51
|
Akwii RG, Mikelis CM. Targeting the Angiopoietin/Tie Pathway: Prospects for Treatment of Retinal and Respiratory Disorders. Drugs 2021; 81:1731-1749. [PMID: 34586603 PMCID: PMC8479497 DOI: 10.1007/s40265-021-01605-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2021] [Indexed: 12/21/2022]
Abstract
Anti-angiogenic approaches have significantly advanced the treatment of vascular-related pathologies. The ephemeral outcome and known side effects of the current vascular endothelial growth factor (VEGF)-based anti-angiogenic treatments have intensified research on other growth factors. The angiopoietin/Tie (Ang/Tie) family has an established role in vascular physiology and regulates angiogenesis, vascular permeability, and inflammatory responses. The Ang/Tie family consists of angiopoietins 1-4, their receptors, tie1 and 2 and the vascular endothelial-protein tyrosine phosphatase (VE-PTP). Modulation of Tie2 activation has provided a promising outcome in preclinical models and has led to clinical trials of Ang/Tie-targeting drug candidates for retinal disorders. Although less is known about the role of Ang/Tie in pulmonary disorders, several studies have revealed great potential of the Ang/Tie family members as drug targets for pulmonary vascular disorders as well. In this review, we summarize the functions of the Ang/Tie pathway in retinal and pulmonary vascular physiology and relevant disorders and highlight promising drug candidates targeting this pathway currently being or expected to be under clinical evaluation for retinal and pulmonary vascular disorders.
Collapse
Affiliation(s)
- Racheal Grace Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1406 S. Coulter St., Amarillo, TX, 79106, USA
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1406 S. Coulter St., Amarillo, TX, 79106, USA.
| |
Collapse
|
52
|
New mechanism-based approaches to treating and evaluating the vasculopathy of scleroderma. Curr Opin Rheumatol 2021; 33:471-479. [PMID: 34402454 DOI: 10.1097/bor.0000000000000830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Utilizing recent insight into the vasculopathy of scleroderma (SSc), the review will highlight new opportunities for evaluating and treating the disease by promoting stabilization and protection of the microvasculature. RECENT FINDINGS Endothelial junctional signaling initiated by vascular endothelial-cadherin (VE-cadherin) and Tie2 receptors, which are fundamental to promoting vascular health and stability, are disrupted in SSc. This would be expected to not only diminish their protective activity, but also increase pathological processes that are normally restrained by these signaling mediators, resulting in pathological changes in vascular function and structure. Indeed, key features of SSc vasculopathy, from the earliest signs of edema and puffy fingers to pathological disruption of hemodynamics, nutritional blood flow, capillary structure and angiogenesis are all consistent with this altered endothelial signaling. It also likely contributes to further progression of the disease including tissue fibrosis, and organ and tissue injury. SUMMARY Restoring protective endothelial junctional signaling should combat the vasculopathy of SSc and prevent further deterioration in vascular and organ function. Indeed, this type of targeted approach has achieved remarkable results in preclinical models for other diseases. Furthermore, tracking this endothelial junctional signaling, for example by assessing vascular permeability, should facilitate insight into disease progression and its response to therapy.
Collapse
|
53
|
Cordes S, Mokhtari Z, Bartosova M, Mertlitz S, Riesner K, Shi Y, Mengwasser J, Kalupa M, McGeary A, Schleifenbaum J, Schrezenmeier J, Bullinger L, Diaz-Ricart M, Palomo M, Carrreras E, Beutel G, Schmitt CP, Beilhack A, Penack O. Endothelial damage and dysfunction in acute graft-versus-host disease. Haematologica 2021; 106:2147-2160. [PMID: 32675225 PMCID: PMC8327719 DOI: 10.3324/haematol.2020.253716] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Clinical studies suggested that endothelial dysfunction and damage could be involved in the development and severity of acute graft-versus-host disease (aGVHD). Accordingly, we found increased percentage of apoptotic Casp3+ blood vessels in duodenal and colonic mucosa biopsies of patients with severe aGVHD. In murine experimental aGVHD, we detected severe microstructural endothelial damage and reduced endothelial pericyte coverage accompanied by reduced expression of endothelial tight junction proteins leading to increased endothelial leakage in aGVHD target organs. During intestinal aGVHD, colonic vasculature structurally changed, reflected by increased vessel branching and vessel diameter. Because recent data demonstrated an association of endothelium-related factors and steroid refractory aGVHD (SR-aGVHD), we analyzed human biopsies and murine tissues from SR-aGVHD. We found extensive tissue damage but low levels of alloreactive T cell infiltration in target organs, providing the rationale for T-cell independent SR-aGVHD treatment strategies. Consequently, we tested the endothelium-protective PDE5 inhibitor sildenafil, which reduced apoptosis and improved metabolic activity of endothelial cells in vitro. Accordingly, sildenafil treatment improved survival and reduced target organ damage during experimental SR-aGVHD. Our results demonstrate extensive damage, structural changes, and dysfunction of the vasculature during aGVHD. Therapeutic intervention by endothelium-protecting agents is an attractive approach for SR-aGVHD complementing current anti-inflammatory treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Shi
- Charité Universitätsmedizin Berlin
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
De novo generation of macrophage from placenta-derived hemogenic endothelium. Dev Cell 2021; 56:2121-2133.e6. [PMID: 34197725 DOI: 10.1016/j.devcel.2021.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 01/31/2023]
Abstract
Macrophages play pivotal roles in immunity, hematopoiesis, and tissue homeostasis. In mammals, macrophages have been shown to originate from yolk-sac-derived erythro-myeloid progenitors and aorta-gonad-mesonephros (AGM)-derived hematopoietic stem cells. However, whether macrophages can arise from other embryonic sites remains unclear. Here, using single-cell RNA sequencing, we profile the transcriptional landscape of mouse fetal placental hematopoiesis. We uncover and experimentally validate that a CD44+ subpopulation of placental endothelial cells (ECs) exhibits hemogenic potential. Importantly, lineage tracing using the newly generated Hoxa13 reporter line shows that Hoxa13-labeled ECs can produce placental macrophages, named Hofbauer cell (HBC)-like cells. Furthermore, we identify two subtypes of HBC-like cells, and cell-cell interaction analysis identifies their potential roles in angiogenesis and antigen presentation, separately. Our study provides a comprehensive understanding of placental hematopoiesis and highlights the placenta as a source of macrophages, which has important implications for both basic and translational research.
Collapse
|
55
|
Villalba N, Baby S, Yuan SY. The Endothelial Glycocalyx as a Double-Edged Sword in Microvascular Homeostasis and Pathogenesis. Front Cell Dev Biol 2021; 9:711003. [PMID: 34336864 PMCID: PMC8316827 DOI: 10.3389/fcell.2021.711003] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/22/2021] [Indexed: 12/27/2022] Open
Abstract
Expressed on the endothelial cell (EC) surface of blood vessels, the glycocalyx (GCX), a mixture of carbohydrates attached to proteins, regulates the access of cells and molecules in the blood to the endothelium. Besides protecting endothelial barrier integrity, the dynamic microstructure of the GCX confers remarkable functions including mechanotransduction and control of vascular tone. Recently, a novel perspective has emerged supporting the pleiotropic roles of the endothelial GCX (eGCX) in cardiovascular health and disease. Because eGCX degradation occurs in certain pathological states, the circulating levels of eGCX degradation products have been recognized to have diagnostic or prognostic values. Beyond their biomarker roles, certain eGCX fragments serve as pathogenic factors in disease progression. Pharmacological interventions that attenuate eGCX degradation or restore its integrity have been sought. This review provides our current understanding of eGCX structure and function across the microvasculature in different organs. We also discuss disease or injury states, such as infection, sepsis and trauma, where eGCX dysfunction contributes to severe inflammatory vasculopathy.
Collapse
Affiliation(s)
- Nuria Villalba
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sheon Baby
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
56
|
Sanwal R, Joshi K, Ditmans M, Tsai SSH, Lee WL. Ultrasound and Microbubbles for Targeted Drug Delivery to the Lung Endothelium in ARDS: Cellular Mechanisms and Therapeutic Opportunities. Biomedicines 2021; 9:biomedicines9070803. [PMID: 34356867 PMCID: PMC8301318 DOI: 10.3390/biomedicines9070803] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by increased permeability of the alveolar–capillary membrane, a thin barrier composed of adjacent monolayers of alveolar epithelial and lung microvascular endothelial cells. This results in pulmonary edema and severe hypoxemia and is a common cause of death after both viral (e.g., SARS-CoV-2) and bacterial pneumonia. The involvement of the lung in ARDS is notoriously heterogeneous, with consolidated and edematous lung abutting aerated, less injured regions. This makes treatment difficult, as most therapeutic approaches preferentially affect the normal lung regions or are distributed indiscriminately to other organs. In this review, we describe the use of thoracic ultrasound and microbubbles (USMB) to deliver therapeutic cargo (drugs, genes) preferentially to severely injured areas of the lung and in particular to the lung endothelium. While USMB has been explored in other organs, it has been under-appreciated in the treatment of lung injury since ultrasound energy is scattered by air. However, this limitation can be harnessed to direct therapy specifically to severely injured lungs. We explore the cellular mechanisms governing USMB and describe various permutations of cargo administration. Lastly, we discuss both the challenges and potential opportunities presented by USMB in the lung as a tool for both therapy and research.
Collapse
Affiliation(s)
- Rajiv Sanwal
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kushal Joshi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Mihails Ditmans
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Scott S. H. Tsai
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence: ; Tel.: +416-864-6060 (ext. 77655)
| |
Collapse
|
57
|
Khan KA, Wu FTH, Cruz‐Munoz W, Kerbel RS. Ang2 inhibitors and Tie2 activators: potential therapeutics in perioperative treatment of early stage cancer. EMBO Mol Med 2021; 13:e08253. [PMID: 34125494 PMCID: PMC8261516 DOI: 10.15252/emmm.201708253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic drugs targeting the VEGF pathway are most effective in advanced metastatic disease settings of certain types of cancers, whereas they have been unsuccessful as adjuvant therapies of micrometastatic disease in numerous phase III trials involving early-stage (resectable) cancers. Newer investigational anti-angiogenic drugs have been designed to inhibit the Angiopoietin (Ang)-Tie pathway. Acting through Tie2 receptors, the Ang1 ligand is a gatekeeper of endothelial quiescence. Ang2 is a dynamically expressed pro-angiogenic destabilizer of endothelium, and its upregulation is associated with poor prognosis in cancer. Besides using Ang2 blockers as inhibitors of tumor angiogenesis, little attention has been paid to their use as stabilizers of blood vessels to suppress tumor cell extravasation and metastasis. In clinical trials, Ang2 blockers have shown limited efficacy in advanced metastatic disease settings. This review summarizes preclinical evidence suggesting the potential utility of Ang2 inhibitors or Tie2 activators as neoadjuvant or adjuvant therapies in the prevention or treatment of early-stage micrometastatic disease. We further discuss the rationale and potential of combining these strategies with immunotherapy, including immune checkpoint targeting antibodies.
Collapse
Affiliation(s)
- Kabir A Khan
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Florence TH Wu
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - William Cruz‐Munoz
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Robert S Kerbel
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| |
Collapse
|
58
|
Bhatraju PK, Morrell ED, Zelnick L, Sathe NA, Chai XY, Sakr SS, Sahi SK, Sader A, Lum DM, Liu T, Koetje N, Garay A, Barnes E, Lawson J, Cromer G, Bray MK, Pipavath S, Kestenbaum BR, Liles WC, Fink SL, West TE, Evans L, Mikacenic C, Wurfel MM. Comparison of host endothelial, epithelial and inflammatory response in ICU patients with and without COVID-19: a prospective observational cohort study. Crit Care 2021; 25:148. [PMID: 33874973 PMCID: PMC8054255 DOI: 10.1186/s13054-021-03547-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/22/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Analyses of blood biomarkers involved in the host response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral infection can reveal distinct biological pathways and inform development and testing of therapeutics for COVID-19. Our objective was to evaluate host endothelial, epithelial and inflammatory biomarkers in COVID-19. METHODS We prospectively enrolled 171 ICU patients, including 78 (46%) patients positive and 93 (54%) negative for SARS-CoV-2 infection from April to September, 2020. We compared 22 plasma biomarkers in blood collected within 24 h and 3 days after ICU admission. RESULTS In critically ill COVID-19 and non-COVID-19 patients, the most common ICU admission diagnoses were respiratory failure or pneumonia, followed by sepsis and other diagnoses. Similar proportions of patients in both groups received invasive mechanical ventilation at the time of study enrollment. COVID-19 and non-COVID-19 patients had similar rates of acute respiratory distress syndrome, severe acute kidney injury, and in-hospital mortality. While concentrations of interleukin 6 and 8 were not different between groups, markers of epithelial cell injury (soluble receptor for advanced glycation end products, sRAGE) and acute phase proteins (serum amyloid A, SAA) were significantly higher in COVID-19 compared to non-COVID-19, adjusting for demographics and APACHE III scores. In contrast, angiopoietin 2:1 (Ang-2:1 ratio) and soluble tumor necrosis factor receptor 1 (sTNFR-1), markers of endothelial dysfunction and inflammation, were significantly lower in COVID-19 (p < 0.002). Ang-2:1 ratio and SAA were associated with mortality only in non-COVID-19 patients. CONCLUSIONS These studies demonstrate that, unlike other well-studied causes of critical illness, endothelial dysfunction may not be characteristic of severe COVID-19 early after ICU admission. Pathways resulting in elaboration of acute phase proteins and inducing epithelial cell injury may be promising targets for therapeutics in COVID-19.
Collapse
Affiliation(s)
- Pavan K Bhatraju
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA.
- Sepsis Center of Research Excellence - University of Washington (SCORE-UW), Seattle, WA, USA.
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, USA.
| | - Eric D Morrell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Leila Zelnick
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, USA
| | - Neha A Sathe
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Xin-Ya Chai
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Sana S Sakr
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Sharon K Sahi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Anthony Sader
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Dawn M Lum
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, USA
| | - Ted Liu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Neall Koetje
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Ashley Garay
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Elizabeth Barnes
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Jonathan Lawson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Gail Cromer
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Mary K Bray
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | | | - Bryan R Kestenbaum
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, USA
| | - W Conrad Liles
- Sepsis Center of Research Excellence - University of Washington (SCORE-UW), Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - T Eoin West
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Laura Evans
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Carmen Mikacenic
- Translational Research, Benaroya Research Institute, Seattle, WA, USA
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA, 98104, USA
- Sepsis Center of Research Excellence - University of Washington (SCORE-UW), Seattle, WA, USA
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, USA
| |
Collapse
|
59
|
Oshima K, King SI, McMurtry SA, Schmidt EP. Endothelial Heparan Sulfate Proteoglycans in Sepsis: The Role of the Glycocalyx. Semin Thromb Hemost 2021; 47:274-282. [PMID: 33794552 DOI: 10.1055/s-0041-1725064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is increasing recognition of the importance of the endothelial glycocalyx and its in vivo manifestation, the endothelial surface layer, in vascular homeostasis. Heparan sulfate proteoglycans (HSPGs) are a major structural constituent of the endothelial glycocalyx and serve to regulate vascular permeability, microcirculatory tone, leukocyte and platelet adhesion, and hemostasis. During sepsis, endothelial HSPGs are shed through the induction of "sheddases" such as heparanase and matrix metalloproteinases, leading to loss of glycocalyx integrity and consequent vascular dysfunction. Less well recognized is that glycocalyx degradation releases HSPG fragments into the circulation, which can shape the systemic consequences of sepsis. In this review, we will discuss (1) the normal, homeostatic functions of HSPGs within the endothelial glycocalyx, (2) the pathological changes in HSPGs during sepsis and their consequences on the local vascular bed, and (3) the systemic consequences of HSPG degradation. In doing so, we will identify potential therapeutic targets to improve vascular function during sepsis as well as highlight key areas of uncertainty that require further mechanistic investigation.
Collapse
Affiliation(s)
- Kaori Oshima
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Samantha I King
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sarah A McMurtry
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Eric P Schmidt
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Medicine, Denver Health Medical Center, Denver, Colorado
| |
Collapse
|
60
|
Li Y, Suo L, Fu Z, Li G, Zhang J. Pivotal role of endothelial cell autophagy in sepsis. Life Sci 2021; 276:119413. [PMID: 33794256 DOI: 10.1016/j.lfs.2021.119413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
Sepsis is a fatal organ dysfunction resulting from a disordered host response to infection. Endothelial cells (ECs) are usually the primary targets of inflammatory mediators in sepsis; damage to ECs plays a pivotal part in vital organ failure. In recent studies, autophagy was suggested to play a critical role in the ECs injury although the mechanisms by which ECs are injured in sepsis are not well elucidated. Autophagy is a highly conserved catabolic process that includes sequestrating plasma contents and transporting cargo to lysosomes for recycling the vital substrates required for metabolism. This pathway also counteracts microbial invasion to balance and retain homeostasis, especially during sepsis. Increasing evidence indicates that autophagy is closely associated with endothelial function. The role of autophagy in sepsis may or may not be favorable depending upon conditions. In the present review, the current knowledge of autophagy in the process of sepsis and its influence on ECs was evaluated. In addition, the potential of targeting EC autophagy for clinical treatment of sepsis was discussed.
Collapse
Affiliation(s)
- Yuexian Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, PR China
| | - Liangyuan Suo
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shengjing Hospital of China Medical University, No. 44 Xiaoheyan Road, Shengyang, Liaoning 110042, PR China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, PR China
| | - Guoqing Li
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning 116001, PR China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|
61
|
Grant ZL, Whitehead L, Wong VH, He Z, Yan RY, Miles AR, Benest AV, Bates DO, Prahst C, Bentley K, Bui BV, Symons RC, Coultas L. Blocking endothelial apoptosis revascularizes the retina in a model of ischemic retinopathy. J Clin Invest 2021; 130:4235-4251. [PMID: 32427589 PMCID: PMC7410052 DOI: 10.1172/jci127668] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
Aberrant, neovascular retinal blood vessel growth is a vision-threatening complication in ischemic retinal diseases. It is driven by retinal hypoxia frequently caused by capillary nonperfusion and endothelial cell (EC) loss. We investigated the role of EC apoptosis in this process using a mouse model of ischemic retinopathy, in which vessel closure and EC apoptosis cause capillary regression and retinal ischemia followed by neovascularization. Protecting ECs from apoptosis in this model did not prevent capillary closure or retinal ischemia. Nonetheless, it prevented the clearance of ECs from closed capillaries, delaying vessel regression and allowing ECs to persist in clusters throughout the ischemic zone. In response to hypoxia, these preserved ECs underwent a vessel sprouting response and rapidly reassembled into a functional vascular network. This alleviated retinal hypoxia, preventing subsequent pathogenic neovascularization. Vessel reassembly was not limited by VEGFA neutralization, suggesting it was not dependent on the excess VEGFA produced by the ischemic retina. Neutralization of ANG2 did not prevent vessel reassembly, but did impair subsequent angiogenic expansion of the reassembled vessels. Blockade of EC apoptosis may promote ischemic tissue revascularization by preserving ECs within ischemic tissue that retain the capacity to reassemble a functional network and rapidly restore blood supply.
Collapse
Affiliation(s)
- Zoe L Grant
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, and
| | - Lachlan Whitehead
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, and
| | - Vickie Hy Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Richard Y Yan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Abigail R Miles
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Andrew V Benest
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, School of Medicine, University of Nottingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Birmingham, United Kingdom
| | - David O Bates
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, School of Medicine, University of Nottingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Birmingham, United Kingdom
| | - Claudia Prahst
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Katie Bentley
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA.,Beijer Laboratory for Gene and Neuroscience Research, Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Robert Ca Symons
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia.,Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Department of Ophthalmology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Leigh Coultas
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, and
| |
Collapse
|
62
|
Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res 2021; 117:60-73. [PMID: 32215570 PMCID: PMC7810126 DOI: 10.1093/cvr/cvaa070] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Sepsis accounts for nearly 700 000 deaths in Europe annually and is caused by an overwhelming host response to infection resulting in organ failure. The endothelium is an active contributor to sepsis and as such represents a major target for therapy. During sepsis, endothelial cells amplify the immune response and activate the coagulation system. They are both a target and source of inflammation and serve as a link between local and systemic immune responses. In response to cytokines produced by immune cells, the endothelium expresses adhesion molecules and produces vasoactive compounds, inflammatory cytokines, and chemoattractants, thus switching from an anticoagulant to procoagulant state. These responses contribute to local control of infection, but systemic activation can lead to microvascular thrombosis, capillary permeability, hypotension, tissue hypoxia, and ultimately tissue damage. This review focuses on the role of the endothelium in leucocyte adhesion and transmigration as well as production of reactive oxygen and nitrogen species, microRNAs and cytokines, formation of signalling microparticles, and disseminated intravascular coagulation. We also discuss alterations in endothelial permeability and apoptosis. Finally, we review the diagnostic potential of endothelial markers and endothelial pathways as therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Elena V Dolmatova
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Keke Wang
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Rohan Mandavilli
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| |
Collapse
|
63
|
Li Z, Korhonen EA, Merlini A, Strauss J, Wihuri E, Nurmi H, Antila S, Paech J, Deutsch U, Engelhardt B, Chintharlapalli S, Koh GY, Flügel A, Alitalo K. Angiopoietin-2 blockade ameliorates autoimmune neuroinflammation by inhibiting leukocyte recruitment into the CNS. J Clin Invest 2020; 130:1977-1990. [PMID: 32149735 PMCID: PMC7108925 DOI: 10.1172/jci130308] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 01/03/2020] [Indexed: 12/12/2022] Open
Abstract
Angiopoietin-2 (Ang2), a ligand of the endothelial Tie2 tyrosine kinase, is involved in vascular inflammation and leakage in critically ill patients. However, the role of Ang2 in demyelinating central nervous system (CNS) autoimmune diseases is unknown. Here, we report that Ang2 is critically involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a rodent model of multiple sclerosis. Ang2 expression was induced in CNS autoimmunity, and transgenic mice overexpressing Ang2 specifically in endothelial cells (ECs) developed a significantly more severe EAE. In contrast, treatment with Ang2-blocking Abs ameliorated neuroinflammation and decreased spinal cord demyelination and leukocyte infiltration into the CNS. Similarly, Ang2-binding and Tie2-activating Ab attenuated the development of CNS autoimmune disease. Ang2 blockade inhibited expression of EC adhesion molecules, improved blood-brain barrier integrity, and decreased expression of genes involved in antigen presentation and proinflammatory responses of microglia and macrophages, which was accompanied by inhibition of α5β1 integrin activation in microglia. Taken together, our data suggest that Ang2 provides a target for increasing Tie2 activation in ECs and inhibiting proinflammatory polarization of CNS myeloid cells via α5β1 integrin in neuroinflammation. Thus, Ang2 targeting may serve as a therapeutic option for the treatment of CNS autoimmune disease.
Collapse
Affiliation(s)
- Zhilin Li
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emilia A Korhonen
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Arianna Merlini
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Judith Strauss
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Eleonoora Wihuri
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Harri Nurmi
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Salli Antila
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jennifer Paech
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | | | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
64
|
Zhao YT, Fallas JA, Saini S, Ueda G, Somasundaram L, Zhou Z, Xavier I, Ehnes D, Xu C, Carter L, Wrenn S, Mathieu J, Sellers DL, Baker D, Ruohola-Baker H. F-domain valency determines outcome of signaling through the angiopoietin pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33501432 PMCID: PMC7836102 DOI: 10.1101/2020.09.19.304188] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Angiopoietin 1 and 2 (Ang1 and Ang2) modulate angiogenesis and vascular homeostasis through engagement of their very similar F-domain modules with the Tie2 receptor tyrosine kinase on endothelial cells. Despite this similarity in the underlying receptor binding interaction, the two angiopoietins have opposite effects: Ang1 induces phosphorylation of protein kinase B (AKT), strengthens cell-cell junctions and enhances endothelial cell survival while Ang2 antagonizes these effects1–4. To investigate the molecular basis for the opposing effects, we examined the protein kinase activation and morphological phenotypes produced by a series of computationally designed protein scaffolds presenting the Ang1 F-domain in a wide range of valencies and geometries. We find two broad phenotypic classes distinguished by the number of presented F-domains: scaffolds presenting 4 F-domains have Ang2 like activity, upregulating pFAK and pERK but not pAKT, and failing to induce cell migration and tube formation, while scaffolds presenting 6 or more F-domains have Ang1 like activity, upregulating pAKT and inducing migration and tube formation. The scaffolds with 8 or more F-domains display superagonist activity, producing stronger phenotypes at lower concentrations than Ang1. When examined in vivo, superagonist icosahedral self-assembling nanoparticles caused significant revascularization in hemorrhagic brains after a controlled cortical impact injury.
Collapse
|
65
|
Juffermans NP, van den Brom CE, Kleinveld DJB. Targeting Endothelial Dysfunction in Acute Critical Illness to Reduce Organ Failure. Anesth Analg 2020; 131:1708-1720. [PMID: 33186159 DOI: 10.1213/ane.0000000000005023] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During hyperinflammatory conditions that can occur in acute critical illness, such as shock or hypoperfusion, inflammatory mediators activate the endothelium, fueling a proinflammatory host-response as well as procoagulant processes. These changes result in shedding of the glycocalyx, endothelial hyperpermeability, edema formation, and lead to disturbed microcirculatory perfusion and organ failure. Different fluid strategies that are used in shock may have differential effects on endothelial integrity. Collectively, low protein content fluids seem to have negative effects on the endothelial glycocalyx, aggravating endothelial hyperpermeability, whereas fluids containing albumin or plasma proteins may be superior to normal saline in protecting the glycocalyx and endothelial barrier function. Targeting the endothelium may be a therapeutic strategy to limit organ failure, which hitherto has not received much attention. Treatment targets aimed at restoring the endothelium should focus on maintaining glycocalyx function and/or targeting coagulation pathways or specific endothelial receptors. Potential treatments could be supplementing glycocalyx constituents or inhibiting glycocalyx breakdown. In this review, we summarize mechanisms of endothelial dysfunction during acute critical illness, such as the systemic inflammatory response, shedding of the glycocalyx, endothelial activation, and activation of coagulation. In addition, this review focuses on the effects of different fluid strategies on endothelial permeability. Also, potential mechanisms for treatment options to reduce endothelial hyperpermeability with ensuing organ failure are evaluated. Future research is needed to elucidate these pathways and to translate these data to the first human safety and feasibility trials.
Collapse
Affiliation(s)
- Nicole P Juffermans
- From the Department of Intensive Care, Onze Lieve Vrouwe Gasthuis, Amsterdam, the Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Charissa E van den Brom
- Department of Anesthesiology, Amsterdam UMC, VU Amsterdam, Amsterdam, the Netherlands.,Experimental Laboratory for Vital Signs, Amsterdam UMC, VU Amsterdam, Amsterdam, the Netherlands
| | - Derek J B Kleinveld
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
66
|
Zhang P, Peng J, Ren YQ, Zheng H, Yan H. Dexmedetomidine protects against endothelial injury in septic rats induced by cecal ligation and puncture by decreasing angiopoietin 2 and increasing vascular endothelial cadherin levels. Exp Ther Med 2020; 21:111. [PMID: 33335574 PMCID: PMC7739856 DOI: 10.3892/etm.2020.9543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/05/2020] [Indexed: 12/02/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of dexmedetomidine (Dex) on endothelial injury in a cecal ligation and puncture (CLP)-induced rat model of sepsis. A total of 36 male Sprague-Dawley rats were divided into three groups: Sham, CLP and CLP + Dex. The wet/dry (W/D) ratio of lung weight, hematoxylin and eosin (H&E) staining of lung tissue, plasma levels of angiopoietin (Ang)1 and 2, ratio of Ang2/1 and vascular endothelial (VE)-cadherin protein expression levels in lung tissue were determined. The W/D ratio of lung tissue in the CLP + Dex group was significantly lower than that in the CLP group (P<0.01). The H&E staining results indicated that Dex treatment reduced the levels of CLP-induced alveolar septum widening, infiltrating white blood cells and congestion, when compared with CLP alone. In addition, the expression levels of plasma Ang2 and the Ang2/1 ratio in the CLP + Dex group were significantly lower than those of the CLP rats (P<0.01). Furthermore, the level of VE-cadherin protein in lung tissue of the CLP + Dex group was higher than that of the CLP group (P<0.05). The results indicated that Dex had a protective effect against CLP-induced endothelial injury, through the ability to reduce expression of the endothelial injury factor Ang2 and increase the expression of the endothelial adhesion junction factor VE-cadherin in a septic rat model. These data suggest a potential application of Dex in the clinical treatment of sepsis.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Ji Peng
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Yun-Qin Ren
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Han Zheng
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Hong Yan
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
67
|
McBride A, Chanh HQ, Fraser JF, Yacoub S, Obonyo NG. Microvascular dysfunction in septic and dengue shock: Pathophysiology and implications for clinical management. Glob Cardiol Sci Pract 2020; 2020:e202029. [PMID: 33447608 PMCID: PMC7773436 DOI: 10.21542/gcsp.2020.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The microcirculation comprising of arterioles, capillaries and post-capillary venules is the terminal vascular network of the systemic circulation. Microvascular homeostasis, comprising of a balance between vasoconstriction, vasodilation and endothelial permeability in healthy states, regulates tissue perfusion. In severe infections, systemic inflammation occurs irrespective of the infecting microorganism(s), resulting in microcirculatory dysregulation and dysfunction, which impairs tissue perfusion and often precedes end-organ failure. The common hallmarks of microvascular dysfunction in both septic shock and dengue shock, are endothelial cell activation, glycocalyx degradation and plasma leak through a disrupted endothelial barrier. Microvascular tone is also impaired by a reduced bioavailability of nitric oxide. In vitro and in vivo studies have however demonstrated that the nature and extent of microvascular dysfunction as well as responses to volume expansion resuscitation differ in these two clinical syndromes. This review compares and contrasts the pathophysiology of microcirculatory dysfunction in septic versus dengue shock and the attendant effects of fluid administration during resuscitation.
Collapse
Affiliation(s)
- Angela McBride
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam.,Brighton and Sussex Medical School, United Kingdom
| | - Ho Q Chanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
| | - John F Fraser
- Critical Care Research Group, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Sophie Yacoub
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam.,Centre for Tropical Medicine and Global Health, University of Oxford, United Kingdom
| | - Nchafatso G Obonyo
- Critical Care Research Group, Brisbane, Australia.,KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.,Initiative to Develop African Research Leaders, Kilifi, Kenya
| |
Collapse
|
68
|
Jeong JH, Ojha U, Lee YM. Pathological angiogenesis and inflammation in tissues. Arch Pharm Res 2020; 44:1-15. [PMID: 33230600 PMCID: PMC7682773 DOI: 10.1007/s12272-020-01287-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022]
Abstract
The role of angiogenesis in the growth of organs and tumors is widely recognized. Vascular-organ interaction is a key mechanism and a concept that enables an understanding of all biological phenomena and normal physiology that is essential for human survival under pathological conditions. Recently, vascular endothelial cells have been classified as a type of innate immune cells that are dependent on the pathological situations. Moreover, inflammatory cytokines and signaling regulators activated upon exposure to infection or various stresses play crucial roles in the pathological function of parenchymal cells, peripheral immune cells, stromal cells, and cancer cells in tissues. Therefore, vascular-organ interactions as a vascular microenvironment or tissue microenvironment under physiological and pathological conditions are gaining popularity as an interesting research topic. Here, we review vascular contribution as a major factor in microenvironment homeostasis in the pathogenesis of normal as well as cancerous tissues. Furthermore, we suggest that the normalization strategy of pathological angiogenesis could be a promising therapeutic target for various diseases, including cancer.
Collapse
Affiliation(s)
- Ji-Hak Jeong
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea.,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea
| | - You Mie Lee
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea. .,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
69
|
Gengenbacher N, Singhal M, Mogler C, Hai L, Milde L, Pari AAA, Besemfelder E, Fricke C, Baumann D, Gehrs S, Utikal J, Felcht M, Hu J, Schlesner M, Offringa R, Chintharlapalli SR, Augustin HG. Timed Ang2-Targeted Therapy Identifies the Angiopoietin-Tie Pathway as Key Regulator of Fatal Lymphogenous Metastasis. Cancer Discov 2020; 11:424-445. [PMID: 33106316 DOI: 10.1158/2159-8290.cd-20-0122] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/13/2020] [Accepted: 10/09/2020] [Indexed: 11/16/2022]
Abstract
Recent clinical and preclinical advances have highlighted the existence of a previously hypothesized lymphogenous route of metastasis. However, due to a lack of suitable preclinical modeling tools, its contribution to long-term disease outcome and relevance for therapy remain controversial. Here, we established a genetically engineered mouse model (GEMM) fragment-based tumor model uniquely sustaining a functional network of intratumoral lymphatics that facilitates seeding of fatal peripheral metastases. Multiregimen survival studies and correlative patient data identified primary tumor-derived Angiopoietin-2 (Ang2) as a potent therapeutic target to restrict lymphogenous tumor cell dissemination. Mechanistically, tumor-associated lymphatic endothelial cells (EC), in contrast to blood vascular EC, were found to be critically addicted to the Angiopoietin-Tie pathway. Genetic manipulation experiments in combination with single-cell mapping revealed agonistically acting Ang2-Tie2 signaling as key regulator of lymphatic maintenance. Correspondingly, acute presurgical Ang2 neutralization was sufficient to prolong survival by regressing established intratumoral lymphatics, hence identifying a therapeutic regimen that warrants further clinical evaluation. SIGNIFICANCE: Exploiting multiple mouse tumor models including a unique GEMM-derived allograft system in combination with preclinical therapy designs closely matching the human situation, this study provides fundamental insight into the biology of tumor-associated lymphatic EC and defines an innovative presurgical therapeutic window of migrastatic Ang2 neutralization to restrict lymphogenous metastasis.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Nicolas Gengenbacher
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Munich, Germany
| | - Ling Hai
- Junior Group Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Milde
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Ashik Ahmed Abdul Pari
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Eva Besemfelder
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Claudine Fricke
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Daniel Baumann
- Faculty of Biosciences, Heidelberg University, Mannheim, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie Gehrs
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Moritz Felcht
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Matthias Schlesner
- Junior Group Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rienk Offringa
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany. .,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
70
|
Liu P, Ryczko M, Xie X, Baardsnes J, Lord-Dufour S, Duroche Y, Hicks EA, Taiyab A, Sheardown H, Quaggin SE, Jin J. New soluble angiopoietin analog of Hepta-ANG1 prevents pathological vascular leakage. Biotechnol Bioeng 2020; 118:423-432. [PMID: 32970320 DOI: 10.1002/bit.27580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 11/09/2022]
Abstract
Vascular leak is a key driver of organ injury in diseases, and strategies that reduce enhanced permeability and vascular inflammation are promising therapeutic targets. Activation of the angiopoietin-1 (ANG1)-Tie2 tyrosine kinase signaling pathway is an important regulator of vascular quiescence. Here we describe the design and construction of a new soluble ANG1 mimetic that is a potent activator of endothelial Tie2 in vitro and in vivo. Using a chimeric fusion strategy, we replaced the extracellular matrix (ECM) binding and oligomerization domain of ANG1 with a heptameric scaffold derived from the C-terminus of serum complement protein C4-binding protein α. We refer to this new fusion protein biologic as Hepta-ANG1, which forms a stable heptamer and induces Tie2 phosphorylation in cultured cells, and in the lung following intravenous injection of mice. Injection of Hepta-ANG1 ameliorates vascular endothelial growth factor- and lipopolysaccharide-induced vascular leakage, in keeping with the known functions of Angpt1-Tie2 in maintaining quiescent vascular stability. The new Hepta-ANG1 fusion is easy to produce and displays remarkable stability with high multimericity that can potently activate Tie2. It could be a new candidate ANG1 mimetic therapy for treatments of inflammatory vascular leak, such as acute respiratory distress syndrome and sepsis.
Collapse
Affiliation(s)
- Pan Liu
- Department of Medicine/Nephrology, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| | | | - Xinfang Xie
- Department of Medicine/Nephrology, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA.,Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, Qubec, Canada
| | - Simon Lord-Dufour
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, Qubec, Canada
| | - Yves Duroche
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, Qubec, Canada
| | - Emily Anne Hicks
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Aftab Taiyab
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Heather Sheardown
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Susan E Quaggin
- Department of Medicine/Nephrology, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Jing Jin
- Department of Medicine/Nephrology, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
71
|
Kapiainen E, Kihlström MK, Pietilä R, Kaakinen M, Ronkainen VP, Tu H, Heikkinen A, Devarajan R, Miinalainen I, Laitakari A, Ansarizadeh M, Zhang Q, Wei GH, Ruddock L, Pihlajaniemi T, Elamaa H, Eklund L. The Amino-Terminal Oligomerization Domain of Angiopoietin-2 Affects Vascular Remodeling, Mammary Gland Tumor Growth, and Lung Metastasis in Mice. Cancer Res 2020; 81:129-143. [PMID: 33037065 DOI: 10.1158/0008-5472.can-19-1904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 05/03/2020] [Accepted: 10/06/2020] [Indexed: 11/16/2022]
Abstract
Angiopoietin-2 (ANGPT2) is a context-dependent TIE2 agonistic or antagonistic ligand that induces diverse responses in cancer. Blocking ANGPT2 provides a promising strategy for inhibiting tumor growth and metastasis, yet variable effects of targeting ANGPT2 have complicated drug development. ANGPT2443 is a naturally occurring, lower oligomeric protein isoform whose expression is increased in cancer. Here, we use a knock-in mouse line (mice expressing Angpt2443), a genetic model for breast cancer and metastasis (MMTV-PyMT), a syngeneic melanoma lung colonization model (B16F10), and orthotopic injection of E0771 breast cancer cells to show that alternative forms increase the diversity of Angpt2 function. In a mouse retina model of angiogenesis, expression of Angpt2443 caused impaired venous development, suggesting enhanced function as a competitive antagonist for Tie2. In mammary gland tumor models, Angpt2443 differentially affected primary tumor growth and vascularization; these varying effects were associated with Angpt2 protein localization in the endothelium or in the stromal extracellular matrix as well as the frequency of Tie2-positive tumor blood vessels. In the presence of metastatic cells, Angpt2443 promoted destabilization of pulmonary vasculature and lung metastasis. In vitro, ANGPT2443 was susceptible to proteolytical cleavage, resulting in a monomeric ligand (ANGPT2DAP) that inhibited ANGPT1- or ANGPT4-induced TIE2 activation but did not bind to alternative ANGPT2 receptor α5β1 integrin. Collectively, these data reveal novel roles for the ANGPT2 N-terminal domain in blood vessel remodeling, tumor growth, metastasis, integrin binding, and proteolytic regulation. SIGNIFICANCE: This study identifies the role of the N-terminal oligomerization domain of angiopoietin-2 in vascular remodeling and lung metastasis and provides new insights into mechanisms underlying the versatile functions of angiopoietin-2 in cancer.See related commentary by Kamiyama and Augustin, p. 35.
Collapse
Affiliation(s)
- Emmi Kapiainen
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Minna K Kihlström
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Riikka Pietilä
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | - Hongmin Tu
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Anne Heikkinen
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Raman Devarajan
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Anna Laitakari
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Mohammadhassan Ansarizadeh
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Qin Zhang
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Gong-Hong Wei
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Lloyd Ruddock
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Harri Elamaa
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland. .,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
72
|
Yoon C, Kim D, Lim JH, Lee GM. Forskolin Increases cAMP Levels and Enhances Recombinant Antibody Production in CHO Cell Cultures. Biotechnol J 2020. [DOI: 10.1002/biot.202000264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Chansik Yoon
- Department of Biological Sciences KAIST Daejeon 34141 Republic of Korea
| | - Dongil Kim
- Department of Biological Sciences KAIST Daejeon 34141 Republic of Korea
| | - Ju Hyeon Lim
- New Drug Development Center Cheongju 28160 Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences KAIST Daejeon 34141 Republic of Korea
| |
Collapse
|
73
|
Zhang PY, Fan Y, Tan T, Yu Y. Generation of Artificial Gamete and Embryo From Stem Cells in Reproductive Medicine. Front Bioeng Biotechnol 2020; 8:781. [PMID: 32793569 PMCID: PMC7387433 DOI: 10.3389/fbioe.2020.00781] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022] Open
Abstract
In addition to the great growing need for assisted reproduction technologies (ART), additional solutions for patients without functional gametes are strongly needed. Due to ethical restrictions, limited studies can be performed on human gametes and embryos; however, artificial gametes and embryos represent a new hope for clinical application and basic research in the field of reproductive medicine. Here, we provide a review of the research progress and possible application of artificial gametes and embryos from different species, including mice, monkeys and humans. Gametes specification from adult stem cells and embryonic stem cells (ESCs) as well as propagation of stem cells from the reproductive system and from organized embryos, which are similar to blastocysts, have been realized in some nonhuman mammals, but not all achievements can be replicated in humans. This area of research remains noteworthy and requires further study and effort to achieve the reconstitution of the entire cycle of gametogenesis and embryo development in vitro.
Collapse
Affiliation(s)
- Pu-Yao Zhang
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tao Tan
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
74
|
Mirando AC, Lima e Silva R, Chu Z, Campochiaro PA, Pandey NB, Popel AS. Suppression of Ocular Vascular Inflammation through Peptide-Mediated Activation of Angiopoietin-Tie2 Signaling. Int J Mol Sci 2020; 21:ijms21145142. [PMID: 32708100 PMCID: PMC7404316 DOI: 10.3390/ijms21145142] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023] Open
Abstract
Persistent inflammation is a complication associated with many ocular diseases. Changes in ocular vessels can amplify disease responses and contribute to vision loss by influencing the delivery of leukocytes to the eye, vascular leakage, and perfusion. Here, we report the anti-inflammatory activity for AXT107, a non-RGD, 20-mer αvβ3 and α5β1 integrin-binding peptide that blocks vascular endothelial growth factor (VEGF)-signaling and activates tyrosine kinase with immunoglobulin and EGF-like domains 2 (Tie2) using the normally inhibitory ligand angiopoietin 2 (Ang2). Tumor necrosis factor α (TNFα), a central inflammation mediator, induces Ang2 release from endothelial cells to enhance its stimulation of inflammation and vascular leakage. AXT107 resolves TNFα-induced vascular inflammation in endothelial cells by converting the endogenously released Ang2 into an agonist of Tie2 signaling, thereby disrupting both the synergism between TNFα and Ang2 while also preventing inhibitor of nuclear factor-κB α (IκBα) degradation directly through Tie2 signaling. This recovery of IκBα prevents nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) nuclear localization, thereby blocking NF-κB-induced inflammatory responses, including the production of VCAM-1 and ICAM-1, leukostasis, and vascular leakage in cell and mouse models. AXT107 also decreased the levels of pro-inflammatory TNF receptor 1 (TNFR1) without affecting levels of the more protective TNFR2. These data suggest that AXT107 may provide multiple benefits in the treatment of retinal/choroidal and other vascular diseases by suppressing inflammation and promoting vascular stabilization.
Collapse
MESH Headings
- Angiopoietin-1/metabolism
- Angiopoietin-2/metabolism
- Animals
- Capillary Permeability/drug effects
- Choroid Diseases/drug therapy
- Collagen Type IV/pharmacology
- Collagen Type IV/therapeutic use
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Human Umbilical Vein Endothelial Cells
- Humans
- I-kappa B Kinase/metabolism
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Leukostasis/drug therapy
- Leukostasis/metabolism
- Mice
- Mice, Inbred C57BL
- Peptide Fragments/pharmacology
- Peptide Fragments/therapeutic use
- Receptor, TIE-2/agonists
- Receptor, TIE-2/metabolism
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Retinal Diseases/drug therapy
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/pharmacology
- Vascular Cell Adhesion Molecule-1/metabolism
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Adam C. Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
| | - Raquel Lima e Silva
- Department of Ophthalmology and the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.L.e.S.); (P.A.C.)
| | - Zenny Chu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
| | - Peter A. Campochiaro
- Department of Ophthalmology and the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.L.e.S.); (P.A.C.)
| | - Niranjan B. Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
- AsclepiX Therapeutics, Inc., Baltimore, MD 21211, USA
- Correspondence: (N.B.P.); (A.S.P.); Tel.: +908-328-2019 (N.B.P.); +410-955-6419 (A.S.P.)
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
- Correspondence: (N.B.P.); (A.S.P.); Tel.: +908-328-2019 (N.B.P.); +410-955-6419 (A.S.P.)
| |
Collapse
|
75
|
Bhatraju PK, Cohen M, Nagao RJ, Morrell ED, Kosamo S, Chai XY, Nance R, Dmyterko V, Delaney J, Christie JD, Liu KD, Mikacenic C, Gharib SA, Liles WC, Zheng Y, Christiani DC, Himmelfarb J, Wurfel MM. Genetic variation implicates plasma angiopoietin-2 in the development of acute kidney injury sub-phenotypes. BMC Nephrol 2020; 21:284. [PMID: 32680471 PMCID: PMC7368773 DOI: 10.1186/s12882-020-01935-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 07/07/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND We previously identified two acute kidney injury (AKI) sub-phenotypes (AKI-SP1 and AKI-SP2) with different risk of poor clinical outcomes and response to vasopressor therapy. Plasma biomarkers of endothelial dysfunction (tumor necrosis factor receptor-1, angiopoietin-1 and 2) differentiated the AKI sub-phenotypes. However, it is unknown whether these biomarkers are simply markers or causal mediators in the development of AKI sub-phenotypes. METHODS We tested for associations between single-nucleotide polymorphisms within the Angiopoietin-1, Angiopoietin-2, and Tumor Necrosis Factor Receptor 1A genes and AKI- SP2 in 421 critically ill subjects of European ancestry. Top performing single-nucleotide polymorphisms (FDR < 0.05) were tested for cis-biomarker expression and whether genetic risk for AKI-SP2 is mediated through circulating biomarkers. We also completed in vitro studies using human kidney microvascular endothelial cells. Finally, we calculated the renal clearance of plasma biomarkers using 20 different timed urine collections. RESULTS A genetic variant, rs2920656C > T, near ANGPT2 was associated with reduced risk of AKI-SP2 (odds ratio, 0.45; 95% CI, 0.31-0.66; adjusted FDR = 0.003) and decreased plasma angiopoietin-2 (p = 0.002). Causal inference analysis showed that for each minor allele (T) the risk of developing AKI-SP2 decreases by 16%. Plasma angiopoietin-2 mediated 41.5% of the rs2920656 related risk for AKI-SP2. Human kidney microvascular endothelial cells carrying the T allele of rs2920656 produced numerically lower levels of angiopoietin-2 although this was not statistically significant (p = 0.07). Finally, analyses demonstrated that angiopoietin-2 is minimally renally cleared in critically ill subjects. CONCLUSION Genetic mediation analysis provides supportive evidence that angiopoietin-2 plays a causal role in risk for AKI-SP2.
Collapse
Affiliation(s)
- Pavan K. Bhatraju
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA ,grid.34477.330000000122986657Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, USA
| | - Max Cohen
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - Ryan J. Nagao
- grid.34477.330000000122986657Department of Bioengineering, University of Washington and Center for Cardiovascular Biology, Seattle, USA ,grid.34477.330000000122986657Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Eric D. Morrell
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - Susanna Kosamo
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - Xin-Ya Chai
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - Robin Nance
- grid.34477.330000000122986657Department of Epidemiology, University of Washington, Seattle, USA
| | - Victoria Dmyterko
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - Joseph Delaney
- grid.34477.330000000122986657Department of Epidemiology, University of Washington, Seattle, USA
| | - Jason D. Christie
- grid.25879.310000 0004 1936 8972Division of Pulmonary, Allergy, and Critical Care and Center for Clinical Epidemiology and Biostatistics, Department of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Kathleen D. Liu
- grid.266102.10000 0001 2297 6811Divisions of Nephrology and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Carmen Mikacenic
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - Sina A. Gharib
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA
| | - W. Conrad Liles
- grid.34477.330000000122986657Department of Medicine, University of Washington, Seattle, USA
| | - Ying Zheng
- grid.34477.330000000122986657Department of Bioengineering, University of Washington and Center for Cardiovascular Biology, Seattle, USA ,grid.34477.330000000122986657Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - David C. Christiani
- grid.38142.3c000000041936754XDepartments of Environmental Health and Epidemiology, Harvard TH Chan School of Public Health, Harvard University and Pulmonary and Critical Care Division, Cambridge, USA ,Department of Medicine, MA General Hospital/Harvard Medical School, Boston, USA
| | - Jonathan Himmelfarb
- grid.34477.330000000122986657Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, USA
| | - Mark M. Wurfel
- grid.34477.330000000122986657Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Seattle, WA 98104 USA ,grid.34477.330000000122986657Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, USA
| |
Collapse
|
76
|
Probst CK, Montesi SB, Medoff BD, Shea BS, Knipe RS. Vascular permeability in the fibrotic lung. Eur Respir J 2020; 56:13993003.00100-2019. [PMID: 32265308 PMCID: PMC9977144 DOI: 10.1183/13993003.00100-2019] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 03/26/2020] [Indexed: 12/26/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is thought to result from aberrant tissue repair processes in response to chronic or repetitive lung injury. The origin and nature of the injury, as well as its cellular and molecular targets, are likely heterogeneous, which complicates accurate pre-clinical modelling of the disease and makes therapeutic targeting a challenge. Efforts are underway to identify central pathways in fibrogenesis which may allow targeting of aberrant repair processes regardless of the initial injury stimulus. Dysregulated endothelial permeability and vascular leak have long been studied for their role in acute lung injury and repair. Evidence that these processes are of importance to the pathogenesis of fibrotic lung disease is growing. Endothelial permeability is increased in non-fibrosing lung diseases, but it resolves in a self-limited fashion in conditions such as bacterial pneumonia and acute respiratory distress syndrome. In progressive fibrosing diseases such as IPF, permeability appears to persist, however, and may also predict mortality. In this hypothesis-generating review, we summarise available data on the role of endothelial permeability in IPF and focus on the deleterious consequences of sustained endothelial hyperpermeability in response to and during pulmonary inflammation and fibrosis. We propose that persistent permeability and vascular leak in the lung have the potential to establish and amplify the pro-fibrotic environment. Therapeutic interventions aimed at recognising and "plugging" the leak may therefore be of significant benefit for preventing the transition from lung injury to fibrosis and should be areas for future research.
Collapse
Affiliation(s)
- Clemens K. Probst
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sydney B. Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Barry S. Shea
- Division of Pulmonary and Critical Care Medicine, Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Rachel S. Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
77
|
Hypoxia: Turning vessels into vassals of cancer immunotolerance. Cancer Lett 2020; 487:74-84. [PMID: 32470491 DOI: 10.1016/j.canlet.2020.05.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a universal feature of solid cancers caused by a mismatch between cellular oxygen supply and consumption. To meet the increased demand for oxygen, hypoxic cancer cells (CCs) induce a multifaceted process known as angiogenesis, wherein new vessels are formed by the sprouting of pre-existing ones. In addition to providing oxygen for growth and an exit route for dissemination, angiogenic vessels and factors are co-opted by CCs to enable the generation of an immunotolerant, hypoxic tumor microenvironment, leading to therapeutic failure and mortality. In this review, we discuss how hypoxia-inducible factors (HIFs), the mechanistic target of rapamycin (mTOR), and the unfolded protein response (UPR) control angiogenic factors serving both vascular and immunomodulatory functions in the tumor microenvironment. Possible therapeutic strategies, wherein targeting oxygen sensing might enhance anti-angiogenic and immunologically-mediated anti-cancer responses, are suggested.
Collapse
|
78
|
Zhao H, Zhu Y, Zhang J, Wu Y, Xiang X, Zhang Z, Li T, Liu L. The Beneficial Effect of HES on Vascular Permeability and Its Relationship With Endothelial Glycocalyx and Intercellular Junction After Hemorrhagic Shock. Front Pharmacol 2020; 11:597. [PMID: 32457611 PMCID: PMC7227604 DOI: 10.3389/fphar.2020.00597] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background Vascular leakage is a common complication of hemorrhagic shock. Endothelial glycocalyx plays a crucial role in the protection of vascular endothelial barrier function. Hydroxyethyl starch (HES) is a commonly used resuscitation fluid for hemorrhagic shock. However, whether the protective effect of HES on vascular permeability after hemorrhagic shock is associated with the endothelial glycocalyx is unclear. Methods Using hemorrhagic shock rat model and hypoxia treated vascular endothelial cells (VECs), effects of HES (130/0.4) on pulmonary vascular permeability and the relationship to endothelial glycocalyx were observed. Results Pulmonary vascular permeability was significantly increased after hemorrhagic shock, as evidenced by the increased permeability of pulmonary vessels to albumin-fluorescein isothiocyanate conjugate (FITC-BSA) and Evans blue, the decreased transendothelial electrical resistance of VECs and the increased transmittance of FITC-BSA. The structure of the endothelial glycocalyx was destroyed, showing a decrease in thickness. The expression of heparan sulfate, hyaluronic acid, and chondroitin sulfate, the components of the endothelial glycocalyx, was significantly decreased. HES (130/0.4) significantly improved the vascular barrier function, recovered the thickness and the expression of components of the endothelial glycocalyx by down-regulating the expression of heparinase, hyaluronidase, and neuraminidase, and meanwhile increased the expression of intercellular junction proteins ZO-1, occludin, and VE-cadherin. Degradation of endothelial glycocalyx with degrading enzyme (heparinase, hyaluronidase, and neuraminidase) abolished the beneficial effect of HES on vascular permeability, but had no significant effect on the recovery of the expression of endothelial intercellular junction proteins induced by HES (130/0.4). HES (130/0.4) decreased the expression of cleaved-caspase-3 induced by hemorrhagic shock. Conclusions HES (130/0.4) has protective effect on vascular barrier function after hemorrgic shock.The mechanism is mainly related to the protective effect of HES on endothelial glycocalyx and intercellular junction proteins. The protective effect of HES on endothelial glycocalyx was associated with the down-regulated expression of heparinase, hyaluronidase, and neuraminidase. HES (130/0.4) had an anti-apoptotic effect in hemorrhagic shock.
Collapse
Affiliation(s)
- Hongliang Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xinming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zisen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
79
|
Pike DP, Vogel MJ, McHowat J, Mikuzis PA, Schulte KA, Ford DA. 2-Chlorofatty acids are biomarkers of sepsis mortality and mediators of barrier dysfunction in rats. J Lipid Res 2020; 61:1115-1127. [PMID: 32376642 DOI: 10.1194/jlr.ra120000829] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/05/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis is defined as the systemic, dysregulated host immune response to an infection that leads to injury to host organ systems and, often, death. Complex interactions between pathogens and their hosts elicit microcirculatory dysfunction. Neutrophil myeloperoxidase (MPO) is critical for combating pathogens, but MPO-derived hypochlorous acid (HOCl) can react with host molecular species as well. Plasmalogens are targeted by HOCl, leading to the production of 2-chlorofatty acids (2-CLFAs). 2-CLFAs are associated with human sepsis mortality, decrease in vitro endothelial barrier function, and activate human neutrophil extracellular trap formation. Here, we sought to examine 2-CLFAs in an in vivo rat sepsis model. Intraperitoneal cecal slurry sepsis with clinically relevant rescue therapies led to ∼73% mortality and evidence of microcirculatory dysfunction. Plasma concentrations of 2-CLFAs assessed 8 h after sepsis induction were lower in rats that survived sepsis than in nonsurvivors. 2-CLFA levels were elevated in kidney, liver, spleen, lung, colon, and ileum in septic animals. In vivo, exogenous 2-CLFA treatments increased kidney permeability, and in in vitro experiments, 2-CLFA also increased epithelial surface expression of vascular cell adhesion molecule 1 and decreased epithelial barrier function. Collectively, these studies support a role of free 2-CLFAs as biomarkers of sepsis mortality, potentially mediated, in part, by 2-CLFA-elicited endothelial and epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Daniel P Pike
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Michael J Vogel
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Jane McHowat
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104; Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Paul A Mikuzis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Kevin A Schulte
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104. mailto:
| |
Collapse
|
80
|
The ratio of plasma angiopoietin-2 to angiopoietin-1 as a prognostic biomarker in patients with sepsis. Cytokine 2020; 129:155029. [DOI: 10.1016/j.cyto.2020.155029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/01/2023]
|
81
|
LaRivière WB, Liao S, McMurtry SA, Oshima K, Han X, Zhang F, Yan S, Haeger SM, Ransom M, Bastarache JA, Linhardt RJ, Schmidt EP, Yang Y. Alveolar heparan sulfate shedding impedes recovery from bleomycin-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1198-L1210. [PMID: 32320623 DOI: 10.1152/ajplung.00063.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pulmonary epithelial glycocalyx, an anionic cell surface layer enriched in glycosaminoglycans such as heparan sulfate and chondroitin sulfate, contributes to the alveolar barrier. Direct injury to the pulmonary epithelium induces shedding of heparan sulfate into the air space; the impact of this shedding on recovery after lung injury is unknown. Using mass spectrometry, we found that heparan sulfate was shed into the air space for up to 3 wk after intratracheal bleomycin-induced lung injury and coincided with induction of matrix metalloproteinases (MMPs), including MMP2. Delayed inhibition of metalloproteinases, beginning 7 days after bleomycin using the nonspecific MMP inhibitor doxycycline, attenuated heparan sulfate shedding and improved lung function, suggesting that heparan sulfate shedding may impair lung recovery. While we also observed an increase in air space heparanase activity after bleomycin, pharmacological and transgenic inhibition of heparanase in vivo failed to attenuate heparan sulfate shedding or protect against bleomycin-induced lung injury. However, experimental augmentation of airway heparanase activity significantly worsened post-bleomycin outcomes, confirming the importance of epithelial glycocalyx integrity to lung recovery. We hypothesized that MMP-associated heparan sulfate shedding contributed to delayed lung recovery, in part, by the release of large, highly sulfated fragments that sequestered lung-reparative growth factors such as hepatocyte growth factor. In vitro, heparan sulfate bound hepatocyte growth factor and attenuated growth factor signaling, suggesting that heparan sulfate shed into the air space after injury may directly impair lung repair. Accordingly, administration of exogenous heparan sulfate to mice after bleomycin injury increased the likelihood of death due to severe lung dysfunction. Together, our findings demonstrate that alveolar epithelial heparan sulfate shedding impedes lung recovery after bleomycin.
Collapse
Affiliation(s)
- W B LaRivière
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - S Liao
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - S A McMurtry
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - K Oshima
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - X Han
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, New York
| | - F Zhang
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, New York
| | - S Yan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,College of Life Sciences, Henan Normal University, Xinxiang, China
| | - S M Haeger
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - M Ransom
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - J A Bastarache
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - R J Linhardt
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, New York
| | - E P Schmidt
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, Denver Health Medical Center, Denver, Colorado
| | - Y Yang
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
82
|
Filewod NC, Lee WL. Inflammation without Vascular Leakage. Science Fiction No Longer? Am J Respir Crit Care Med 2020; 200:1472-1476. [PMID: 31381867 DOI: 10.1164/rccm.201905-1011cp] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vascular leakage is a characteristic of critical illnesses such as septic shock and acute respiratory distress syndrome. It results in hypotension and tissue edema and contributes to organ dysfunction. It has long been taught that increased vascular permeability is a natural consequence of inflammation; in particular, many clinicians believe that it occurs inevitably during leukocyte recruitment to a site of infection. In fact, abundant research now indicates that vascular leakage and leukocyte emigration do not necessarily occur together in a blood vessel. The molecular mechanisms underpinning these processes-allowing leukocytes to exit the circulation without increasing vascular permeability-are starting to be elucidated and establish vascular leakage as a viable therapeutic target. Several preclinical studies indicate that vascular leakage can be reduced without impairing cytokine production, leukocyte recruitment, and pathogen clearance. The realization that leukocyte traffic and vascular permeability can be regulated separately should spur development of therapies that decrease vascular leakage and tissue edema without compromising the immune response.
Collapse
Affiliation(s)
- Niall C Filewod
- Department of Critical Care Medicine and.,Keenan Research Centre for Biomedical Sciences, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Warren L Lee
- Department of Critical Care Medicine and.,Keenan Research Centre for Biomedical Sciences, St. Michael's Hospital, Toronto, Ontario, Canada; and.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
83
|
Crist AM, Zhou X, Garai J, Lee AR, Thoele J, Ullmer C, Klein C, Zabaleta J, Meadows SM. Angiopoietin-2 Inhibition Rescues Arteriovenous Malformation in a Smad4 Hereditary Hemorrhagic Telangiectasia Mouse Model. Circulation 2020; 139:2049-2063. [PMID: 30744395 DOI: 10.1161/circulationaha.118.036952] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hereditary hemorrhagic telangiectasia is an autosomal dominant vascular disorder caused by heterozygous, loss-of-function mutations in 4 transforming growth factor beta (TGFβ) pathway members, including the central transcriptional mediator of the TGFβ pathway, Smad4. Loss of Smad4 causes the formation of inappropriate, fragile connections between arteries and veins called arteriovenous malformations (AVMs), which can hemorrhage leading to stroke, aneurysm, or death. Unfortunately, the molecular mechanisms underlying AVM pathogenesis remain poorly understood, and the TGFβ downstream effectors responsible for hereditary hemorrhagic telangiectasia-associated AVM formation are currently unknown. METHODS To identify potential biological targets of the TGFβ pathway involved in AVM formation, we performed RNA- and chromatin immunoprecipitation-sequencing experiments on BMP9 (bone morphogenetic protein 9)-stimulated endothelial cells (ECs) and isolated ECs from a Smad4-inducible, EC-specific knockout ( Smad4-iECKO) mouse model that develops retinal AVMs. These sequencing studies identified the angiopoietin-Tek signaling pathway as a downstream target of SMAD4. We used monoclonal blocking antibodies to target a specific component in this pathway and assess its effects on AVM development. RESULTS Sequencing studies uncovered 212 potential biological targets involved in AVM formation, including the EC surface receptor, TEK (TEK receptor tyrosine kinase) and its antagonistic ligand, ANGPT2 (angiopoietin-2). In Smad4-iECKO mice, Angpt2 expression is robustly increased, whereas Tek levels are decreased, resulting in an overall reduction in angiopoietin-Tek signaling. We provide evidence that SMAD4 directly represses Angpt2 transcription in ECs. Inhibition of ANGPT2 function in Smad4-deficient mice, either before or after AVMs form, prevents and alleviates AVM formation and normalizes vessel diameters. These rescue effects are attributed to a reversion in EC morphological changes, such as cell size and shape that are altered in the absence of Smad4. CONCLUSIONS Our studies provide a novel mechanism whereby the loss of Smad4 causes increased Angpt2 transcription in ECs leading to AVM formation, increased blood vessel calibers, and changes in EC morphology in the retina. Blockade of ANGPT2 function in an in vivo Smad4 model of hereditary hemorrhagic telangiectasia alleviated these vascular phenotypes, further implicating ANGPT2 as an important TGFβ downstream mediator of AVM formation. Therefore, alternative approaches that target ANGPT2 function may have therapeutic value for the alleviation of hereditary hemorrhagic telangiectasia symptoms, such as AVMs.
Collapse
Affiliation(s)
- Angela M Crist
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA (A.M.C., X.Z., A.R.L., S.M.M.)
| | - Xingyan Zhou
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA (A.M.C., X.Z., A.R.L., S.M.M.)
| | - Jone Garai
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans (J.G., J.Z.)
| | - Amanda R Lee
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA (A.M.C., X.Z., A.R.L., S.M.M.)
| | - Janina Thoele
- Roche Innovation Center, Basel, Switzerland (J.T., C.U.)
| | | | | | - Jovanny Zabaleta
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans (J.G., J.Z.)
| | - Stryder M Meadows
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA (A.M.C., X.Z., A.R.L., S.M.M.)
| |
Collapse
|
84
|
Endothelial Glycocalyx Impairment in Disease: Focus on Hyaluronan Shedding. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:768-780. [PMID: 32035885 DOI: 10.1016/j.ajpath.2019.11.016] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023]
Abstract
Hyaluronan (HA) is a ubiquitous glycosaminoglycan of the extracellular matrix. It is present in the endothelial glycocalyx covering the apical surface of endothelial cells. The endothelial glycocalyx regulates blood vessel permeability and homeostasis. HA plays a central role in numerous functions of the endothelial surface layer, protecting the endothelial cells, regulating the barrier permeability, and ensuring mechanosensing, which is essential to nitric oxide production and flow-induced vasodilation. During acute injury, inflammatory conditions, or many other pathologic conditions, the endothelial glycocalyx is damaged, and its degradation is accompanied by shedding of one or more glycocalyx components into the blood. Syndecan-1, heparan sulfate, and HA are the main components whose shedding has been claimed to represent the endothelial glycocalyx state of health. This review focuses on endothelial glycocalyx HA and highlights its key roles in the functions of the endothelial glycocalyx, its shedding in several pathologic conditions such as sepsis, diabetes, chronic and acute kidney injury, ischemia/reperfusion, atherosclerosis, and inflammation, which are all accompanied by increased circulating HA levels. Plasma/serum HA level is becoming recognized as a biomarker of endothelial glycocalyx damage in select pathologies. Hyaluronidase, the main HA-degrading enzyme, and its involvement in the impairment of endothelial glycocalyx are also addressed.
Collapse
|
85
|
Bhatraju PK, Zelnick LR, Herting J, Katz R, Mikacenic C, Kosamo S, Morrell ED, Robinson-Cohen C, Calfee CS, Christie JD, Liu KD, Matthay MA, Hahn WO, Dmyterko V, Slivinski NSJ, Russell JA, Walley KR, Christiani DC, Liles WC, Himmelfarb J, Wurfel MM. Identification of Acute Kidney Injury Subphenotypes with Differing Molecular Signatures and Responses to Vasopressin Therapy. Am J Respir Crit Care Med 2020; 199:863-872. [PMID: 30334632 DOI: 10.1164/rccm.201807-1346oc] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Currently, no safe and effective pharmacologic interventions exist for acute kidney injury (AKI). One reason may be that heterogeneity exists within the AKI population, thereby hampering the identification of specific pathophysiologic pathways and therapeutic targets. OBJECTIVE The aim of this study was to identify and test whether AKI subphenotypes have prognostic and therapeutic implications. METHODS First, latent class analysis methodology was applied independently in two critically ill populations (discovery [n = 794] and replication [n = 425]) with AKI. Second, a parsimonious classification model was developed to identify AKI subphenotypes. Third, the classification model was applied to patients with AKI in VASST (Vasopressin and Septic Shock Trial; n = 271), and differences in treatment response were determined. In all three populations, AKI was defined using serum creatinine and urine output. MEASUREMENTS AND MAIN RESULTS A two-subphenotype latent class analysis model had the best fit in both the discovery (P = 0.004) and replication (P = 0.004) AKI groups. The risk of 7-day renal nonrecovery and 28-day mortality was greater with AKI subphenotype 2 (AKI-SP2) relative to AKI subphenotype 1 (AKI-SP1). The AKI subphenotypes discriminated risk for poor clinical outcomes better than the Kidney Disease: Improving Global Outcomes stages of AKI. A three-variable model that included markers of endothelial dysfunction and inflammation accurately determined subphenotype membership (C-statistic 0.92). In VASST, vasopressin compared with norepinephrine was associated with improved 90-day mortality in AKI-SP1 (27% vs. 46%, respectively; P = 0.02), but no significant difference was observed in AKI-SP2 (45% vs. 49%, respectively; P = 0.99) and the P value for interaction was 0.05. CONCLUSIONS This analysis identified two molecularly distinct AKI subphenotypes with different clinical outcomes and responses to vasopressin therapy. Identification of AKI subphenotypes could improve risk prognostication and may be useful for predictive enrichment in clinical trials.
Collapse
Affiliation(s)
- Pavan K Bhatraju
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine.,2 Kidney Research Institute, Division of Nephrology, and
| | | | | | - Ronit Katz
- 2 Kidney Research Institute, Division of Nephrology, and
| | | | - Susanna Kosamo
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine
| | - Eric D Morrell
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine
| | | | - Carolyn S Calfee
- 4 Department of Medicine.,5 Department of Anesthesia and Perioperative Care.,6 Cardiovascular Research Institute
| | - Jason D Christie
- 7 Division of Pulmonary, Allergy, and Critical Care and.,8 Center for Clinical Epidemiology and Biostatistics, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kathleen D Liu
- 9 Division of Nephrology, and.,10 Division of Critical Care Medicine, University of California, San Francisco, San Francisco, California
| | - Michael A Matthay
- 4 Department of Medicine.,5 Department of Anesthesia and Perioperative Care.,6 Cardiovascular Research Institute
| | - William O Hahn
- 11 Division of Allergy and Infectious Diseases, Department of Medicine
| | | | | | - Jim A Russell
- 13 Centre for Heart Lung Innovation and.,14 Division of Critical Care Medicine, Department of Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Keith R Walley
- 13 Centre for Heart Lung Innovation and.,14 Division of Critical Care Medicine, Department of Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - David C Christiani
- 15 Department of Environmental Health and.,16 Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, Massachusetts; and.,17 Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - W Conrad Liles
- 18 Department of Medicine, University of Washington, Seattle, Washington
| | | | - Mark M Wurfel
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine.,2 Kidney Research Institute, Division of Nephrology, and
| |
Collapse
|
86
|
Koch PD, Ahmed MS, Kohler RH, Li R, Weissleder R. Imaging of Tie2 with a Fluorescently Labeled Small Molecule Affinity Ligand. ACS Chem Biol 2020; 15:151-157. [PMID: 31809013 DOI: 10.1021/acschembio.9b00724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The receptor tyrosine kinase inhibitor, Tie2, has significant roles in endothelial signaling and angiogenesis and is relevant in the pathophysiology of several diseases. However, there are relatively few small molecule probes available to study Tie2, making the evaluation of its activity in vivo difficult. Recently, it was discovered that the small molecule rebastinib (DCC-2036) is a potent Tie2 inhibitor. We hypothesized that fluorescent derivatives of rebastinib could be used as imaging agents for Tie2. On the basis of crystallography structures, we synthesized three fluorescent derivatives, which we then evaluated in both in vitro and in vivo assays. We found that the Rebastinib-BODIPY TMR (Reb-TMR) derivative has superior imaging characteristics in vitro, and we successfully labeled endothelial cells in vivo. We propose that this probe could be further used in in vivo applications for studying the role of Tie2 in disease.
Collapse
Affiliation(s)
- Peter David Koch
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
| | - Maaz S. Ahmed
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
87
|
Gianni-Barrera R, Di Maggio N, Melly L, Burger MG, Mujagic E, Gürke L, Schaefer DJ, Banfi A. Therapeutic vascularization in regenerative medicine. Stem Cells Transl Med 2020; 9:433-444. [PMID: 31922362 PMCID: PMC7103618 DOI: 10.1002/sctm.19-0319] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Therapeutic angiogenesis, that is, the generation of new vessels by delivery of specific factors, is required both for rapid vascularization of tissue‐engineered constructs and to treat ischemic conditions. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis. However, uncontrolled expression can lead to aberrant vascular growth and vascular tumors (angiomas). Major challenges to fully exploit VEGF potency for therapy include the need to precisely control in vivo distribution of growth factor dose and duration of expression. In fact, the therapeutic window of VEGF delivery depends on its amount in the microenvironment around each producing cell rather than on the total dose, since VEGF remains tightly bound to extracellular matrix (ECM). On the other hand, short‐term expression of less than about 4 weeks leads to unstable vessels, which promptly regress following cessation of the angiogenic stimulus. Here, we will briefly overview some key aspects of the biology of VEGF and angiogenesis and discuss their therapeutic implications with a particular focus on approaches using gene therapy, genetically modified progenitors, and ECM engineering with recombinant factors. Lastly, we will present recent insights into the mechanisms that regulate vessel stabilization and the switch between normal and aberrant vascular growth after VEGF delivery, to identify novel molecular targets that may improve both safety and efficacy of therapeutic angiogenesis.
Collapse
Affiliation(s)
- Roberto Gianni-Barrera
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Nunzia Di Maggio
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Ludovic Melly
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Cardiac, Vascular, and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Maximilian G Burger
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Plastic and Reconstructive Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Edin Mujagic
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Lorenz Gürke
- Vascular Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Plastic and Reconstructive Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Plastic and Reconstructive Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
88
|
Hauschildt J, Schrimpf C, Thamm K, Retzlaff J, Idowu TO, von Kaisenberg C, Haller H, David S. Dual Pharmacological Inhibition of Angiopoietin-2 and VEGF-A in Murine Experimental Sepsis. J Vasc Res 2019; 57:34-45. [PMID: 31726451 DOI: 10.1159/000503787] [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: 09/02/2019] [Accepted: 09/30/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Sepsis is a pathological host response to infection leading to vascular barrier breakdown due to elevated levels of angiopoietin-2 (Angpt-2) and vascular endothelial growth factor-A (VEGF-A). Here, we tested a novel heterodimeric bispecific monoclonal IgG1-cross antibody of Angpt-2 and VEGF - termed "A2V." METHODS Cecal ligation and puncture was used to induce murine polymicrobial sepsis. Organs and blood were harvested for fluorescence immunohistochemistry and RT-PCR, and survival was recorded. In vitro endothelial cells were stimulated with plasma from septic shock patients costimulated with A2V or IgG antibody followed by immunocytochemistry and real-time transendothelial electrical resistance. RESULTS Septic mice treated with A2V had a reduced induction of the endothelial adhesion molecule ICAM-1, leading to a trend towards less transmigration of inflammatory cells (A2V: 42.2 ± 1.0 vs. IgG 48.5 ± 1.7 Gr-1+ cells/HPF, p = 0.08) and reduced tissue levels of inflammatory cytokines (e.g., IL-6 mRNA: A2V 9.4 ± 3.2 vs. IgG 83.9 ± 36.7-fold over control, p = 0.03). Endothelial permeability was improved in vivo and in vitro in stimulated endothelial cells with septic plasma. Survival was improved by 38% (p = 0.02). CONCLUSION Dual inhibition of Angpt-2 and VEGF-A improves murine sepsis morbidity and mortality, making it a potential therapeutic against vascular barrier breakdown.
Collapse
Affiliation(s)
- Janine Hauschildt
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Claudia Schrimpf
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Kristina Thamm
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jennifer Retzlaff
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Temitayo O Idowu
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | | | - Hermann Haller
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Sascha David
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany,
| |
Collapse
|
89
|
Zhang Y, Kontos CD, Annex BH, Popel AS. Angiopoietin-Tie Signaling Pathway in Endothelial Cells: A Computational Model. iScience 2019; 20:497-511. [PMID: 31655061 PMCID: PMC6806670 DOI: 10.1016/j.isci.2019.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/21/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
The angiopoietin-Tie signaling pathway is an important vascular signaling pathway involved in angiogenesis, vascular stability, and quiescence. Dysregulation in the pathway is linked to the impairments in vascular function associated with many diseases, including cancer, ocular diseases, systemic inflammation, and cardiovascular diseases. The present study uses a computational signaling pathway model validated against experimental data to quantitatively study various mechanistic aspects of the angiopoietin-Tie signaling pathway, including receptor activation, trafficking, turnover, and molecular mechanisms of its regulation. The model provides mechanistic insights into the controversial role of Ang2 and its regulators vascular endothelial protein tyrosine phosphatase (VE-PTP) and Tie1 and predicts synergistic effects of inhibition of VE-PTP, Tie1, and Tie2 cleavage on enhancing the vascular protective actions of Tie2.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Christopher D Kontos
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Brian H Annex
- Department of Medicine and the Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
90
|
Zhang C, Guo F, Chang M, Zhou Z, Yi L, Gao C, Huang X, Huan J. Exosome-delivered syndecan-1 rescues acute lung injury via a FAK/p190RhoGAP/RhoA/ROCK/NF-κB signaling axis and glycocalyx enhancement. Exp Cell Res 2019; 384:111596. [PMID: 31487506 DOI: 10.1016/j.yexcr.2019.111596] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/30/2019] [Accepted: 09/01/2019] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is characterized by protein-rich pulmonary edema, critical hypoxemia, and influx of pro-inflammatory cytokines and cells. There are currently no effective pharmacon therapies in clinical practice. Syndecan-1 in endothelial cells has potential to protect barrier function of endothelium and suppress inflammation response. Thus, the present study was to identify whether exosomes with encapsulation of syndecan-1 could achieve ideal therapeutic effects in ALI. Exosomes were isolated from the conditional medium of lentivirus-transfected mouse pulmonary microvascular endothelial cells (MPMVECs) and characterized by nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), and western blotting. ALI mouse models were induced via intratracheal administration of lipopolysaccharide (LPS) and treated with exosomes. Lung edema, inflammation, and glycocalyx thickness were examined. The possible mechanism was verified by immunoblotting in MPMVECs. The purified exosomes included SDC1-high-Exos and SDC1-low-Exos which loaded with up-regulated syndecan-1 and down-regulated syndecan-1 respectively. Compared with SDC1-low-Exos, administration of SDC1-high-Exos could ameliorate lung edema and inflammation, attenuate number of cells and protein levels in bronchoalveolar lavage fluid (BALF), and preserve glycocalyx. Furthermore, SDC1-high-Exos also mitigated the expression of pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6 following LPS challenge. In MPMVECs, SDC1-high-Exos decreased stress fiber formation and ameliorated monolayer hyper-permeability after LPS stimulation. Western blotting analysis demonstrated that FAK/p190RhoGAP/RhoA/ROCK/NF-κB signaling pathway may be involved in LPS-induced ALI. In conclusion, SDC1-high-Exos play a pivotal role in ameliorating LPS-stimulated ALI models and may be served as a potential therapeutic agent for clinical application in the future.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Feng Guo
- Department of Plastic Surgery, Affiliated Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Mengling Chang
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Zengding Zhou
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Lei Yi
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Chengjin Gao
- Emergency Department, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaoqin Huang
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China.
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China.
| |
Collapse
|
91
|
Abstract
Objective: The endothelial glycocalyx (eGC) is a dynamic and multicomponent layer of macromolecules found at the surface of vascular endothelium, which is largely underappreciated. It has recently been recognized that eGC is a major regulator of endothelial function and may have therapeutic value in organ injuries. This study aimed to explore the role of the eGC in various pathologic and physiologic conditions, by reviewing the basic research findings pertaining to the detection of the eGC and its clinical significance. We also explored different pharmacologic agents used to protect and rebuild the eGC. Data sources: An in-depth search was performed in the PubMed database, focusing on research published after 2003 with keywords including eGC, permeability, glycocalyx and injuries, and glycocalyx protection. Study selection: Several authoritative reviews and original studies were identified and reviewed to summarize the characteristics of the eGC under physiologic and pathologic conditions as well as the detection and protection of the eGC. Results: The eGC degradation is closely associated with pathophysiologic changes such as vascular permeability, edema formation, mechanotransduction, and clotting cascade, together with neutrophil and platelet adhesion in diverse injury and disease states including inflammation (sepsis and trauma), ischemia-reperfusion injury, shock, hypervolemia, hypertension, hyperglycemia, and high Na+ as well as diabetes and atherosclerosis. Therapeutic strategies for protecting and rebuilding the eGC should be explored through experimental test and clinical verifications. Conclusions: Disturbance of the eGC usually occurs at early stages of various clinical pathophysiologies which can be partly prevented and reversed by protecting and restoring the eGC. The eGC seems to be a promising diagnostic biomarker and therapeutic target in clinical settings.
Collapse
|
92
|
Hippensteel JA, Uchimido R, Tyler PD, Burke RC, Han X, Zhang F, McMurtry SA, Colbert JF, Lindsell CJ, Angus DC, Kellum JA, Yealy DM, Linhardt RJ, Shapiro NI, Schmidt EP. Intravenous fluid resuscitation is associated with septic endothelial glycocalyx degradation. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:259. [PMID: 31337421 PMCID: PMC6652002 DOI: 10.1186/s13054-019-2534-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Intravenous fluids, an essential component of sepsis resuscitation, may paradoxically worsen outcomes by exacerbating endothelial injury. Preclinical models suggest that fluid resuscitation degrades the endothelial glycocalyx, a heparan sulfate-enriched structure necessary for vascular homeostasis. We hypothesized that endothelial glycocalyx degradation is associated with the volume of intravenous fluids administered during early sepsis resuscitation. METHODS We used mass spectrometry to measure plasma heparan sulfate (a highly sensitive and specific index of systemic endothelial glycocalyx degradation) after 6 h of intravenous fluids in 56 septic shock patients, at presentation and after 24 h of intravenous fluids in 100 sepsis patients, and in two groups of non-infected patients. We compared plasma heparan sulfate concentrations between sepsis and non-sepsis patients, as well as between sepsis survivors and sepsis non-survivors. We used multivariable linear regression to model the association between volume of intravenous fluids and changes in plasma heparan sulfate. RESULTS Consistent with previous studies, median plasma heparan sulfate was elevated in septic shock patients (118 [IQR, 113-341] ng/ml 6 h after presentation) compared to non-infected controls (61 [45-79] ng/ml), as well as in a second cohort of sepsis patients (283 [155-584] ng/ml) at emergency department presentation) compared to controls (177 [144-262] ng/ml). In the larger sepsis cohort, heparan sulfate predicted in-hospital mortality. In both cohorts, multivariable linear regression adjusting for age and severity of illness demonstrated a significant association between volume of intravenous fluids administered during resuscitation and plasma heparan sulfate. In the second cohort, independent of disease severity and age, each 1 l of intravenous fluids administered was associated with a 200 ng/ml increase in circulating heparan sulfate (p = 0.006) at 24 h after enrollment. CONCLUSIONS Glycocalyx degradation occurs in sepsis and septic shock and is associated with in-hospital mortality. The volume of intravenous fluids administered during sepsis resuscitation is independently associated with the degree of glycocalyx degradation. These findings suggest a potential mechanism by which intravenous fluid resuscitation strategies may induce iatrogenic endothelial injury.
Collapse
Affiliation(s)
| | - Ryo Uchimido
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Patrick D Tyler
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ryan C Burke
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Xiaorui Han
- Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Fuming Zhang
- Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Sarah A McMurtry
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - James F Colbert
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | | | - Derek C Angus
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John A Kellum
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald M Yealy
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert J Linhardt
- Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Nathan I Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Eric P Schmidt
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA. .,Department of Medicine, Denver Health Medical Center, Denver, CO, USA.
| |
Collapse
|
93
|
Huang X, Zhang X, Zhao DX, Yin J, Hu G, Evans CE, Zhao YY. Endothelial Hypoxia-Inducible Factor-1α Is Required for Vascular Repair and Resolution of Inflammatory Lung Injury through Forkhead Box Protein M1. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1664-1679. [PMID: 31121134 PMCID: PMC6680254 DOI: 10.1016/j.ajpath.2019.04.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/03/2019] [Accepted: 04/18/2019] [Indexed: 12/30/2022]
Abstract
Endothelial barrier dysfunction is a central factor in the pathogenesis of persistent lung inflammation and protein-rich edema formation, the hallmarks of acute respiratory distress syndrome. However, little is known about the molecular mechanisms that are responsible for vascular repair and resolution of inflammatory injury after sepsis challenge. Herein, we show that hypoxia-inducible factor-1α (HIF-1α), expressed in endothelial cells (ECs), is the critical transcriptional factor mediating vascular repair and resolution of inflammatory lung injury. After sepsis challenge, HIF-1α but not HIF-2α expression was rapidly induced in lung vascular ECs, and mice with EC-restricted disruption of Hif1α (Hif1af/f/Tie2Cre+) exhibited defective vascular repair, persistent inflammation, and increased mortality in contrast with the wild-type littermates after polymicrobial sepsis or endotoxemia challenge. Hif1af/f/Tie2Cre+ lungs exhibited marked decrease of EC proliferation during recovery after sepsis challenge, which was associated with inhibited expression of forkhead box protein M1 (Foxm1), a reparative transcription factor. Therapeutic restoration of endothelial Foxm1 expression, via liposomal delivery of Foxm1 plasmid DNA to Hif1af/f/Tie2Cre+ mice, resulted in reactivation of the vascular repair program and improved survival. Together, our studies, for the first time, delineate the essential role of endothelial HIF-1α in driving the vascular repair program. Thus, therapeutic activation of HIF-1α-dependent vascular repair may represent a novel and effective therapy to treat inflammatory vascular diseases, such as sepsis and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Xiaojia Huang
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; Division of Critical Care, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Xianming Zhang
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; Division of Critical Care, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - David X Zhao
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois; Department of Medicine, University of Chicago, Chicago, Illinois
| | - Jun Yin
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois
| | - Colin E Evans
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; Division of Critical Care, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; Division of Critical Care, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
94
|
Dekker NAM, van Leeuwen ALI, van Strien WWJ, Majolée J, Szulcek R, Vonk ABA, Hordijk PL, Boer C, van den Brom CE. Microcirculatory perfusion disturbances following cardiac surgery with cardiopulmonary bypass are associated with in vitro endothelial hyperpermeability and increased angiopoietin-2 levels. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:117. [PMID: 30975180 PMCID: PMC6460737 DOI: 10.1186/s13054-019-2418-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/01/2019] [Indexed: 12/31/2022]
Abstract
Background Endothelial hyperpermeability following cardiopulmonary bypass (CPB) contributes to microcirculatory perfusion disturbances and postoperative complications after cardiac surgery. We investigated the postoperative course of renal and pulmonary endothelial barrier function and the association with microcirculatory perfusion and angiopoietin-2 levels in patients after CPB. Methods Clinical data, sublingual microcirculatory data, and plasma samples were collected from patients undergoing coronary artery bypass graft surgery with CPB (n = 17) before and at several time points up to 72 h after CPB. Renal and pulmonary microvascular endothelial cells were incubated with patient plasma, and in vitro endothelial barrier function was assessed using electric cell–substrate impedance sensing. Plasma levels of angiopoietin-1,-2, and soluble Tie2 were measured, and the association with in vitro endothelial barrier function and in vivo microcirculatory perfusion was determined. Results A plasma-induced reduction of renal and pulmonary endothelial barrier function was observed in all samples taken within the first three postoperative days (P < 0.001 for all time points vs. pre-CPB). Angiopoietin-2 and soluble Tie2 levels increased within 72 h after CPB (5.7 ± 4.4 vs. 1.7 ± 0.4 ng/ml, P < 0.0001; 16.3 ± 4.7 vs. 11.9 ± 1.9 ng/ml, P = 0.018, vs. pre-CPB), whereas angiopoietin-1 remained stable. Interestingly, reduced in vitro renal and pulmonary endothelial barrier moderately correlated with reduced in vivo microcirculatory perfusion after CPB (r = 0.47, P = 0.005; r = 0.79, P < 0.001). In addition, increased angiopoietin-2 levels moderately correlated with reduced in vitro renal and pulmonary endothelial barrier (r = − 0.46, P < 0.001; r = − 0.40, P = 0.005) and reduced in vivo microcirculatory perfusion (r = − 0.43, P = 0.01; r = − 0.41, P = 0.03). Conclusions CPB is associated with an impairment of in vitro endothelial barrier function that continues in the first postoperative days and correlates with reduced postoperative microcirculatory perfusion and increased circulating angiopoietin-2 levels. These results suggest that angiopoietin-2 is a biomarker for postoperative endothelial hyperpermeability, which may contribute to delayed recovery of microcirculatory perfusion after CPB. Trial registration NTR4212. Electronic supplementary material The online version of this article (10.1186/s13054-019-2418-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole A M Dekker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands. .,Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands. .,Amsterdam UMC, Vrije Universiteit Amsterdam, Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Anoek L I van Leeuwen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Willem W J van Strien
- Amsterdam UMC, Vrije Universiteit Amsterdam, Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jisca Majolée
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Robert Szulcek
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Pulmonology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Alexander B A Vonk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Peter L Hordijk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Christa Boer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Charissa E van den Brom
- Amsterdam UMC, Vrije Universiteit Amsterdam, Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Experimental Laboratory for Vital Signs, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
95
|
Park I, Kim M, Choe K, Song E, Seo H, Hwang Y, Ahn J, Lee SH, Lee JH, Jo YH, Kim K, Koh GY, Kim P. Neutrophils disturb pulmonary microcirculation in sepsis-induced acute lung injury. Eur Respir J 2019; 53:13993003.00786-2018. [PMID: 30635296 PMCID: PMC6437604 DOI: 10.1183/13993003.00786-2018] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 12/27/2018] [Indexed: 02/02/2023]
Abstract
The lung is highly vulnerable during sepsis, yet its functional deterioration accompanied by disturbances in the pulmonary microcirculation is poorly understood. This study aimed to investigate how the pulmonary microcirculation is distorted in sepsis-induced acute lung injury (ALI) and reveal the underlying cellular pathophysiologic mechanism. Using a custom-made intravital lung microscopic imaging system in a murine model of sepsis-induced ALI, we achieved direct real-time visualisation of the pulmonary microcirculation and circulating cells in vivo. We derived the functional capillary ratio (FCR) as a quantitative parameter for assessing the fraction of functional microvasculature in the pulmonary microcirculation and dead space. We identified that the FCR rapidly decreases in the early stage of sepsis-induced ALI. The intravital imaging revealed that this decrease resulted from the generation of dead space, which was induced by prolonged neutrophil entrapment within the capillaries. We further showed that the neutrophils had an extended sequestration time and an arrest-like dynamic behaviour, both of which triggered neutrophil aggregates inside the capillaries and arterioles. Finally, we found that Mac-1 (CD11b/CD18) was upregulated in the sequestered neutrophils and that a Mac-1 inhibitor restored the FCR and improved hypoxaemia. Using the intravital lung imaging system, we observed that Mac-1-upregulated neutrophil aggregates led to the generation of dead space in the pulmonary microcirculation that was recovered by a Mac-1 inhibitor in sepsis-induced ALI. Neutrophils induce dead space in the pulmonary microcirculation in sepsis-induced ALI, recovered by a Mac-1 inhibitorhttp://ow.ly/vUzO30nbUyU
Collapse
Affiliation(s)
- Inwon Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Dept of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam-si, Republic of Korea
| | - Mingyo Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Division of Rheumatology, Dept of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Kibaek Choe
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eunjoo Song
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Howon Seo
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yoonha Hwang
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jinhyo Ahn
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jae Hyuk Lee
- Dept of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam-si, Republic of Korea
| | - You Hwan Jo
- Dept of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam-si, Republic of Korea
| | - Kyuseok Kim
- Dept of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam-si, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,Joint lead authors
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Joint lead authors
| |
Collapse
|
96
|
Higher Plasma Endothelial Markers in Adults with Cystic Fibrosis Compared with Healthy Age-matched Control Subjects. Ann Am Thorac Soc 2019; 16:768-771. [PMID: 30889361 DOI: 10.1513/annalsats.201812-844rl] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
97
|
Carota IA, Kenig-Kozlovsky Y, Onay T, Scott R, Thomson BR, Souma T, Bartlett CS, Li Y, Procissi D, Ramirez V, Yamaguchi S, Tarjus A, Tanna CE, Li C, Eremina V, Vestweber D, Oladipupo SS, Breyer MD, Quaggin SE. Targeting VE-PTP phosphatase protects the kidney from diabetic injury. J Exp Med 2019; 216:936-949. [PMID: 30886059 PMCID: PMC6446875 DOI: 10.1084/jem.20180009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 11/10/2018] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetic nephropathy is a leading cause of kidney failure. VE-PTP phosphatase expression is increased in the endothelium of rodents with diabetes and hypertension. Genetic deletion of VE-PTP reduces kidney injury in diabetic mice, suggesting it may be a therapeutic target. Diabetic nephropathy is a leading cause of end-stage kidney failure. Reduced angiopoietin-TIE2 receptor tyrosine kinase signaling in the vasculature leads to increased vascular permeability, inflammation, and endothelial cell loss and is associated with the development of diabetic complications. Here, we identified a mechanism to explain how TIE2 signaling is attenuated in diabetic animals. Expression of vascular endothelial protein tyrosine phosphatase VE-PTP (also known as PTPRB), which dephosphorylates TIE2, is robustly up-regulated in the renal microvasculature of diabetic rodents, thereby reducing TIE2 activity. Increased VE-PTP expression was dependent on hypoxia-inducible factor transcriptional activity in vivo. Genetic deletion of VE-PTP restored TIE2 activity independent of ligand availability and protected kidney structure and function in a mouse model of severe diabetic nephropathy. Mechanistically, inhibition of VE-PTP activated endothelial nitric oxide synthase and led to nuclear exclusion of the FOXO1 transcription factor, reducing expression of pro-inflammatory and pro-fibrotic gene targets. In sum, we identify inhibition of VE-PTP as a promising therapeutic target to protect the kidney from diabetic injury.
Collapse
Affiliation(s)
- Isabel A Carota
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL.,Eli Lilly & Company, Biotechnology Discovery Research, Indianapolis, IN
| | - Yael Kenig-Kozlovsky
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Tuncer Onay
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rizaldy Scott
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Benjamin R Thomson
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Tomokazu Souma
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Christina S Bartlett
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Yanyang Li
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Daniele Procissi
- Department of Radiology and Biomedical Engineering, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Veronica Ramirez
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shinji Yamaguchi
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Antoine Tarjus
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Christine E Tanna
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Chengjin Li
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Vera Eremina
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | - Matthew D Breyer
- Eli Lilly & Company, Biotechnology Discovery Research, Indianapolis, IN
| | - Susan E Quaggin
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL .,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
98
|
Mirando AC, Shen J, Silva RLE, Chu Z, Sass NC, Lorenc VE, Green JJ, Campochiaro PA, Popel AS, Pandey NB. A collagen IV-derived peptide disrupts α5β1 integrin and potentiates Ang2/Tie2 signaling. JCI Insight 2019; 4:122043. [PMID: 30668550 PMCID: PMC6478425 DOI: 10.1172/jci.insight.122043] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 01/11/2019] [Indexed: 01/08/2023] Open
Abstract
The angiopoietin (Ang)/Tie2 signaling pathway is essential for maintaining vascular homeostasis, and its dysregulation is associated with several diseases. Interactions between Tie2 and α5β1 integrin have emerged as part of this control; however, the mechanism is incompletely understood. AXT107, a collagen IV-derived peptide, has strong antipermeability activity and has enabled the elucidation of this previously undetermined mechanism. Previously, AXT107 was shown to inhibit VEGFR2 and other growth factor signaling via receptor tyrosine kinase association with specific integrins. AXT107 disrupts α5β1 and stimulates the relocation of Tie2 and α5 to cell junctions. In the presence of Ang2 and AXT107, junctional Tie2 is activated, downstream survival signals are upregulated, F-actin is rearranged to strengthen junctions, and, as a result, endothelial junctional permeability is reduced. These data suggest that α5β1 sequesters Tie2 in nonjunctional locations in endothelial cell membranes and that AXT107-induced disruption of α5β1 promotes clustering of Tie2 at junctions and converts Ang2 into a strong agonist, similar to responses observed when Ang1 levels greatly exceed those of Ang2. The potentiation of Tie2 activation by Ang2 even extended to mouse models in which AXT107 induced Tie2 phosphorylation in a model of hypoxia and inhibited vascular leakage in an Ang2-overexpression transgenic model and an LPS-induced inflammation model. Because Ang2 levels are very high in ischemic diseases, such as diabetic macular edema, neovascular age-related macular degeneration, uveitis, and cancer, targeting α5β1 with AXT107 provides a potentially more effective approach to treat these diseases.
Collapse
Affiliation(s)
| | - Jikui Shen
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Raquel Lima e Silva
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zenny Chu
- Department of Biomedical Engineering and
| | | | - Valeria E. Lorenc
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jordan J. Green
- Department of Biomedical Engineering and
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- AsclepiX Therapeutics, Inc., Baltimore, Maryland, USA
| | - Peter A. Campochiaro
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering and
- AsclepiX Therapeutics, Inc., Baltimore, Maryland, USA
| | - Niranjan B. Pandey
- Department of Biomedical Engineering and
- AsclepiX Therapeutics, Inc., Baltimore, Maryland, USA
| |
Collapse
|
99
|
A MST1-FOXO1 cascade establishes endothelial tip cell polarity and facilitates sprouting angiogenesis. Nat Commun 2019; 10:838. [PMID: 30783090 PMCID: PMC6381131 DOI: 10.1038/s41467-019-08773-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 01/28/2019] [Indexed: 12/29/2022] Open
Abstract
Hypoxia is a main driver of sprouting angiogenesis, but how tip endothelial cells are directed to hypoxic regions remains poorly understood. Here, we show that an endothelial MST1–FOXO1 cascade is essential for directional migration of tip cells towards hypoxic regions. In mice, endothelial‐specific deletion of either MST1 or FOXO1 leads to the loss of tip cell polarity and subsequent impairment of sprouting angiogenesis. Mechanistically, MST1 is activated by reactive oxygen species (ROS) produced in mitochondria in response to hypoxia, and activated MST1 promotes the nuclear import of FOXO1, thus augmenting its transcriptional regulation of polarity and migration‐associated genes. Furthermore, endothelial MST1‐FOXO1 cascade is required for revascularization and neovascularization in the oxygen-induced retinopathy model. Together, the results of our study delineate a crucial coupling between extracellular hypoxia and an intracellular ROS‐MST1‐FOXO1 cascade in establishing endothelial tip cell polarity during sprouting angiogenesis. Angiogenesis is driven by the directed migration of tip endothelial cells towards hypoxic tissues. Here, Kim et al. show that the generation of reactive oxygen species in endothelial cells upon hypoxia activates MST1, which subsequently promotes the nuclear translocation of FOXO1, and thus activates a pro-migratory transcriptional programme in endothelial tip cells.
Collapse
|
100
|
Increased gene copy number of DEFA1/DEFA3 worsens sepsis by inducing endothelial pyroptosis. Proc Natl Acad Sci U S A 2019; 116:3161-3170. [PMID: 30718392 PMCID: PMC6386704 DOI: 10.1073/pnas.1812947116] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sepsis claims an estimated 30 million episodes and 6 million deaths per year, and treatment options are rather limited. Human neutrophil peptides 1-3 (HNP1-3) are the most abundant neutrophil granule proteins but their neutrophil content varies because of unusually extensive gene copy number polymorphism. A genetic association study found that increased copy number of the HNP-encoding gene DEFA1/DEFA3 is a risk factor for organ dysfunction during sepsis development. However, direct experimental evidence demonstrating that these risk alleles are pathogenic for sepsis is lacking because the genes are present only in some primates and humans. Here, we generate DEFA1/DEFA3 transgenic mice with neutrophil-specific expression of the peptides. We show that mice with high copy number of DEFA1/DEFA3 genes have more severe sepsis-related vital organ damage and mortality than mice with low copy number of DEFA1/DEFA3 or wild-type mice, resulting from more severe endothelial barrier dysfunction and endothelial cell pyroptosis after sepsis challenge. Mechanistically, HNP-1 induces endothelial cell pyroptosis via P2X7 receptor-mediating canonical caspase-1 activation in a NLRP3 inflammasome-dependent manner. Based on these findings, we engineered a monoclonal antibody against HNP-1 to block the interaction with P2X7 and found that the blocking antibody protected mice carrying high copy number of DEFA1/DEFA3 from lethal sepsis. We thus demonstrate that DEFA1/DEFA3 copy number variation strongly modulates sepsis development in vivo and explore a paradigm for the precision treatment of sepsis tailored by individual genetic information.
Collapse
|