angiogenesis and tissue homeostasis. The aim of this current review is to present an overview of what is known about BMP9 and BMP10 on vascular regulation with a particular emphasis on recent results and the many questions that remain unanswered regarding the roles and specificities between BMP9 and BMP10. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Emmanuelle Tillet
- Laboratory BioSanté, Univ. Grenoble Alpes, INSERM, CEA, Grenoble, France
| | - Claire Bouvard
- Laboratory BioSanté, Univ. Grenoble Alpes, INSERM, CEA, Grenoble, France
| | - Sabine Bailly
- Laboratory BioSanté, Univ. Grenoble Alpes, INSERM, CEA, Grenoble, France
| |
Collapse
|
22
|
Han O, Pak B, Jin SW. The Role of BMP Signaling in Endothelial Heterogeneity. Front Cell Dev Biol 2021; 9:673396. [PMID: 34235147 PMCID: PMC8255612 DOI: 10.3389/fcell.2021.673396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023] Open
Abstract
Bone morphogenetic proteins (BMPs), which compose the largest group of the transforming growth factor-β (TGF-ß) superfamily, have been implied to play a crucial role in diverse physiological processes. The most intriguing feature of BMP signaling is that it elicits heterogeneous responses from cells with equivalent identity, thus permitting highly context-dependent signaling outcomes. In endothelial cells (ECs), which are increasingly perceived as a highly heterogeneous population of cells with respect to their morphology, function, as well as molecular characteristics, BMP signaling has shown to elicit diverse and often opposite effects, illustrating the innate complexity of signaling responses. In this review, we provide a concise yet comprehensive overview of how outcomes of BMP signaling are modulated in a context-dependent manner with an emphasis on the underlying molecular mechanisms and summarize how these regulations of the BMP signaling promote endothelial heterogeneity.
Collapse
Affiliation(s)
- Orjin Han
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Boryeong Pak
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Suk-Won Jin
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| |
Collapse
|
23
|
The BMP Pathway in Blood Vessel and Lymphatic Vessel Biology. Int J Mol Sci 2021; 22:ijms22126364. [PMID: 34198654 PMCID: PMC8232321 DOI: 10.3390/ijms22126364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) were originally identified as the active components in bone extracts that can induce ectopic bone formation. In recent decades, their key role has broadly expanded beyond bone physiology and pathology. Nowadays, the BMP pathway is considered an important player in vascular signaling. Indeed, mutations in genes encoding different components of the BMP pathway cause various severe vascular diseases. Their signaling contributes to the morphological, functional and molecular heterogeneity among endothelial cells in different vessel types such as arteries, veins, lymphatic vessels and capillaries within different organs. The BMP pathway is a remarkably fine-tuned pathway. As a result, its signaling output in the vessel wall critically depends on the cellular context, which includes flow hemodynamics, interplay with other vascular signaling cascades and the interaction of endothelial cells with peri-endothelial cells and the surrounding matrix. In this review, the emerging role of BMP signaling in lymphatic vessel biology will be highlighted within the framework of BMP signaling in the circulatory vasculature.
Collapse
|
24
|
He M, He Q, Cai X, Chen Z, Lao S, Deng H, Liu X, Zheng Y, Liu X, Liu J, Xie Z, Yao M, Liang W, He J. Role of lymphatic endothelial cells in the tumor microenvironment-a narrative review of recent advances. Transl Lung Cancer Res 2021; 10:2252-2277. [PMID: 34164274 PMCID: PMC8182726 DOI: 10.21037/tlcr-21-40] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background As lymphatic vessel is a major route for solid tumor metastasis, they are considered an essential part of tumor drainage conduits. Apart from forming the walls of lymphatic vessels, lymphatic endothelial cells (LECs) have been found to play multiple other roles in the tumor microenvironment, calling for a more in-depth review. We hope that this review may help researchers gain a detailed understanding of this fast-developing field and shed some light upon future research. Methods To achieve an informative review of recent advance, we carefully searched the Medline database for English literature that are openly published from the January 1995 to December 2020 and covered the topic of LEC or lymphangiogenesis in tumor progression and therapies. Two different authors independently examined the literature abstracts to exclude possible unqualified ones, and 310 papers with full texts were finally retrieved. Results In this paper, we discussed the structural and molecular basis of tumor-associated LECs, together with their roles in tumor metastasis and drug therapy. We then focused on their impacts on tumor cells, tumor stroma, and anti-tumor immunity, and the molecular and cellular mechanisms involved. Special emphasis on lung cancer and possible therapeutic targets based on LECs were also discussed. Conclusions LECs can play a much more complex role than simply forming conduits for tumor cell dissemination. Therapies targeting tumor-associated lymphatics for lung cancer and other tumors are promising, but more research is needed to clarify the mechanisms involved.
Collapse
Affiliation(s)
- Miao He
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qihua He
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiuyu Cai
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zisheng Chen
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Respiratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Shen Lao
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongsheng Deng
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiwen Liu
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongmei Zheng
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Liu
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun Liu
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhanhong Xie
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Maojin Yao
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,The First People Hospital of Zhaoqing, Zhaoqing, China
| | - Jianxing He
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
25
|
Marziano C, Genet G, Hirschi KK. Vascular endothelial cell specification in health and disease. Angiogenesis 2021; 24:213-236. [PMID: 33844116 PMCID: PMC8205897 DOI: 10.1007/s10456-021-09785-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/17/2021] [Indexed: 02/08/2023]
Abstract
There are two vascular networks in mammals that coordinately function as the main supply and drainage systems of the body. The blood vasculature carries oxygen, nutrients, circulating cells, and soluble factors to and from every tissue. The lymphatic vasculature maintains interstitial fluid homeostasis, transports hematopoietic cells for immune surveillance, and absorbs fat from the gastrointestinal tract. These vascular systems consist of highly organized networks of specialized vessels including arteries, veins, capillaries, and lymphatic vessels that exhibit different structures and cellular composition enabling distinct functions. All vessels are composed of an inner layer of endothelial cells that are in direct contact with the circulating fluid; therefore, they are the first responders to circulating factors. However, endothelial cells are not homogenous; rather, they are a heterogenous population of specialized cells perfectly designed for the physiological demands of the vessel they constitute. This review provides an overview of the current knowledge of the specification of arterial, venous, capillary, and lymphatic endothelial cell identities during vascular development. We also discuss how the dysregulation of these processes can lead to vascular malformations, and therapeutic approaches that have been developed for their treatment.
Collapse
Affiliation(s)
- Corina Marziano
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Gael Genet
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Department of Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
26
|
Abstract
The lymphatic vasculature is a vital component of the vertebrate vascular system that mediates tissue fluid homeostasis, lipid uptake and immune surveillance. The development of the lymphatic vasculature starts in the early vertebrate embryo, when a subset of blood vascular endothelial cells of the cardinal veins acquires lymphatic endothelial cell fate. These cells sprout from the veins, migrate, proliferate and organize to give rise to a highly structured and unique vascular network. Cellular cross-talk, cell-cell communication and the interpretation of signals from surrounding tissues are all essential for coordinating these processes. In this chapter, we highlight new findings and review research progress with a particular focus on LEC migration and guidance, expansion of the LEC lineage, network remodeling and morphogenesis of the lymphatic vasculature.
Collapse
|
27
|
Eye lymphatic defects induced by bone morphogenetic protein 9 deficiency have no functional consequences on intraocular pressure. Sci Rep 2020; 10:16040. [PMID: 32994463 PMCID: PMC7524742 DOI: 10.1038/s41598-020-71877-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/18/2020] [Indexed: 11/08/2022] Open
Abstract
Aqueous humor drainage is essential for the regulation of intraocular pressure (IOP), a major risk factor for glaucoma. The Schlemm's canal and the non-conventional uveoscleral pathway are known to drain aqueous humor from the eye anterior chamber. It has recently been reported that lymphatic vessels are involved in this process, and that the Schlemm's canal responds to some lymphatic regulators. We have previously shown a critical role for bone morphogenetic protein 9 (BMP9) in lymphatic vessel maturation and valve formation, with repercussions in drainage efficiency. Here, we imaged eye lymphatic vessels and analyzed the consequences of Bmp9 (Gdf2) gene invalidation. A network of lymphatic vessel hyaluronan receptor 1 (LYVE-1)-positive lymphatic vessels was observed in the corneolimbus and the conjunctiva. In contrast, LYVE-1-positive cells present in the ciliary bodies were belonging to the macrophage lineage. Although enlarged conjunctival lymphatic trunks and a reduced valve number were observed in Bmp9-KO mice, there were no morphological differences in the Schlemm's canal compared to wild type animals. Moreover, there were no functional consequences on IOP in both basal control conditions and after laser-induced ocular hypertonia. Thus, the BMP9-activated signaling pathway does not constitute a wise target for new glaucoma therapeutic strategies.
Collapse
|
28
|
Gutierrez-Miranda L, Yaniv K. Cellular Origins of the Lymphatic Endothelium: Implications for Cancer Lymphangiogenesis. Front Physiol 2020; 11:577584. [PMID: 33071831 PMCID: PMC7541848 DOI: 10.3389/fphys.2020.577584] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022] Open
Abstract
The lymphatic system plays important roles in physiological and pathological conditions. During cancer progression in particular, lymphangiogenesis can exert both positive and negative effects. While the formation of tumor associated lymphatic vessels correlates with metastatic dissemination, increased severity and poor patient prognosis, the presence of functional lymphatics is regarded as beneficial for anti-tumor immunity and cancer immunotherapy delivery. Therefore, a profound understanding of the cellular origins of tumor lymphatics and the molecular mechanisms controlling their formation is required in order to improve current strategies to control malignant spread. Data accumulated over the last decades have led to a controversy regarding the cellular sources of tumor-associated lymphatic vessels and the putative contribution of non-endothelial cells to this process. Although it is widely accepted that lymphatic endothelial cells (LECs) arise mainly from pre-existing lymphatic vessels, additional contribution from bone marrow-derived cells, myeloid precursors and terminally differentiated macrophages, has also been claimed. Here, we review recent findings describing new origins of LECs during embryonic development and discuss their relevance to cancer lymphangiogenesis.
Collapse
Affiliation(s)
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
29
|
Cell Fate Determination of Lymphatic Endothelial Cells. Int J Mol Sci 2020; 21:ijms21134790. [PMID: 32640757 PMCID: PMC7370169 DOI: 10.3390/ijms21134790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/18/2022] Open
Abstract
The lymphatic vasculature, along with the blood vasculature, is a vascular system in our body that plays important functions in fluid homeostasis, dietary fat uptake, and immune responses. Defects in the lymphatic system are associated with various diseases such as lymphedema, atherosclerosis, fibrosis, obesity, and inflammation. The first step in lymphangiogenesis is determining the cell fate of lymphatic endothelial cells. Several genes involved in this commitment step have been identified using animal models, including genetically modified mice. This review provides an overview of these genes in the mammalian system and related human diseases.
Collapse
|
30
|
Aukema SM, Ten Brinke GA, Timens W, Vos YJ, Accord RE, Kraft KE, Santing MJ, Morssink LP, Streefland E, van Diemen CC, Vrijlandt EJ, Hulzebos CV, Kerstjens-Frederikse WS. A homozygous variant in growth and differentiation factor 2 (GDF2) may cause lymphatic dysplasia with hydrothorax and nonimmune hydrops fetalis. Am J Med Genet A 2020; 182:2152-2160. [PMID: 32618121 DOI: 10.1002/ajmg.a.61743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/08/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023]
Abstract
The etiology of nonimmune hydrops fetalis is extensive and includes genetic disorders. We describe a term-born female neonate with late onset extensive nonimmune hydrops, that is, polyhydramnios, edema, and congenital bilateral chylothorax. This newborn was successfully treated with repetitive thoracocentesis, total parenteral feeding, octreotide intravenously and finally surgical pleurodesis and corticosteroids. A genetic cause seemed plausible as the maternal history revealed a fatal nonimmune hydrops fetalis. A homozygous truncating variant in GDF2 (c.451C>T, p.(Arg151*)) was detected with exome sequencing. Genetic analysis of tissue obtained from the deceased fetal sibling revealed the same homozygous variant. The parents and two healthy siblings were heterozygous for the GDF2 variant. Skin and lung biopsies in the index patient, as well as the revised lung biopsy of the deceased fetal sibling, showed lymphatic dysplasia and lymphangiectasia. To the best of our knowledge, this is the first report of an association between a homozygous variant in GDF2 with lymphatic dysplasia, hydrothorax and nonimmune hydrops fetalis.
Collapse
Affiliation(s)
- Sietse M Aukema
- Department of Clinical Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerdien A Ten Brinke
- Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Yvonne J Vos
- Department of Clinical Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ryan E Accord
- Department of Congenital Cardiothoracic Surgery, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Center for Congenital Heart Diseases, Groningen, The Netherlands
| | - Karianne E Kraft
- Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel J Santing
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Leonard P Morssink
- Department of Obstetrics and Gynaecology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Esther Streefland
- Department of Obstetrics and Gynecology/Prenatal diagnosis, University Medical Centre of Groningen, University of Groningen, Groningen, The Netherlands
| | - Cleo C van Diemen
- Department of Clinical Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elianne Jle Vrijlandt
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Christian V Hulzebos
- Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | | |
Collapse
|
31
|
Yoshimatsu Y, Kimuro S, Pauty J, Takagaki K, Nomiyama S, Inagawa A, Maeda K, Podyma-Inoue KA, Kajiya K, Matsunaga YT, Watabe T. TGF-beta and TNF-alpha cooperatively induce mesenchymal transition of lymphatic endothelial cells via activation of Activin signals. PLoS One 2020; 15:e0232356. [PMID: 32357159 PMCID: PMC7194440 DOI: 10.1371/journal.pone.0232356] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Lymphatic systems play important roles in the maintenance of fluid homeostasis and undergo anatomical and physiological changes during inflammation and aging. While lymphatic endothelial cells (LECs) undergo mesenchymal transition in response to transforming growth factor-β (TGF-β), the molecular mechanisms underlying endothelial-to-mesenchymal transition (EndMT) of LECs remain largely unknown. In this study, we examined the effect of TGF-β2 and tumor necrosis factor-α (TNF-α), an inflammatory cytokine, on EndMT using human skin-derived lymphatic endothelial cells (HDLECs). TGF-β2-treated HDLECs showed increased expression of SM22α, a mesenchymal cell marker accompanied by increased cell motility and vascular permeability, suggesting HDLECs to undergo EndMT. Our data also revealed that TNF-α could enhance TGF-β2-induced EndMT of HDLECs. Furthermore, both cytokines induced the production of Activin A while decreasing the expression of its inhibitory molecule Follistatin, and thus enhancing EndMT. Finally, we demonstrated that human dermal lymphatic vessels underwent EndMT during aging, characterized by double immunostaining for LYVE1 and SM22α. These results suggest that both TGF-β and TNF-α signals play a central role in EndMT of LECs and could be potential targets for senile edema.
Collapse
Affiliation(s)
- Yasuhiro Yoshimatsu
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Division of Pharmacology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shiori Kimuro
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Joris Pauty
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | | | | | - Akihiko Inagawa
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kentaro Maeda
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Katarzyna A. Podyma-Inoue
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | | | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- * E-mail:
| |
Collapse
|
32
|
TMEM100 is a key factor for specification of lymphatic endothelial progenitors. Angiogenesis 2020; 23:339-355. [PMID: 32112176 DOI: 10.1007/s10456-020-09713-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND TMEM100 is identified as a downstream gene of bone morphogenetic protein 9 (BMP9) signaling via activin receptor-like kinase 1 (ALK1), which is known to participate in lymphangiogenesis as well as angiogenesis. TMEM100 has been shown to be important for blood vessel formation and maintenance, but its role in the development of lymphatic vasculature remains unknown. The objective is to investigate the role of TMEM100 in development of the lymphatic system. METHODS AND RESULTS Global Tmem100 gene deletion was induced by tamoxifen on 10.5 days post-coitus. Tmem100-inducible knockout (iKO) embryos in embryonic days (E)14.5-16.5 exhibited edema and blood-filled enlarged lymphatics with misconnections between veins and lymphatic vessels. For a reciprocal approach, we have generated a novel mouse line in which TMEM100 overexpression (OE) can be induced in endothelial cells by intercrossing with Tie2-Cre driver. TMEM100-OE embryos at E12.5-14.5 exhibited edema with small size and number of lymphatic vessels, the exact opposite phenotypes of Tmem100-iKOs. In Tmem100-iKO embryos, the number of progenitors of lymphatic endothelial cells (LECs) in the cardinal vein was increased, while it was decreased in TMEM100-OE embryos. The activity of NOTCH signaling, which limits the number of progenitors of LECs in the cardinal vein, was decreased in Tmem100-iKO embryos, whereas it was increased in TMEM100-OE embryos. CONCLUSION TMEM100 plays an important role in the specification of LECs in the cardinal veins, at least in part, by regulating the NOTCH signaling.
Collapse
|
33
|
Sun Z, Liu C, Jiang WG, Ye L. Deregulated bone morphogenetic proteins and their receptors are associated with disease progression of gastric cancer. Comput Struct Biotechnol J 2020; 18:177-188. [PMID: 31988704 PMCID: PMC6965205 DOI: 10.1016/j.csbj.2019.12.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 12/05/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
Bone morphogenetic proteins (BMP) are members of the transforming growth factor β superfamily (TGF-β). BMPs are involved in tumourigenesis and disease progression of certain malignancies. To date, the role played by BMPs in gastric cancer (GC) remains largely unknown. In the present study, we systematically analysed the expression and clinical significance of BMP and BMP receptors (BMPR) in TCGA gastric cancer database and GEO database and explored the possible mechanism of action. BMP5 is reduced in gastric cancer tissues, while ACVRL1, ACVR1, TGFBR1, and BMPR2 were significantly increased in the gastric tumours. BMP3, ACVR1, TGFBR1, BMPR1B (also known as ALK6), TGFBR2 and BMPR2 were significantly associated with poorer overall survival of GC patients. A negative correlation was seen between BMP/BMPR and proliferation markers which was supported by their correlation with the cell cycle promoters and inhibitors. More interestingly, further analyses showed that BMPs and their receptors are positively correlated with matrix metalloproteinases (MMPs), epithelial mesenchymal transition (EMT) markers and stemness in GC. Furthermore, positive correlations were also frequently seen between BMP receptors and markers/regulators of angiogenesis and lymphangiogenesis in the gastric tumours. Taken together, these findings suggest that BMPs play dual roles in GC. They may inhibit proliferation of GC cells. On the other hand, they can also promote disease progression through a promotion of invasion, EMT and stemness. The elevated expression of BMP receptors in GC were also highly associated with tumour associated angiogenesis and lymphangiogenesis which facilitate tumour growth, expansion and spread.
Collapse
Affiliation(s)
- Zhiwei Sun
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.,VIP-II Division of Medical Department, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Chang Liu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
34
|
Fan Y, Guo L, Zheng H, Ji C, Wang W, Sun H. BMP-9 is a novel marker for colorectal tumorigenesis undergoing the normal mucosa-adenoma-adenocarcinoma sequence and is associated with colorectal cancer prognosis. Oncol Lett 2020; 19:271-282. [PMID: 31897139 PMCID: PMC6923933 DOI: 10.3892/ol.2019.11125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 10/10/2019] [Indexed: 01/29/2023] Open
Abstract
Depending on the type of cancer, bone morphogenetic protein-9 (BMP-9) can promote or inhibit tumorigenesis; however, the function of BMP-9 in colorectal cancer remains unclear. The aim of the present study was to evaluate the clinicopathological importance of BMP-9 expression in the tumorigenesis of normal colorectal epithelial tissue, and subsequent transformation into adenoma and carcinoma. In addition, the present study aimed to determine the prognostic value of BMP-9 on the survival of patients with colorectal cancer (CRC). A total of 65 patients with pathologically confirmed colorectal adenocarcinoma and a history of adenoma were enrolled. BMP-9 and Ki-67 expression was assessed retrospectively using paraffin-embedded samples of normal colorectal mucosa, colorectal adenoma and CRC obtained from each patient. The prognostic value of BMP-9 expression was analyzed in a group comprising 48 patients with CRC and a mean follow-up duration of 39.1 months. Bioinformatics analyses were performed in order to validate the results of the present study using published CRC datasets. The results from the present study suggested that the expression of BMP-9 gradually increased during the transition from normal mucosa to adenoma and subsequent adenocarcinoma (P<0.05); however, no significant association between the expression levels of BMP-9 and the clinicopathological parameters of patients was reported. Kaplan-Meier analysis revealed that patients with high expression levels of BMP-9 exhibited shorter overall survival rate than those with low levels of expression (54.7 vs. 41.3 months; log-rank test, P<0.05). Furthermore, regardless of tumor location and the presence of blood vessel tumor emboli, the univariate and multivariate analyses indicated that BMP-9 expression may be an independent prognostic factor for the overall survival rate of patients with CRC. The results of the present study suggested that BMP-9 may serve an oncogenic role and possess prognostic value in CRC.
Collapse
Affiliation(s)
- Yinjie Fan
- Department of General Surgery, The Affiliated Zhengzhou Central Hospital of Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Lingxiang Guo
- Department of General Surgery, The Affiliated Zhengzhou Central Hospital of Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Huachuan Zheng
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Chunyong Ji
- Department of General Surgery, The Affiliated Zhengzhou Central Hospital of Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Wenbin Wang
- Department of Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Hongzhi Sun
- Department of Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China.,Key Laboratory of Tumor Clinical Metabolomics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
35
|
Esposito E, Ahn BJ, Shi J, Nakamura Y, Park JH, Mandeville ET, Yu Z, Chan SJ, Desai R, Hayakawa A, Ji X, Lo EH, Hayakawa K. Brain-to-cervical lymph node signaling after stroke. Nat Commun 2019; 10:5306. [PMID: 31757960 PMCID: PMC6876639 DOI: 10.1038/s41467-019-13324-w] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 10/27/2019] [Indexed: 12/24/2022] Open
Abstract
After stroke, peripheral immune cells are activated and these systemic responses may amplify brain damage, but how the injured brain sends out signals to trigger systemic inflammation remains unclear. Here we show that a brain-to-cervical lymph node (CLN) pathway is involved. In rats subjected to focal cerebral ischemia, lymphatic endothelial cells proliferate and macrophages are rapidly activated in CLNs within 24 h, in part via VEGF-C/VEGFR3 signalling. Microarray analyses of isolated lymphatic endothelium from CLNs of ischemic mice confirm the activation of transmembrane tyrosine kinase pathways. Blockade of VEGFR3 reduces lymphatic endothelial activation, decreases pro-inflammatory macrophages, and reduces brain infarction. In vitro, VEGF-C/VEGFR3 signalling in lymphatic endothelial cells enhances inflammatory responses in co-cultured macrophages. Lastly, surgical removal of CLNs in mice significantly reduces infarction after focal cerebral ischemia. These findings suggest that modulating the brain-to-CLN pathway may offer therapeutic opportunities to ameliorate systemic inflammation and brain injury after stroke.
Collapse
Affiliation(s)
- Elga Esposito
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Bum Ju Ahn
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Jingfei Shi
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA ,0000 0004 0369 153Xgrid.24696.3fChina-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yoshihiko Nakamura
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA ,0000 0004 0594 9821grid.411556.2Department of Emergency and Critical Care Medicine, Fukuoka University Hospital, Jonan, Fukuoka Japan
| | - Ji Hyun Park
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Emiri T. Mandeville
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Zhanyang Yu
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Su Jing Chan
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA ,0000 0001 2180 6431grid.4280.eDepartment of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Rakhi Desai
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Ayumi Hayakawa
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Xunming Ji
- 0000 0004 0369 153Xgrid.24696.3fChina-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Eng H. Lo
- 0000 0004 0386 9924grid.32224.35Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA USA
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
36
|
The wonders of BMP9: From mesenchymal stem cell differentiation, angiogenesis, neurogenesis, tumorigenesis, and metabolism to regenerative medicine. Genes Dis 2019; 6:201-223. [PMID: 32042861 PMCID: PMC6997590 DOI: 10.1016/j.gendis.2019.07.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/07/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Although bone morphogenetic proteins (BMPs) initially showed effective induction of ectopic bone growth in muscle, it has since been determined that these proteins, as members of the TGF-β superfamily, play a diverse and critical array of biological roles. These roles include regulating skeletal and bone formation, angiogenesis, and development and homeostasis of multiple organ systems. Disruptions of the members of the TGF-β/BMP superfamily result in severe skeletal and extra-skeletal irregularities, suggesting high therapeutic potential from understanding this family of BMP proteins. Although it was once one of the least characterized BMPs, BMP9 has revealed itself to have the highest osteogenic potential across numerous experiments both in vitro and in vivo, with recent studies suggesting that the exceptional potency of BMP9 may result from unique signaling pathways that differentiate it from other BMPs. The effectiveness of BMP9 in inducing bone formation was recently revealed in promising experiments that demonstrated efficacy in the repair of critical sized cranial defects as well as compatibility with bone-inducing bio-implants, revealing the great translational promise of BMP9. Furthermore, emerging evidence indicates that, besides its osteogenic activity, BMP9 exerts a broad range of biological functions, including stem cell differentiation, angiogenesis, neurogenesis, tumorigenesis, and metabolism. This review aims to summarize our current understanding of BMP9 across biology and the body.
Collapse
|
37
|
Akatsu Y, Takahashi N, Yoshimatsu Y, Kimuro S, Muramatsu T, Katsura A, Maishi N, Suzuki HI, Inazawa J, Hida K, Miyazono K, Watabe T. Fibroblast growth factor signals regulate transforming growth factor-β-induced endothelial-to-myofibroblast transition of tumor endothelial cells via Elk1. Mol Oncol 2019; 13:1706-1724. [PMID: 31094056 PMCID: PMC6670013 DOI: 10.1002/1878-0261.12504] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/31/2019] [Accepted: 05/14/2019] [Indexed: 02/04/2023] Open
Abstract
The tumor microenvironment contains various components, including cancer cells, tumor vessels, and cancer-associated fibroblasts, the latter of which are comprised of tumor-promoting myofibroblasts and tumor-suppressing fibroblasts. Multiple lines of evidence indicate that transforming growth factor-β (TGF-β) induces the formation of myofibroblasts and other types of mesenchymal (non-myofibroblastic) cells from endothelial cells. Recent reports show that fibroblast growth factor 2 (FGF2) modulates TGF-β-induced mesenchymal transition of endothelial cells, but the molecular mechanisms behind the signals that control transcriptional networks during the formation of different groups of fibroblasts remain largely unclear. Here, we studied the roles of FGF2 during the regulation of TGF-β-induced mesenchymal transition of tumor endothelial cells (TECs). We demonstrated that auto/paracrine FGF signals in TECs inhibit TGF-β-induced endothelial-to-myofibroblast transition (End-MyoT), leading to suppressed formation of contractile myofibroblast cells, but on the other hand can also collaborate with TGF-β in promoting the formation of active fibroblastic cells which have migratory and proliferative properties. FGF2 modulated TGF-β-induced formation of myofibroblastic and non-myofibroblastic cells from TECs via transcriptional regulation of various mesenchymal markers and growth factors. Furthermore, we observed that TECs treated with TGF-β were more competent in promoting in vivo tumor growth than TECs treated with TGF-β and FGF2. Mechanistically, we showed that Elk1 mediated FGF2-induced inhibition of End-MyoT via inhibition of TGF-β-induced transcriptional activation of α-smooth muscle actin promoter by myocardin-related transcription factor-A. Our data suggest that TGF-β and FGF2 oppose and cooperate with each other during the formation of myofibroblastic and non-myofibroblastic cells from TECs, which in turn determines the characteristics of mesenchymal cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Yuichi Akatsu
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan.,Biomedicine Group, Pharmaceutical Research Laboratories, Pharmaceutical Group, Nippon Kayaku Co., Ltd., Tokyo, Japan
| | - Naoya Takahashi
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Japan
| | - Yasuhiro Yoshimatsu
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Japan
| | - Shiori Kimuro
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Japan
| | - Tomoki Muramatsu
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Akihiro Katsura
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Nako Maishi
- Department of Vascular Biology and Molecular Pathology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi I Suzuki
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Johji Inazawa
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Japan
| |
Collapse
|
38
|
Tamura R, Yoshida K, Toda M. Current understanding of lymphatic vessels in the central nervous system. Neurosurg Rev 2019; 43:1055-1064. [PMID: 31209659 DOI: 10.1007/s10143-019-01133-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/29/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022]
Abstract
Lymphangiogenesis is associated with some pathological conditions such as inflammation, tissue repair, and tumor growth. Recently, a paradigm shift occurred following the discovery of meningeal lymphatic structures in the human central nervous system (CNS); these structures may be a key drainage route for cerebrospinal fluid (CSF) into the peripheral blood and may also contribute to inflammatory reaction and immune surveillance of the CNS. Lymphatic vessels located along the dural sinuses absorb CSF from the adjacent subarachnoid space and brain interstitial fluid via the glymphatic system, which is composed of aquaporin-4 water channels expressed on perivascular astrocytic end-feet membranes. Magnetic resonance imaging (MRI) clearly visualized these lymphatic vessels in the human dura mater. The conception of some neurological disorders, such as multiple sclerosis and Alzheimer's disease, has been changed by this paradigm shift. Meningeal lymphatic vessels could be a promising therapeutic target for the prevention of neurological disorders. However, the involvement of meningeal lymphatic vessels in the pathophysiology has not been fully elucidated and is the subject of future investigations. In this article, to understand the involvement of meningeal lymphatic vessels in neurological disorders, we review the differences between lymphangiogenesis in the CNS and in other tissues during both developmental and adulthood stages, and pathological conditions that may be associated with meningeal lymphatic vessels in the CNS.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
39
|
Liu R, Hu W, Li X, Pu D, Yang G, Liu H, Tan M, Zhu D. Association of circulating BMP9 with coronary heart disease and hypertension in Chinese populations. BMC Cardiovasc Disord 2019; 19:131. [PMID: 31146694 PMCID: PMC6543594 DOI: 10.1186/s12872-019-1095-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 05/03/2019] [Indexed: 12/11/2022] Open
Abstract
Background Bone morphogenetic protein9 (BMP9) has been reported to have a role in vascular development. However, there is still a lack of information regarding the association between circulating BMP9 levels and cardiovascular disease in humans. The goal of this study is to measure circulating BMP9 concentrations in patients with essential hypertension (HTN), coronary heart disease (CHD) and HTN + CHD, and evaluates the relationship between circulating BMP9 and these cardiovascular diseases. Methods A total of 417 individuals were recruited for this cross-sectional study from June 2015 to December 2017. These subjects were screened for HTN and CHD. Circulating BMP9 concentrations were measured by ELISA. Results Circulating BMP9 concentrations were significantly low in HTN, CHD and HTN + CHD individuals relative to those of the healthy individuals. Circulating BMP9 correlated negatively with SBP, FIns and HOMA-IR in HTN patients and correlated negatively with FBG and 2 h-BG in CHD patients. In both HTN and CHD patients, circulating BMP9 correlated negatively with BMI, WHR, FAT%, BP and TG. Multivariate logistic regression analysis showed that circulating BMP9 levels were associated with HTN, HTN + CHD and CHD. Individuals with low quartile of circulating BMP9 had a significantly high risk of HTN or/and CHD as compared with those in high quartile. Conclusions BMP9 is likely to be a biomarker for cardiovascular disease in humans, and it may play a role in the progression of cardiovascular disease. Trial registration ChiCTR-OPC-14005324. Electronic supplementary material The online version of this article (10.1186/s12872-019-1095-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Liu
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, China.,Department of Endocrinology, the Second Affiliated Hospital Chongqing Medical University, Chongqing, China
| | - Wenjing Hu
- Chongqing Prevention and Treatment Hospital for Occupational Diseases, Chongqing, China
| | - Xiaoqiang Li
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Danlan Pu
- Department of Endocrinology, Pepople's Hospital of Chongqing Banan District, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital Chongqing Medical University, Chongqing, China
| | - Hua Liu
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216-4505, USA
| | - Minghong Tan
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, China.
| | - Danping Zhu
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, China.
| |
Collapse
|
40
|
Interleukin-13 receptor α2 is a novel marker and potential therapeutic target for human melanoma. Sci Rep 2019; 9:1281. [PMID: 30718742 PMCID: PMC6362032 DOI: 10.1038/s41598-019-39018-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/31/2018] [Indexed: 12/23/2022] Open
Abstract
Malignant melanoma is one of the untreatable cancers in which conventional therapeutic strategies, including chemotherapy, are hardly effective. Therefore, identification of novel therapeutic targets involved in melanoma progression is urgently needed for developing effective therapeutic methods. Overexpression of interleukin-13 receptor α2 (IL13Rα2) is observed in several cancer types including glioma and pancreatic cancer. Although IL13Rα2 is implicated in the progression of various types of cancer, its expression and roles in the malignant melanoma have not yet been elucidated. In the present study, we showed that IL13Rα2 was expressed in approximately 7.5% melanoma patients. While IL13Rα2 expression in human melanoma cells decreased their proliferation in vitro, it promoted in vivo tumour growth and angiogenesis in melanoma xenograft mouse model. We also found that the expression of amphiregulin, a member of the epidermal growth factor (EGF) family, was correlated with IL13Rα2 expression in cultured melanoma cells, xenograft tumour tissues and melanoma clinical samples. Furthermore, expression of amphiregulin promoted tumour growth, implicating causal relationship between the expression of IL13Rα2 and amphiregulin. These results suggest that IL13Rα2 enhances tumorigenicity by inducing angiogenesis in malignant melanoma, and serves as a potential therapeutic target of malignant melanoma.
Collapse
|
41
|
Subileau M, Merdzhanova G, Ciais D, Collin-Faure V, Feige JJ, Bailly S, Vittet D. Bone Morphogenetic Protein 9 Regulates Early Lymphatic-Specified Endothelial Cell Expansion during Mouse Embryonic Stem Cell Differentiation. Stem Cell Reports 2018; 12:98-111. [PMID: 30595547 PMCID: PMC6335586 DOI: 10.1016/j.stemcr.2018.11.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 01/06/2023] Open
Abstract
Exogenous cues involved in the regulation of the initial steps of lymphatic endothelial development remain largely unknown. We have used an in vitro model based on the co-culture of vascular precursors derived from mouse embryonic stem cell (ESC) differentiation and OP9 stromal cells to examine the first steps of lymphatic specification and expansion. We found that bone morphogenetic protein 9 (BMP9) induced a dose-dependent biphasic effect on ESC-derived vascular precursors. At low concentrations, below 1 ng/mL, BMP9 expands the LYVE-1-positive lymphatic progeny and activates the calcineurin phosphatase/NFATc1 signaling pathway. In contrast, higher BMP9 concentrations preferentially enhance the formation of LYVE-1-negative endothelial cells. This effect results from an OP9 stromal cell-mediated VEGF-A secretion. RNA-silencing experiments indicate specific involvement of ALK1 and ALK2 receptors in these different BMP9 responses. BMP9 at low concentrations may be a useful tool to generate lymphatic endothelial cells from stem cells for cell-replacement strategies. Low doses of BMP9 raise lymph-vasculogenesis during ESC differentiation NFATc1 signaling operates in BMP9-induced lymphatic endothelial cell expansion High doses of BMP9 increase LYVE-1-negative endothelial cell formation A specific differential involvement of ALK1 and ALK2 mediates the BMP9 effects
Collapse
Affiliation(s)
- Mariela Subileau
- Univ. Grenoble Alpes, Inserm, CEA, BIG-BCI, Grenoble 38000, France
| | | | - Delphine Ciais
- Univ. Grenoble Alpes, Inserm, CEA, BIG-BCI, Grenoble 38000, France
| | | | | | - Sabine Bailly
- Univ. Grenoble Alpes, Inserm, CEA, BIG-BCI, Grenoble 38000, France
| | - Daniel Vittet
- Univ. Grenoble Alpes, Inserm, CEA, BIG-BCI, Grenoble 38000, France.
| |
Collapse
|
42
|
Wang S, Zhou H, Wu D, Ni H, Chen Z, Chen C, Xiang Y, Dai K, Chen X, Li X. MicroRNA let-7a regulates angiogenesis by targeting TGFBR3 mRNA. J Cell Mol Med 2018; 23:556-567. [PMID: 30467960 PMCID: PMC6307798 DOI: 10.1111/jcmm.13960] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/16/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis has a great impact on human health, owing to its participation in development, wound healing and the pathogenesis of several diseases. It has been reported that let-7a is a tumour suppressor, but whether it plays a role in angiogenesis is unclear. Here we showed that let-7a, a microRNA conserved in vertebrates, regulated angiogenesis by concomitantly down-regulating TGFBR3. Overexpression of let-7a or knockdown of TGFBR3 in cell culture inhibited the tube formation and reduced migration rate. Moreover, xenograft experiments showed that overexpression of let-7a or knockdown of TGFBR3 had smaller tumour size. Downstream genes, such as VEGFC and MMP9, were also down-regulated in let-7a overexpression or TGFBR3 knockdown groups. Therefore, our results revealed a novel mechanism that let-7a regulate angiogenesis through post-transcriptional regulation of TGFBR3.
Collapse
Affiliation(s)
- Shao Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huandong Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dazhou Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huajing Ni
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongliang Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youqun Xiang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kezhi Dai
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoming Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Li
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Wu J, Jackson-Weaver O, Xu J. The TGFβ superfamily in cardiac dysfunction. Acta Biochim Biophys Sin (Shanghai) 2018; 50:323-335. [PMID: 29462261 DOI: 10.1093/abbs/gmy007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Indexed: 12/23/2022] Open
Abstract
TGFβ superfamily includes the transforming growth factor βs (TGFβs), bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs) and Activin/Inhibin families of ligands. Among the 33 members of TGFβ superfamily ligands, many act on multiple types of cells within the heart, including cardiomyocytes, cardiac fibroblasts/myofibroblasts, coronary endothelial cells, smooth muscle cells, and immune cells (e.g. monocytes/macrophages and neutrophils). In this review, we highlight recent discoveries on TGFβs, BMPs, and GDFs in different cardiac residential cellular components, in association with functional impacts in heart development, injury repair, and dysfunction. Specifically, we will review the roles of TGFβs, BMPs, and GDFs in cardiac hypertrophy, fibrosis, contractility, metabolism, angiogenesis, and regeneration.
Collapse
Affiliation(s)
- Jian Wu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Olan Jackson-Weaver
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Jian Xu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
44
|
Zhang L, Ye Y, Long X, Xiao P, Ren X, Yu J. BMP signaling and its paradoxical effects in tumorigenesis and dissemination. Oncotarget 2018; 7:78206-78218. [PMID: 27661009 PMCID: PMC5363655 DOI: 10.18632/oncotarget.12151] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/14/2016] [Indexed: 01/04/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) play important roles in embryonic and postnatal development by regulating cell differentiation, proliferation, motility, and survival, thus maintaining homeostasis during organ and tissue development. BMPs can lead to tumorigenesis and regulate cancer progression in different stages. Therefore, we summarized studies on BMP expression, the clinical significance of BMP dysfunction in various cancer types, and the molecular regulation of various BMP-related signaling pathways. We emphasized on the paradoxical effects of BMPs on various aspects of carcinogenesis, including epithelial–mesenchymal transition (EMT), cancer stem cells (CSCs), and angiogenesis. We also reviewed the molecular mechanisms by which BMPs regulate tumor generation and progression as well as potential therapeutic targets against BMPs that might be valuable in preventing tumor growth and invasion.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Yingnan Ye
- Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Xinxin Long
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Pei Xiao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jinpu Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China.,Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| |
Collapse
|
45
|
Hanna DL, Loupakis F, Yang D, Cremolini C, Schirripa M, Li M, Matsusaka S, Berger MD, Miyamoto Y, Zhang W, Ning Y, Antoniotti C, Salvatore L, Moran M, Zeger G, Astrow SH, Falcone A, Lenz HJ. Prognostic Value of ACVRL1 Expression in Metastatic Colorectal Cancer Patients Receiving First-line Chemotherapy With Bevacizumab: Results From the Triplet Plus Bevacizumab (TRIBE) Study. Clin Colorectal Cancer 2018; 17:e471-e488. [PMID: 29636300 DOI: 10.1016/j.clcc.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND No biomarkers exist to predict benefit from antiangiogenic therapy in metastatic colorectal cancer patients. ACVRL1 (activin receptor like-protein 1) encodes for ALK1, a member of the transforming growth factor-β receptor family, which directs pathologic angiogenesis. We examined the intratumoral expression of ACVRL1 and other angiogenesis pathway-related genes to identify molecular markers in the TRIBE study. MATERIALS AND METHODS Of 503 randomized patients, 228 had sufficient tissue for analysis. Formalin-fixed paraffin-embedded specimens were examined for expression of VEGF-A, VEGF-B, VEGF-C, VEGFR1, VEGFR2, ACVRL1, EphB4, and EGFL7 using reverse transcription polymerase chain reaction. A maximal χ2 approach was used to determine the messenger RNA levels associated with progression-free survival (PFS), overall survival (OS), response rate, early tumor shrinkage, and depth of response. Recursive partitioning trees were constructed to identify composite prognostic biomarker profiles. External validation was conducted in silico using the Oncomine database. RESULTS High ACVRL1 expression was associated with superior OS in both treatment arms (FOLFOXIRI [5-fluorouracil, leucovorin, oxaliplatin, irinotecan]-bevacizumab, 32.7 vs. 13.5 months, hazard ratio [HR], 0.38, P = .023; FOLFIRI [5-fluorouracil, leucovorin, irinotecan]-bevacizumab, 35.1 vs. 22.0 months, HR, 0.36, P = .006) and prolonged PFS (11.7 vs. 5.9 months, multivariate HR, 0.17; P = .001) for patients receiving FOLFOXIRI-bevacizumab on univariate and multivariate analyses. In recursive partitioning analysis, ACVRL1 was the strongest discriminator of the response rate, PFS, and OS in patients receiving FOLFOXIRI-bevacizumab and of OS in patients receiving FOLFIRI-bevacizumab. In silico validation revealed significant associations between ACVRL1 expression, disease recurrence, and 1-year survival (P < .05) among all colorectal cancer stages. CONCLUSION ACVRL1 expression could serve as a prognostic biomarker in metastatic colorectal cancer patients receiving chemotherapy and bevacizumab and warrants further evaluation in prospective studies.
Collapse
Affiliation(s)
- Diana L Hanna
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA; Hoag Family Cancer Institute, Newport Beach, CA
| | - Fotios Loupakis
- Unito of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Dongyun Yang
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| | - Chiara Cremolini
- Unito of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Marta Schirripa
- Unito of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Meng Li
- Health Sciences Bioinformatics Core, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Satoshi Matsusaka
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| | - Martin D Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| | - Yuji Miyamoto
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| | - Yan Ning
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| | - Carlotta Antoniotti
- Unito of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Lisa Salvatore
- Unito of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | | | | | | | - Alfredo Falcone
- Unito of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.
| |
Collapse
|
46
|
Eleftheriou NM, Sjölund J, Bocci M, Cortez E, Lee SJ, Cunha SI, Pietras K. Compound genetically engineered mouse models of cancer reveal dual targeting of ALK1 and endoglin as a synergistic opportunity to impinge on angiogenic TGF-β signaling. Oncotarget 2018; 7:84314-84325. [PMID: 27741515 PMCID: PMC5341292 DOI: 10.18632/oncotarget.12604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/03/2016] [Indexed: 01/21/2023] Open
Abstract
Angiogenesis occurs early in tumor development, sustains primary tumor growth and provides a route for metastatic escape. The TGF-β family receptors modulate angiogenesis via endothelial-cell specific pathways. Here we investigate the interaction of two such receptors, ALK1 and endoglin, in pancreatic neuroendocrine tumors (PanNET). Independently, ALK1 and endoglin deficiencies exhibited genetically divergent phenotypes, while both highly correlate to an endothelial metagene in human and mouse PanNETs. A concurrent deficiency of both receptors synergistically decreased tumor burden to a greater extent than either individual knockdown. Furthermore, the knockout of Gdf2 (BMP9), the primary ligand for ALK1 and endoglin, exhibited a mixed phenotype from each of ALK1 and endoglin deficiencies; overall primary tumor burden decreased, but hepatic metastases increased. Tumors lacking BMP9 display a hyperbranching vasculature, and an increase in vascular mesenchymal-marker expression, which may be implicit in the increase in metastases. Taken together, our work cautions against singular blockade of BMP9 and instead demonstrates the utility of dual blockade of ALK1 and endoglin as a strategy for anti-angiogenic therapy in PanNET.
Collapse
Affiliation(s)
- Nikolas M Eleftheriou
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Matteo Bocci
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Eliane Cortez
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Se-Jin Lee
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara I Cunha
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| |
Collapse
|
47
|
Jung JW, Yoon SM, Kim S, Jeon YH, Yoon BH, Yang SG, Kim MK, Choe S, Kuo MMC. Bone morphogenetic protein-9 is a potent growth inhibitor of hepatocellular carcinoma and reduces the liver cancer stem cells population. Oncotarget 2018; 7:73754-73768. [PMID: 27650540 PMCID: PMC5342011 DOI: 10.18632/oncotarget.12062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/02/2016] [Indexed: 12/25/2022] Open
Abstract
The biological role of BMP-9 signaling in liver cancer remains dubious. To explore the potential use of BMP-9 signaling for anti-cancer therapy, we used recombinant human BMP-9, which we referred to as MB109, to study the effect on growth of fifteen hepatocellular carcinoma (HCC) cell lines. MB109 effectively inhibits the proliferation of nine HCC cells in vitro. The anti-proliferative effect was found to be induced by turning on p21 signaling, which caused survivin suppression and G0/G1 cell cycle arrest. ID3 was identified to be the mediator of the MB109-induced p21 expression. Blocking the activity of p38 MAPK diminished ID3 and p21 expression, indicating that MB109 signals through a p38 MAPK/ID3/p21 pathway to arrest cell cycle progression. Moreover, prolonged MB109 treatment suppressed the expression of five prominent liver cancer stem cell (LCSC) markers, including CD44, CD90, AFP, GPC3 and ANPEP. Xenograft model confirmed the anti-tumor and LCSC-suppression capability of MB109 in vivo. Contrary to ongoing efforts of suppressing BMP-9 signaling to inhibit angiogenesis of cancer tissue, these results demonstrate an unexpected therapeutic potential of MB109 to stimulate BMP-9 signaling for anti-cancer therapies.
Collapse
Affiliation(s)
- Jae Woo Jung
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea.,Current address: Interdisciplinary Program in Genetic Engineering, Seoul National University, Seoul 151-742, South Korea
| | - So-Mi Yoon
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea
| | - Subin Kim
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea
| | - Yun-Hui Jeon
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea
| | - Byung-Hak Yoon
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea
| | - Su-Geun Yang
- Department of New Drug Development, School of Medicine, Inha University, Incheon 400-712, South Korea
| | - Min Kyoung Kim
- Department of New Drug Development, School of Medicine, Inha University, Incheon 400-712, South Korea
| | - Senyon Choe
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea.,Drug Discovery Collaboratory, University of California, San Diego, La Jolla, CA 92037, United States of America
| | - Mario Meng-Chiang Kuo
- Protein Engineering Laboratory, Joint Center for Biosciences, Songdo Smart Valley, Yeonsu-gu, Incheon 406-840, South Korea.,Drug Discovery Collaboratory, University of California, San Diego, La Jolla, CA 92037, United States of America.,Current address: Polaris Pharmaceuticals, Inc., San Diego, CA 92121, United States of America
| |
Collapse
|
48
|
Circulating bone morphogenetic protein-9 levels are associated with hypertension and insulin resistance in humans. ACTA ACUST UNITED AC 2018; 12:372-380. [PMID: 29550458 DOI: 10.1016/j.jash.2018.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 02/02/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022]
Abstract
It has been demonstrated that bone morphogenetic protein-9 (BMP-9) may have an important role in vascular development and stability. However, the association of circulating BMP-9 with essential hypertension (HTN) has not been established in humans. The objective of this study is to observe the changes of circulating BMP-9 levels in patients with HTN and to investigate the association of circulation BMP-9 and insulin resistance (IR) in a cross-sectional study. Two hundred twenty-five individuals, including 132 patients with hypertension, and 93 healthy controls, were included in the present study. Circulating BMP-9 concentrations were measured with an ELISA kit. The association of circulating BMP-9 with other parameters was analyzed. When compared with healthy subjects, circulating BMP-9 concentrations were markedly lower in HTN patients (46.20 [31.85-62.80] vs. 77.21 [39.33-189.15], P < .01) and correlated negatively with blood pressure and the homeostasis model assessment of insulin resistance (P < .05 or P < .01). Decreasing levels of BMP-9 were independently and markedly related to HTN. In a multiple linear regression analysis, only systolic blood pressure and free fatty acid concentrations were independently associated with circulating BMP-9. Our findings suggest that BMP-9 may be a serum biomarker for HTN and IR.
Collapse
|
49
|
Goumans MJ, Ten Dijke P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022210. [PMID: 28348036 DOI: 10.1101/cshperspect.a022210] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic studies in animals and humans indicate that gene mutations that functionally perturb transforming growth factor β (TGF-β) signaling are linked to specific hereditary vascular syndromes, including Osler-Rendu-Weber disease or hereditary hemorrhagic telangiectasia and Marfan syndrome. Disturbed TGF-β signaling can also cause nonhereditary disorders like atherosclerosis and cardiac fibrosis. Accordingly, cell culture studies using endothelial cells or smooth muscle cells (SMCs), cultured alone or together in two- or three-dimensional cell culture assays, on plastic or embedded in matrix, have shown that TGF-β has a pivotal effect on endothelial and SMC proliferation, differentiation, migration, tube formation, and sprouting. Moreover, TGF-β can stimulate endothelial-to-mesenchymal transition, a process shown to be of key importance in heart valve cushion formation and in various pathological vascular processes. Here, we discuss the roles of TGF-β in vasculogenesis, angiogenesis, and lymphangiogenesis and the deregulation of TGF-β signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
50
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|