1
|
Li J, Hou H, Li J, Zhang K. Angiopoietins/Tie2 signaling axis and its role in angiogenesis of psoriasis. Acta Histochem 2025; 127:152228. [PMID: 39752990 DOI: 10.1016/j.acthis.2024.152228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 03/01/2025]
Abstract
Hyperplasia of microvessels in the superficial dermis is the main pathological feature of psoriasis, and is linked to the pathogenesis of psoriasis. Thus, anti-angiogenic therapy may be effective for psoriasis. Angiopoietins (Angs) are crucial angiogenic factors. Ang1 supports a static mature vascular phenotype, while Ang2 is associated with the formation of abnormal vascular structure, vascular leakage and inflammation. The Ang/Tie2 axis and its signal transduction play an important role in regulation of vascular stability, angiogenesis and inflammation. Targeting the Ang/Tie2 signal axis can normalize microvessels in psoriatic lesions. This paper reviews Ang/Tie2 signal axis and its role in angiogenesis of psoriasis, aiming to provide new ideas and strategies for anti-angiogenic therapy of psoriasis.
Collapse
Affiliation(s)
- Jiao Li
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, No. 5 Dong San Dao Xiang, Jiefang Road, Taiyuan, Shanxi Province 030009, China
| | - Hui Hou
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, No. 5 Dong San Dao Xiang, Jiefang Road, Taiyuan, Shanxi Province 030009, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, No. 5 Dong San Dao Xiang, Jiefang Road, Taiyuan, Shanxi Province 030009, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, No. 5 Dong San Dao Xiang, Jiefang Road, Taiyuan, Shanxi Province 030009, China.
| |
Collapse
|
2
|
Li G, Gao J, Ding P, Gao Y. The role of endothelial cell-pericyte interactions in vascularization and diseases. J Adv Res 2025; 67:269-288. [PMID: 38246244 PMCID: PMC11725166 DOI: 10.1016/j.jare.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Endothelial cells (ECs) and pericytes (PCs) are crucial components of the vascular system, with ECs lining the inner layer of blood vessels and PCs surrounding capillaries to regulate blood flow and angiogenesis. Intercellular communication between ECs and PCs is vital for the formation, stability, and function of blood vessels. Various signaling pathways, such as the vascular endothelial growth factor/vascular endothelial growth factor receptor pathway and the platelet-derived growth factor-B/platelet-derived growth factor receptor-β pathway, play roles in communication between ECs and PCs. Dysfunctional communication between these cells is associated with various diseases, including vascular diseases, central nervous system disorders, and certain types of cancers. AIM OF REVIEW This review aimed to explore the diverse roles of ECs and PCs in the formation and reshaping of blood vessels. This review focused on the essential signaling pathways that facilitate communication between these cells and investigated how disruptions in these pathways may contribute to disease. Additionally, the review explored potential therapeutic targets, future research directions, and innovative approaches, such as investigating the impact of EC-PCs in novel systemic diseases, addressing resistance to antiangiogenic drugs, and developing novel antiangiogenic medications to enhance therapeutic efficacy. KEY SCIENTIFIC CONCEPTS OF REVIEW Disordered EC-PC intercellular signaling plays a role in abnormal blood vessel formation, thus contributing to the progression of various diseases and the development of resistance to antiangiogenic drugs. Therefore, studies on EC-PC intercellular interactions have high clinical relevance.
Collapse
Affiliation(s)
- Gan Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
3
|
Dai Y, He J, He F, Chen Z, Jiang Y, Geng Y, Geng J, Zhou Y, Chen X, Li F, Wang Y, Mu X. Exposure to environmentally relevant levels of GenX affects placental and offspring development in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125294. [PMID: 39532251 DOI: 10.1016/j.envpol.2024.125294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Hexafluoropropylene Oxide Dimer Acid (GenX or HFPO-DA), a fluorochemical used in industrial applications such as non-stick coatings and water-repellent textiles, has emerged as a replacement for perfluorooctanoic acid (PFOA). Its widespread use has led to detection in air, soil, and drinking water, raising concerns about potential health impacts. This study investigates the effects of exposure to environmentally relevant levels of GenX on placental and offspring development in mice. Female mice were exposed to GenX for short-term (11 days) or subchronic (100 days) durations. Both short-term and subchronic exposure resulted in alterations in body weight gain, placental weight, and placental efficiency. Dams exposed subchronically also exhibit altered fetal weight and impaired placental structure. In the labyrinth layer of placenta, subchronic exposure to GenX caused disordered vasculature, characterized by enlarged vessel lumens, discontinuous signals of maternal (MCT1) and fetal (MCT4) vascular networks, and impaired mitochondria in fetal sinus endothelial cells. The placental mRNA profile revealed imbalanced expression of factors essential for proper angiogenesis, with increased levels of Vegfa and Angpt1, and decreased levels of Tie2. Additionally, male pups of the subchronically exposed dams exhibited higher birth weight, increased weight gain and reduced anogenital distance (AGD), while premature puberty onset was observed in female pups. This study provides the first evidence that subchronic exposure to environmentally relevant levels of GenX affects placental angiogenesis, potentially contributing to altered development in offspring. These findings offer new insights into the health impacts of GenX on reproductive development and raise concerns about its safety as a PFOA alternative.
Collapse
Affiliation(s)
- Yuhan Dai
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China; JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Junlin He
- JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Fei He
- JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Zhuxiu Chen
- JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yu Jiang
- JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanqing Geng
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China; JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jianwei Geng
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China; JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yongrui Zhou
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China; JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xuemei Chen
- JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Fangfang Li
- JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yingxiong Wang
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China; JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xinyi Mu
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China; JointInternational Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
4
|
Serlo M, Inkinen N, Lakkisto P, Valkonen M, Pulkkinen A, Selander T, Pettilä V, Vaara ST. Fluid bolus increases plasma hyaluronan concentration compared to follow-up strategy without a bolus in oliguric intensive care unit patients. Sci Rep 2024; 14:20808. [PMID: 39242877 PMCID: PMC11379687 DOI: 10.1038/s41598-024-71670-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
Fluid therapy is a fundamental part of supportive therapy in critical care. However, it is also a suspected risk for endothelial glycocalyx degradation which is associated with poor clinical outcomes. This secondary analysis of RESPONSE randomized trial compares the effect of follow-up strategy (FU) on endothelial biomarkers to that of 500 ml crystalloid fluid bolus (FB) in oliguric, hemodynamically optimized intensive care unit (ICU) patients. 130 adult subjects were enrolled in two Finnish ICUs from January 2017 to November 2020. Blood and urine samples of 63 patients in FU group and 67 patients in FB group were collected before and after the intervention and analyzed using enzyme-linked immunosorbent assays. Single fluid bolus, given after median of 3887 ml (interquartile range 2842; 5359 ml) resuscitation fluids in the preceding 24 h, increased plasma hyaluronan concentration compared to the follow-up strategy (difference in medians 29.2 ng/ml with 95% CI [14.5ng/ml; 55.5ng/ml], P < 0.001). No treatment effect was detected in the plasma levels of syndecan-1, , angiopoietin-2, angiopoietin receptors Tie2 and Tie1, or in soluble thrombomodulin in the adjusted median regression analysis. The increase in hyaluronan was independent of its simultaneous renal clearance but correlated moderately with the increase in endothelium-specific Tie1. The follow-up strategy did not show consistent endothelium-sparing effect but protected against hyaluronan increase. The mechanisms and consequences of hyaluronan fluctuations need further clarification. Trial registration: clinicaltrials.gov, NCT02860572. Registered 1 August 2016, https://www.clinicaltrials.gov/study/NCT02860572?term=NCT02860572&rank=1.
Collapse
Affiliation(s)
- Maija Serlo
- Department of Perioperative and Intensive Care, Intensive and Intermediate Care Unit, University of Helsinki and Helsinki University Hospital, P.O. Box 320, 00290 HUS, Helsinki, Finland.
| | - Nina Inkinen
- Department of Anesthesia and Intensive Care, Hospital Nova of Central Finland, Central Finland Health Care District, Hoitajantie 3, 40620, Jyväskylä, Finland
| | - Päivi Lakkisto
- Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Miia Valkonen
- Department of Perioperative and Intensive Care, Intensive and Intermediate Care Unit, University of Helsinki and Helsinki University Hospital, P.O. Box 320, 00290 HUS, Helsinki, Finland
| | - Anni Pulkkinen
- Department of Anesthesia and Intensive Care, Hospital Nova of Central Finland, Central Finland Health Care District, Hoitajantie 3, 40620, Jyväskylä, Finland
| | - Tuomas Selander
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
| | - Ville Pettilä
- Department of Perioperative and Intensive Care, Intensive and Intermediate Care Unit, University of Helsinki and Helsinki University Hospital, P.O. Box 320, 00290 HUS, Helsinki, Finland
| | - Suvi T Vaara
- Department of Perioperative and Intensive Care, Intensive and Intermediate Care Unit, University of Helsinki and Helsinki University Hospital, P.O. Box 320, 00290 HUS, Helsinki, Finland
| |
Collapse
|
5
|
Al-Ostoot FH, Salah S, Khanum SA. An Overview of Cancer Biology, Pathophysiological Development and It's Treatment Modalities: Current Challenges of Cancer anti-Angiogenic Therapy. Cancer Invest 2024; 42:559-604. [PMID: 38874308 DOI: 10.1080/07357907.2024.2361295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 11/22/2021] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
A number of conditions and factors can cause the transformation of normal cells in the body into malignant tissue by changing the normal functions of a wide range of regulatory, apoptotic, and signal transduction pathways. Despite the current deficiency in fully understanding the mechanism of cancer action accurately and clearly, numerous genes and proteins that are causally involved in the initiation, progression, and metastasis of cancer have been identified. But due to the lack of space and the abundance of details on this complex topic, we have emphasized here more recent advances in our understanding of the principles implied tumor cell transformation, development, invasion, angiogenesis, and metastasis. Inhibition of angiogenesis is a significant strategy for the treatment of various solid tumors, that essentially depend on cutting or at least limiting the supply of blood to micro-regions of tumors, leading to pan-hypoxia and pan-necrosis inside solid tumor tissues. Researchers have continued to enhance the efficiency of anti-angiogenic drugs over the past two decades, to identify their potential in the drug interaction, and to discover reasonable interpretations for possible resistance to treatment. In this review, we have discussed an overview of cancer history and recent methods use in cancer therapy, focusing on anti-angiogenic inhibitors targeting angiogenesis formation. Further, this review has explained the molecular mechanism of action of these anti-angiogenic inhibitors in various tumor types and their limitations use. In addition, we described the synergistic mechanisms of immunotherapy and anti-angiogenic therapy and summarizes current clinical trials of these combinations. Many phase III trials found that combining immunotherapy and anti-angiogenic therapy improved survival. Therefore, targeting the source supply of cancer cells to grow and spread with new anti-angiogenic agents in combination with different conventional therapy is a novel method to reduce cancer progression. The aim of this paper is to overview the varying concepts of cancer focusing on mechanisms involved in tumor angiogenesis and provide an overview of the recent trends in anti-angiogenic strategies for cancer therapy.
Collapse
Affiliation(s)
- Fares Hezam Al-Ostoot
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
- Department of Biochemistry, Faculty of Education & Science, Albaydha University, Al-Baydha, Yemen
| | - Salma Salah
- Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
| |
Collapse
|
6
|
Gendy N, Brown L, Staunton MK, Garg K, Hernandez Garcilazo N, Qian L, Yamamoto Y, Ugwuowo U, Obeid W, Al-Qusairi L, Bostom A, Mansour SG. The Role of Angiopoietins in Cardiovascular Outcomes of Kidney Transplant Recipients: An Ancillary Study from the FAVORIT. Am J Nephrol 2024; 55:597-606. [PMID: 38735283 PMCID: PMC11444892 DOI: 10.1159/000538878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/05/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION Kidney transplant recipients (KTRs) have increased risk of cardiovascular disease (CVD) mortality. We investigated vascular biomarkers, angiopoietin-1, and angiopoietin-2 (angpt-1, -2), in CVD development in KTRs. METHODS This ancillary study from the FAVORIT evaluates the associations of baseline plasma angpt-1, -2 levels in CVD development (primary outcome) and graft failure (GF) and death (secondary outcomes) in 2000 deceased donor KTRs. We used Cox regression to analyze the association of biomarker quartiles with outcomes. We adjusted for demographic; CVD- and transplant-related variables; medications; urine albumin-to-creatinine ratio; and randomization status. We calculated areas under the curves (AUCs) to predict CVD or death, and GF or death by incorporating biomarkers alongside clinical variables. RESULTS Participants' median age was 52 IQR [45, 59] years: with 37% women and 73% identifying as white. Median time from transplantation was 3.99 IQR [1.58, 7.93] years and to CVD development was 2.54 IQR [1.11-3.80] years. Quartiles of angpt-1 were not associated with outcomes. Whereas higher levels of angpt-2 (quartile 4) were associated with about 2 times the risk of CVD, GF, and death (aHR 1.85 [1.25-2.73], p < 0.01; 2.24 [1.36-3.70)], p < 0.01; 2.30 [1.48-3.58], p < 0.01, respectively) as compared to quartile 1. Adding angiopoietins to preexisting clinical variables improved prediction of CVD or death (AUC improved from 0.70 to 0.72, p = 0.005) and GF or death (AUC improved from 0.68 to 0.70, p = 0.005). Angpt-2 may partially explain the increased risk of future CVD in KTRs. Further research is needed to assess the utility of using angiopoietins in the clinical care of KTRs. CONCLUSION Angpt-2 may be a useful prognostic tool for future CVD in KTRs. Combining angiopoietins with clinical markers may tailor follow-up to mitigate CVD risk.
Collapse
Affiliation(s)
- Natalie Gendy
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA,
- Schulich School of Medicine, Western University, London, Ontario, Canada,
| | - Liam Brown
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA
| | - Mary Kate Staunton
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA
| | - Kanika Garg
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA
| | | | - Long Qian
- Section of Nephrology, Yale New Haven Hospital, New Haven, Connecticut, USA
| | - Yu Yamamoto
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA
| | - Ugochukwu Ugwuowo
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA
| | - Wassim Obeid
- Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Andrew Bostom
- Department of Family Medicine, Brown University, Providence, Rhode Island, USA
| | - Sherry G Mansour
- Clinical and Translational Research Accelerator, New Haven, Connecticut, USA
- Section of Nephrology, Yale New Haven Hospital, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Bowman C, Rockson SG. Genetic causes of lymphatic disorders: recent updates on the clinical and molecular aspects of lymphatic disease. Curr Opin Cardiol 2024; 39:170-177. [PMID: 38483006 DOI: 10.1097/hco.0000000000001116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW The lymphatic system facilitates several key functions that limit significant morbidity and mortality. Despite the impact and burden of lymphatic disorders, there are many remaining disorders whose genetic substrate remains unknown. The purpose of this review is to provide an update on the genetic causes of lymphatic disorders, while reporting on newly proposed clinical classifications of lymphatic disease. RECENT FINDINGS We reviewed several new mutations in genes that have been identified as potential causes of lymphatic disorders including: MDFIC, EPHB 4 , and ANGPT2. Furthermore, the traditional St. George's Classification system for primary lymphatic anomalies has been updated to reflect the use of genetic testing, both as a tool for the clinical identification of lymphatic disease and as a method through which new sub-classifications of lymphatic disorders have been established within this framework. Finally, we highlighted recent clinical studies that have explored the impact of therapies such as sirolimus, ketoprofen, and acebilustat on lymphatic disorders. SUMMARY Despite a growing body of evidence, current literature demonstrates a persistent gap in the number of known genes responsible for lymphatic disease entities. Recent clinical classification tools have been introduced in order to integrate traditional symptom- and time-based diagnostic approaches with modern genetic classifications, as highlighted in the updated St. George's classification system. With the introduction of this novel approach, clinicians may be better equipped to recognize established disease and, potentially, to identify novel causal mutations. Further research is needed to identify additional genetic causes of disease and to optimize current clinical tools for diagnosis and treatment.
Collapse
Affiliation(s)
- Catharine Bowman
- Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
8
|
Wu Y, Zhang J, Wang X, Xu Y, Zheng J. Saikosaponin-d regulates angiogenesis in idiopathic pulmonary fibrosis through angiopoietin/Tie-2 pathway. Acta Histochem 2023; 125:152100. [PMID: 37837833 DOI: 10.1016/j.acthis.2023.152100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/06/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is considered as a chronic interstitial lung disease with underlying mechanism of IPF remaining unclear, while there are no definitive treatment options. In recent years, scientists have gradually paid attention to the influence of angiogenesis on IPF. Because IPF is a progressive with microvascular remodeling disorder, scientists have postulated that angiogenesis may also be one of the initiating and contributing factors of the disease. Bupleurum is a common natural Chinese herbal medicine with antibacterial, anti-inflammatory, anti-tumor and other pharmacological effects. As the most important active monomer of Bupleurum, Saikosaponin-d (SSd) is a new discovery with anti-pulmonary fibrosis (PF) activity. This study attempts to investigate the role of SSd in the interference of PF through regulation of angiogenesis in IPF through Angiopoietin (Angpt) /Tie receptor 2 (Tie2) pathway. METHODS Randomly, we allocated C57BL/6 mice into four groups (n = 20 in each group). Afterwards, establishment of IPF model was accomplished via intratracheal administration of bleomycin (BLM, 5 mg/kg), while corresponding drug intervention was given accordingly. On 3rd, 7th, 14th and 28th days after modeling, we performed histopathological examination through staining. Meanwhile, immunohistochemistry (IHC) of PF and the expression of related factors were observed, while Ang/Tie2 pathway was assessed by ELISA with the effect of SSd on angiogenesis related proteins in IPF being explored with IHC and Western Blot technique. RESULTS Our results showed that SSd could reduce inflammation and PF levels in lung tissue of experimental mice, while levels of angiogenesis-related factors, namely Tie-2, Ang-1 and ANGPT2 (Ang-2), fibrosis- associated factors like Alpha-smooth muscle actin (α-SMA), collagen-I and hydroxyproline in SSd and dexamethasone (DXM) mice were significantly reduced at each time point compared to BLM (p < 0.01). Additionally, we discovered substantial decreased expressions of Ang-1, Ang-2, Tie-2, α-SMA and collagen-I at protein level in SSd and DXM mice at each time point compared to BLM (p < 0.05). Besides, insignificant differences were observed between SSd and DXM groups (p > 0.05). CONCLUSION This study has demonstrated that SSd could down-regulate the expression of ANG-1, Ang-2 and Tie2 in the Ang/Tie2 pathway, and may reduce lung inflammation and PF in BLM-induced mice via inhibition of angiogenesis.
Collapse
Affiliation(s)
- Yan Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi City, Jiangsu 214122, China
| | - Jun Zhang
- Department of Respiratory and Critical Care Medicine, Aoyang Hospital Affiliated to Jiangsu University, 279 Jingang Dadao, Zhangjiagang City, Jiangsu 215631, China
| | - Xintian Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Jingkou District, Zhenjiang City, Jiangsu 212000, China
| | - Yuncong Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Jingkou District, Zhenjiang City, Jiangsu 212000, China
| | - Jinxu Zheng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Jingkou District, Zhenjiang City, Jiangsu 212000, China.
| |
Collapse
|
9
|
Zhang Y, Xu S, Jiang F, Hu M, Han Y, Wang Y, Liu Z. A comprehensive insight into the role of molecular pathways affected by the Angiopoietin and Tie system involved in hematological malignancies' pathogenesis. Pathol Res Pract 2023; 248:154677. [PMID: 37467636 DOI: 10.1016/j.prp.2023.154677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/15/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023]
Abstract
Angiogenesis has been recognized as a critical factor in developing solid tumors and hematological malignancies. How angiogenesis affects the molecular pathways in malignancies is still a mystery. The angiopoietin family, one of the known molecular mediators for angiogenesis, encourages angiogenesis by attaching to Tie receptors on cell surfaces. Angiopoietin, Tie, and particularly the molecular pathways they mediate have all been the subject of recent studies that have established their diagnostic, prognostic, and therapeutic potential. Here, we've reviewed the function of molecular pathways impacted by the Angiogenin and Tie system in hematological malignancies.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
10
|
Chen S, Wang Y, Kong L, Ji Y, Cui J, Shen W. Role of UDP-glucose ceramide glucosyltransferase in venous malformation. Front Cell Dev Biol 2023; 11:1178045. [PMID: 37274734 PMCID: PMC10235597 DOI: 10.3389/fcell.2023.1178045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/11/2023] [Indexed: 06/06/2023] Open
Abstract
Venous malformation (VM) results from the abnormal growth of the vasculature; however, the detailed molecular mechanism remains unclear. As a glycosyltransferase, UDP-glucose ceramide glucosyltransferase (UGCG) is localized to the Golgi body and is a key enzyme in the first step of glycosphingolipid synthesis. Here, we aimed to explore the relationship between UGCG and the development of VM. First, investigations using RT-qPCR and Western blotting on the diseased vasculature of VM patients and normal vascular tissues revealed that UGCG expression was markedly elevated in the diseased vessels. Subsequently, immunofluorescence assay showed that UGCG was co-localized with CD31, an endothelial cell marker, in tissues from patients with VM and healthy subjects. Then, we established TIE2-L914F-mutant human umbilical vein endothelial cells (HUVECs) by lentivirus transfection. Next, Western blotting revealed that UGCG expression was considerably higher in HUVECsTIE2-L914F. In addition, we established a UGCG-overexpressing HUVECs line by plasmid transfection. With the CCK8 cell proliferation experiment, wound healing assay, and tube formation assay, we found that UGCG could promote the proliferation, migration, and tube formation activity of HUVECs, whereas the inhibition of UGCG could inhibit the proliferation, migration, and tube formation activity of HUVECsTIE2-L914F. Finally, Western blotting revealed that UGCG regulates the AKT/mTOR pathway in HUVECs. These data demonstrated that UGCG can affect the activity of vascular endothelial cells and regulate the AKT/mTOR signaling pathway; this is a potential mechanism underlying VM pathogenesis.
Collapse
|
11
|
Liu X, Liang H, Fang H, Xiao J, Yang C, Zhou Z, Feng J, Chen C. Angiopoietin-1 promotes triple-negative breast cancer cell proliferation by upregulating carboxypeptidase A4. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1487-1495. [PMID: 37162264 PMCID: PMC10520468 DOI: 10.3724/abbs.2023082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/02/2023] [Indexed: 05/11/2023] Open
Abstract
Angiopoietin-1 (ANG1) is a pro-angiogenic regulator that contributes to the progression of solid tumors by stimulating the proliferation, migration and tube formation of vascular endothelial cells, as well as the renewal and stability of blood vessels. However, the functions and mechanisms of ANG1 in triple-negative breast cancer (TNBC) are unclear. The clinical sample database shows that a higher level of ANG1 in TNBC is associated with poor prognosis compared to non-TNBC. In addition, knockdown of ANG1 inhibits TNBC cell proliferation and induces cell cycle G1 phase arrest and apoptosis. Overexpression of ANG1 promotes tumor growth in nude mice. Mechanistically, ANG1 promotes TNBC by upregulating carboxypeptidase A4 (CPA4) expression. Overall, the ANG1-CPA4 axis can be a therapeutic target for TNBC.
Collapse
Affiliation(s)
- Xue Liu
- Medical CollegeAnhui University of Science and TechnologyHuainan232001China
- Department of Laboratory Medicine & Central LaboratoryFengxian District Central Hospital of ShanghaiShanghai201499China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
| | - Huichun Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
| | - Huan Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
| | - Ji Xiao
- College of Life Science and TechnologyGuangzhou Jinan Biomedicine Research and Development CenterJinan UniversityGuangzhou510632China
| | - Chuanyu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
| | - Zhongmei Zhou
- The School of Continuing EducationKunming Medical UniversityKunming650500China
| | - Jing Feng
- Department of Laboratory Medicine & Central LaboratoryFengxian District Central Hospital of ShanghaiShanghai201499China
- The Second Affiliated Hospital of the Chinese University of Hong KongShenzhen518172China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Academy of Biomedical EngineeringKunming Medical UniversityKunming650500China
- The Third Affiliated HospitalKunming Medical UniversityKunming650106China
| |
Collapse
|
12
|
Pericytes in the tumor microenvironment. Cancer Lett 2023; 556:216074. [PMID: 36682706 DOI: 10.1016/j.canlet.2023.216074] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
Pericytes are a type of mural cell located between the endothelial cells of capillaries and the basement membrane, which function to regulate the capillary vasomotor and maintain normal microcirculation of local tissues and organs and have been identified as a significant component in the tumor microenvironment (TME). Pericytes have various interactions with different components of the TME, such as constituting the pre-metastatic niche, promoting the growth of cancer cells and drug resistance through paracrine activity, and inducing M2 macrophage polarization. While changes in the TME can affect the number, phenotype, and molecular markers of pericytes. For example, pericyte detachment from endothelial cells in the TME facilitates tumor cells in situ to invade the circulating blood and is beneficial to local capillary basement membrane enzymatic hydrolysis and endothelial cell proliferation and budding, which contribute to tumor angiogenesis and metastasis. In this review, we discuss the emerging role of pericytes in the TME, and tumor treatment related to pericytes. This review aimed to provide a more comprehensive understanding of the function of pericytes and the relationship between pericytes and tumors and to provide ideas for the treatment and prevention of malignant tumors.
Collapse
|
13
|
Stritt S, Nurden P, Nurden AT, Schved JF, Bordet JC, Roux M, Alessi MC, Trégouët DA, Mäkinen T, Giansily-Blaizot M. APOLD1 loss causes endothelial dysfunction involving cell junctions, cytoskeletal architecture, and Weibel-Palade bodies, while disrupting hemostasis. Haematologica 2023; 108:772-784. [PMID: 35638551 PMCID: PMC9973481 DOI: 10.3324/haematol.2022.280816] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Vascular homeostasis is impaired in various diseases thereby contributing to the progression of their underlying pathologies. The endothelial immediate early gene Apolipoprotein L domain-containing 1 (APOLD1) helps to regulate endothelial function. However, its precise role in endothelial cell biology remains unclear. We have localized APOLD1 to endothelial cell contacts and to Weibel-Palade bodies (WPB) where it associates with von Willebrand factor (VWF) tubules. Silencing of APOLD1 in primary human endothelial cells disrupted the cell junction-cytoskeletal interface, thereby altering endothelial permeability accompanied by spontaneous release of WPB contents. This resulted in an increased presence of WPB cargoes, notably VWF and angiopoietin-2 in the extracellular medium. Autophagy flux, previously recognized as an essential mechanism for the regulated release of WPB, was impaired in the absence of APOLD1. In addition, we report APOLD1 as a candidate gene for a novel inherited bleeding disorder across three generations of a large family in which an atypical bleeding diathesis was associated with episodic impaired microcirculation. A dominant heterozygous nonsense APOLD1:p.R49* variant segregated to affected family members. Compromised vascular integrity resulting from an excess of plasma angiopoietin-2, and locally impaired availability of VWF may explain the unusual clinical profile of APOLD1:p.R49* patients. In summary, our findings identify APOLD1 as an important regulator of vascular homeostasis and raise the need to consider testing of endothelial cell function in patients with inherited bleeding disorders without apparent platelet or coagulation defects.
Collapse
Affiliation(s)
- Simon Stritt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala
| | - Paquita Nurden
- Institut de Rythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France.
| | - Alan T Nurden
- Institut de Rythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France
| | - Jean-François Schved
- Department of Biological Hematology, CHU Montpellier, Université de Montpellier, Montpellier
| | - Jean-Claude Bordet
- Hematology, Hospices civils de Lyon, Bron biology center and Hemostasis- Thrombosis, Lyon-1 University, Lyon
| | | | | | - David-Alexandre Trégouët
- Laboratory of Excellence GENMED (Medical Genomics), Paris; University of Bordeaux, INSERM, Bordeaux Population Health Research Center, U1219, Bordeaux
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Muriel Giansily-Blaizot
- Department of Biological Hematology, CHU Montpellier, Université de Montpellier, Montpellier
| |
Collapse
|
14
|
Poto R, Gambardella AR, Marone G, Schroeder JT, Mattei F, Schiavoni G, Varricchi G. Basophils from allergy to cancer. Front Immunol 2022; 13:1056838. [PMID: 36578500 PMCID: PMC9791102 DOI: 10.3389/fimmu.2022.1056838] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Human basophils, first identified over 140 years ago, account for just 0.5-1% of circulating leukocytes. While this scarcity long hampered basophil studies, innovations during the past 30 years, beginning with their isolation and more recently in the development of mouse models, have markedly advanced our understanding of these cells. Although dissimilarities between human and mouse basophils persist, the overall findings highlight the growing importance of these cells in health and disease. Indeed, studies continue to support basophils as key participants in IgE-mediated reactions, where they infiltrate inflammatory lesions, release pro-inflammatory mediators (histamine, leukotriene C4: LTC4) and regulatory cytokines (IL-4, IL-13) central to the pathogenesis of allergic diseases. Studies now report basophils infiltrating various human cancers where they play diverse roles, either promoting or hampering tumorigenesis. Likewise, this activity bears remarkable similarity to the mounting evidence that basophils facilitate wound healing. In fact, both activities appear linked to the capacity of basophils to secrete IL-4/IL-13, with these cytokines polarizing macrophages toward the M2 phenotype. Basophils also secrete several angiogenic factors (vascular endothelial growth factor: VEGF-A, amphiregulin) consistent with these activities. In this review, we feature these newfound properties with the goal of unraveling the increasing importance of basophils in these diverse pathobiological processes.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy,World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy
| | - Adriana Rosa Gambardella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy,Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), Naples, Italy
| | - John T. Schroeder
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University, Baltimore, MD, United States
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy,*Correspondence: Gilda Varricchi, ; Giovanna Schiavoni,
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy,Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), Naples, Italy,*Correspondence: Gilda Varricchi, ; Giovanna Schiavoni,
| |
Collapse
|
15
|
Baquet-Walscheid K, Wildschütz L, Kasper M, Busch M, Thanos S, Bauer D, Stoll M, König S, Heiligenhaus A. Assessment of angiogenesis-related parameters in juvenile idiopathic arthritis-associated uveitis. Mol Biol Rep 2022; 49:6093-6102. [PMID: 35359237 DOI: 10.1007/s11033-022-07398-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/17/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Juvenile idiopathic arthritis-associated uveitis (JIAU) may run a chronic and treatment-resistant course, and occasionally, alterations of the iris vasculature may be observed clinically. METHODS Iris tissue (IT), aqueous humor (AH) and serum samples from patients with clinically inactive JIAU (n = 30), acute anterior uveitis (AAU; n = 18), and primary open angle glaucoma (POAG; n = 20) were obtained during trabeculectomy or cataract surgery. Samples were analyzed by RNA-Seq, qRT-PCR, LC-IMS, Western-Blot, and LEGENDplex™ analysis. Pattern of iris vasculature in JIAU patients was assessed qualitatively via fluorescein and indocyanine green angiography (FLA/ICGA). RESULTS RNA-Seq of IT showed significantly differential expression (DE) of 136 genes between JIAU and POAG, of which 15 were associated with angiogenesis. qRT-PCR, performed to validate RNA-Seq results, showed upregulation of the angiogenesis-related genes Kdr, Angpt-1, Tie-1, Tie-2 and Mmrn2 in IT (JIAU vs POAG, p > 0.05). LC-IMS of IT revealed a total number of 56 DE proteins (JIAU vs POAG), of which Angiopoetin, Lumican and Decorin were associated with angiogenesis and showed increased (p > 0.05) expression on Western-Blot analysis. LEGENDplex™ analysis showed upregulation of ANGPT-2 in AH from JIAU compared to AAU and POAG, whereas VEGF was upregulated in AAU. Iris vascular leakage, hypoperfusion and neovascularization were observed by FLA/ICGA in JIA patients with treatment-refractory complicated course of uveitis. CONCLUSION Angiogenesis-related factors could play a role in long-standing complicated JIAU, leading to clinically visible alterations in selected cases.
Collapse
Affiliation(s)
- Karoline Baquet-Walscheid
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany. .,University of Duisburg-Essen, Essen, Germany.
| | - Lena Wildschütz
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Maren Kasper
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Martin Busch
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Solon Thanos
- Institute of Experimental Ophthalmology, University of Muenster, Münster, Germany
| | - Dirk Bauer
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Monika Stoll
- Core Facility Genomics, University of Muenster, Münster, Germany.,Department "Genetic Epidemiology", Institute of Human Genetics, University of Münster, Münster, Germany
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, University of Muenster, Münster, Germany
| | - Arnd Heiligenhaus
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany.,University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
16
|
Mansour SG, Bhatraju PK, Coca SG, Obeid W, Wilson FP, Stanaway IB, Jia Y, Thiessen-Philbrook H, Go AS, Ikizler TA, Siew ED, Chinchilli VM, Hsu CY, Garg AX, Reeves WB, Liu KD, Kimmel PL, Kaufman JS, Wurfel MM, Himmelfarb J, Parikh SM, Parikh CR. Angiopoietins as Prognostic Markers for Future Kidney Disease and Heart Failure Events after Acute Kidney Injury. J Am Soc Nephrol 2022; 33:613-627. [PMID: 35017169 PMCID: PMC8975075 DOI: 10.1681/asn.2021060757] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/15/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The mechanisms underlying long-term sequelae after AKI remain unclear. Vessel instability, an early response to endothelial injury, may reflect a shared mechanism and early trigger for CKD and heart failure. METHODS To investigate whether plasma angiopoietins, markers of vessel homeostasis, are associated with CKD progression and heart failure admissions after hospitalization in patients with and without AKI, we conducted a prospective cohort study to analyze the balance between angiopoietin-1 (Angpt-1), which maintains vessel stability, and angiopoietin-2 (Angpt-2), which increases vessel destabilization. Three months after discharge, we evaluated the associations between angiopoietins and development of the primary outcomes of CKD progression and heart failure and the secondary outcome of all-cause mortality 3 months after discharge or later. RESULTS Median age for the 1503 participants was 65.8 years; 746 (50%) had AKI. Compared with the lowest quartile, the highest quartile of the Angpt-1:Angpt-2 ratio was associated with 72% lower risk of CKD progression (adjusted hazard ratio [aHR], 0.28; 95% confidence interval [CI], 0.15 to 0.51), 94% lower risk of heart failure (aHR, 0.06; 95% CI, 0.02 to 0.15), and 82% lower risk of mortality (aHR, 0.18; 95% CI, 0.09 to 0.35) for those with AKI. Among those without AKI, the highest quartile of Angpt-1:Angpt-2 ratio was associated with 71% lower risk of heart failure (aHR, 0.29; 95% CI, 0.12 to 0.69) and 68% less mortality (aHR, 0.32; 95% CI, 0.15 to 0.68). There were no associations with CKD progression. CONCLUSIONS A higher Angpt-1:Angpt-2 ratio was strongly associated with less CKD progression, heart failure, and mortality in the setting of AKI.
Collapse
Affiliation(s)
- Sherry G Mansour
- Clinical Translational Research Accelerator, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut.,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Pavan K Bhatraju
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington.,Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Steven G Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wassim Obeid
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Francis P Wilson
- Clinical Translational Research Accelerator, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut.,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Ian B Stanaway
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Yaqi Jia
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Alan S Go
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, California.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California.,Department of Health Research and Policy, Stanford University, Palo Alto, California.,Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - T Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Edward D Siew
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Vernon M Chinchilli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chi-Yuan Hsu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, California.,Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Amit X Garg
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,ICES, Ontario, Canada
| | - W Brian Reeves
- Division of Nephrology, Department of Medicine, University of Texas Joe and Teresa Long School of Medicine, San Antonio, Texas
| | - Kathleen D Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, California.,Department of Anesthesia, Division of Critical Care Medicine, University of California, San Francisco, San Francisco, California
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - James S Kaufman
- Division of Nephrology, Veterans Affairs New York Harbor Healthcare System and New York University School of Medicine, New York, New York
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington.,Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Samir M Parikh
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | |
Collapse
|
17
|
Ali F, Fang HL, Shah FA, Muhammad SA, Khan A, Li S. Reprofiling analysis of FDA approved drugs with upregulated differential expression genes found in hypertension. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
18
|
Abstract
Brain arteriovenous malformation (bAVM) is the most common cause of intracranial hemorrhage (ICH), particularly in young patients. However, the exact cause of bAVM bleeding and rupture is not yet fully understood. In bAVMs, blood bypasses the entire capillary bed and directly flows from arteries to veins. The vessel walls in bAVMs have structural defects, which impair vascular integrity. Mural cells are essential structural and functional components of blood vessels and play a critical role in maintaining vascular integrity. Changes in mural cell number and coverage have been implicated in bAVMs. In this review, we discussed the roles of mural cells in bAVM pathogenesis. We focused on 1) the recent advances in human and animal studies of bAVMs; 2) the importance of mural cells in vascular integrity; 3) the regulatory signaling pathways that regulate mural cell function. More specifically, the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-β (PDGFR-β), EphrinB2/EphB4, and angiopoietins/tie2 signaling pathways that regulate mural cell-recruitment during vascular remodeling were discussed in detail.
Collapse
|
19
|
Conditional Mutation of Hand1 in the Mouse Placenta Disrupts Placental Vascular Development Resulting in Fetal Loss in Both Early and Late Pregnancy. Int J Mol Sci 2021; 22:ijms22179532. [PMID: 34502440 PMCID: PMC8431056 DOI: 10.3390/ijms22179532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/18/2022] Open
Abstract
Congenital heart defects (CHD) affect approximately 1% of all live births, and often require complex surgeries at birth. We have previously demonstrated abnormal placental vascularization in human placentas from fetuses diagnosed with CHD. Hand1 has roles in both heart and placental development and is implicated in CHD development. We utilized two conditionally activated Hand1A126fs/+ murine mutant models to investigate the importance of cell-specific Hand1 on placental development in early (Nkx2-5Cre) and late (Cdh5Cre) pregnancy. Embryonic lethality occurred in Nkx2-5Cre/Hand1A126fs/+ embryos with marked fetal demise occurring after E10.5 due to a failure in placental labyrinth formation and therefore the inability to switch to hemotrophic nutrition or maintain sufficient oxygen transfer to the fetus. Labyrinthine vessels failed to develop appropriately and vessel density was significantly lower by day E12.5. In late pregnancy, the occurrence of Cdh5Cre+;Hand1A126fs/+ fetuses was reduced from 29% at E12.5 to 20% at E18.5 and remaining fetuses exhibited reduced fetal and placental weights, labyrinth vessel density and placenta angiogenic factor mRNA expression. Our results demonstrate for the first time the necessity of Hand1 in both establishment and remodeling of the exchange area beyond early pregnancy and in patterning vascularization of the placental labyrinth crucial for maintaining pregnancy and successful fetal growth.
Collapse
|
20
|
Festa J, Da Boit M, Hussain A, Singh H. Potential Benefits of Berry Anthocyanins on Vascular Function. Mol Nutr Food Res 2021; 65:e2100170. [PMID: 34346156 DOI: 10.1002/mnfr.202100170] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/04/2021] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD), such as hypertension and atherosclerosis, is the leading cause of global death. Endothelial dysfunction (ED) is a strong predictor for most CVD making it a therapeutic target for both drug and nutrition interventions. It has been previously shown that polyphenols from wine and grape extracts possess vasodilator activities, due to the increased expression and phosphorylation of the endothelial nitric oxide synthase (eNOS), and consequent vasodilator nitric oxide (NO) production. This is vital in the prevention of ED, as NO production contributes to the maintenance of endothelial homeostasis. Moreover, polyphenols have the ability to inhibit reactive oxygen species (ROS), which can cause oxidative stress, as well as suppress the upregulation of inflammatory markers within the endothelium. However, while the majority of the research has focused on red wine, this has overshadowed the potential of other nutritional components for targeting ED, such as the use of berries. Berries are high in anthocyanin flavonoids a subtype of polyphenols with studies suggesting improved vascular function as a result of inducing NO production and reducing oxidative stress and inflammation. This review focuses on the protective effects of berries within the vasculature.
Collapse
Affiliation(s)
- Joseph Festa
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester, LE1 9BH, UK
| | - Mariasole Da Boit
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester, LE1 9BH, UK
| | - Aamir Hussain
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester, LE1 9BH, UK.,Department of Respiratory Sciences, College of Life Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Harprit Singh
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester, LE1 9BH, UK
| |
Collapse
|
21
|
Al-Ostoot FH, Salah S, Khamees HA, Khanum SA. Tumor angiogenesis: Current challenges and therapeutic opportunities. Cancer Treat Res Commun 2021; 28:100422. [PMID: 34147821 DOI: 10.1016/j.ctarc.2021.100422] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis plays an important role in the development of cancer since it allows for the delivery of oxygen, nutrients, and growth factors as well as tumor dissemination to distant organs. Inhibition of angiogenesis is an important strategy for the prevention of multiple solid tumors that depend on cutting or at least reducing the blood supply to tumor micro-regions, resulting in pan-hypoxia and pan-necrosis within solid tumor tissues. These drugs are an important part of treatment for some types of cancer. As a stand-alone therapy, inhibition of tumor angiogenesis can arrest or halt tumor growth, but will not eliminate the tumor. Therefore, anti-angiogenic drugs in combinations with another anti-cancer treatment method, like chemotherapy, lead to being critical for optimum cancer patient outcomes. Over the last two decades, investigations have been made to improve the efficacy of anti-angiogenic drugs, recognize their potential in drug interactions, and come up with plausible explanations for possible treatment resistance. This review will offer an overview of the varying concepts of tumor angiogenesis, several important angiogenic factors; focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
Affiliation(s)
- Fares Hezam Al-Ostoot
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India; Department of Biochemistry, Faculty of Education & Science, Al-Baydha University, Baydha, Yemen.
| | - Salma Salah
- Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Hussien Ahmed Khamees
- Department of Studies in Physics, Manasagangotri, University of Mysore, Mysuru, India
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India.
| |
Collapse
|
22
|
Dalan R, Boehm BO. The implications of COVID-19 infection on the endothelium: A metabolic vascular perspective. Diabetes Metab Res Rev 2021; 37:e3402. [PMID: 32871617 DOI: 10.1002/dmrr.3402] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/10/2020] [Accepted: 08/26/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Bernhard O Boehm
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| |
Collapse
|
23
|
Thakur V, Ratho RK, Kumar P, Bhatia SK, Bora I, Mohi GK, Saxena SK, Devi M, Yadav D, Mehariya S. Multi-Organ Involvement in COVID-19: Beyond Pulmonary Manifestations. J Clin Med 2021; 10:446. [PMID: 33498861 PMCID: PMC7866189 DOI: 10.3390/jcm10030446] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus Disease 19 (COVID-19), due to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has become an on-going global health emergency affecting over 94 million cases with more than 2 million deaths globally. Primarily identified as atypical pneumonia, it has developed into severe acute respiratory distress syndrome (ARDS), a multi-organ dysfunction with associated fatality. Ever since its emergence, COVID-19 with its plethora of clinical presentations has signalled its dynamic nature and versatility of the disease process. Being a disease with droplet transmission has now assumed the proportion of a suspected airborne nature which, once proved, poses a Herculean task to control. Because of the wide distribution of the human angiotensin-converting enzyme-2 (hACE2) receptors, known for its transmission, we envisage its multiorgan spread and extensive disease distribution. Thus, an extensive review of the extrapulmonary organotropism of SARS-CoV-2 with organ-specific pathophysiology and associated manifestations like dermatological complications, myocardial dysfunction, gastrointestinal symptoms, neurologic illnesses, hepatic and renal injury is needed urgently. The plausible mechanism of site-specific viral invasion is also discussed to give a comprehensive understanding of disease complexity, to help us to focus on research priorities and therapeutic strategies to counter the disease progression. A note on the latest advancements in vaccine research will enlighten the scientific world and equip it for better preparedness.
Collapse
Affiliation(s)
- Vikram Thakur
- Department of Virology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India; (V.T.); (I.B.); (G.K.M.)
| | - Radha Kanta Ratho
- Department of Virology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India; (V.T.); (I.B.); (G.K.M.)
| | - Pradeep Kumar
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India;
| | - Shashi Kant Bhatia
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul 05029, Korea;
| | - Ishani Bora
- Department of Virology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India; (V.T.); (I.B.); (G.K.M.)
| | - Gursimran Kaur Mohi
- Department of Virology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India; (V.T.); (I.B.); (G.K.M.)
| | - Shailendra K Saxena
- Centre for Advanced Research, Faculty of Medicine, King George’s Medical University, Lucknow 226003, India;
| | - Manju Devi
- Department of Oral Pathology and Microbiology, RUHS College of Dental Sciences (Government Dental College), RUHS University of Rajasthan, Jaipur, Rajasthan 302016, India;
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Korea
| | - Sanjeet Mehariya
- Department of Engineering, University of Campania ‘Luigi Vanitelli’, Real Casa dell’ Annunziata, Via Roma 29, 81031 Aversa, Italy
| |
Collapse
|
24
|
Roberts KA, Colley L, Agbaedeng TA, Ellison-Hughes GM, Ross MD. Vascular Manifestations of COVID-19 - Thromboembolism and Microvascular Dysfunction. Front Cardiovasc Med 2020; 7:598400. [PMID: 33195487 PMCID: PMC7649150 DOI: 10.3389/fcvm.2020.598400] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
The coronavirus pandemic has reportedly infected over 31.5 million individuals and caused over 970,000 deaths worldwide (as of 22nd Sept 2020). This novel coronavirus, officially named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), although primarily causes significant respiratory distress, can have significant deleterious effects on the cardiovascular system. Severe cases of the virus frequently result in respiratory distress requiring mechanical ventilation, often seen, but not confined to, individuals with pre-existing hypertension and cardiovascular disease, potentially due to the fact that the virus can enter the circulation via the lung alveoli. Here the virus can directly infect vascular tissues, via TMPRSS2 spike glycoprotein priming, thereby facilitating ACE-2-mediated viral entry. Clinical manifestations, such as vasculitis, have been detected in a number of vascular beds (e.g., lungs, heart, and kidneys), with thromboembolism being observed in patients suffering from severe coronavirus disease (COVID-19), suggesting the virus perturbs the vasculature, leading to vascular dysfunction. Activation of endothelial cells via the immune-mediated inflammatory response and viral infection of either endothelial cells or cells involved in endothelial homeostasis, are some of the multifaceted mechanisms potentially involved in the pathogenesis of vascular dysfunction within COVID-19 patients. In this review, we examine the evidence of vascular manifestations of SARS-CoV-2, the potential mechanism(s) of entry into vascular tissue and the contribution of endothelial cell dysfunction and cellular crosstalk in this vascular tropism of SARS-CoV-2. Moreover, we discuss the current evidence on hypercoagulability and how it relates to increased microvascular thromboembolic complications in COVID-19.
Collapse
Affiliation(s)
- Kirsty A. Roberts
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Liam Colley
- School of Sport, Health & Exercise Science, Bangor University, Bangor, United Kingdom
| | - Thomas A. Agbaedeng
- Centre for Heart Rhythm Disorders, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Georgina M. Ellison-Hughes
- Centre for Human and Physiological Sciences, Faculty of Life Sciences & Medicine, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| |
Collapse
|
25
|
Mondal R, Lahiri D, Deb S, Bandyopadhyay D, Shome G, Sarkar S, Paria SR, Thakurta TG, Singla P, Biswas SC. COVID-19: Are we dealing with a multisystem vasculopathy in disguise of a viral infection? J Thromb Thrombolysis 2020; 50:567-579. [PMID: 32627126 PMCID: PMC7335630 DOI: 10.1007/s11239-020-02210-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
After the emergence of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS-CoV) in the last two decades, the world is facing its new challenge in Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic with unprecedented global response. With the expanding domain of presentations in COVID-19 patients, the full range of manifestations is yet to unfold. The classical clinical symptoms for SARS-CoV-2 affected patients are dry cough, high fever, dyspnoea, lethal pneumonia whereas many patients have also been found to be associated with a few additional signs and clinical manifestations of isolated vasculopathy. Albeit a deep and profound knowledge has been gained on the clinical features and management of COVID-19, less clear association has been provided on SARS-CoV-2 mediated direct or indirect vasculopathy and its possible correlation with disease prognosis. The accumulative evidences suggest that novel coronavirus, apart from its primary respiratory confinement, may also invade vascular endothelial cells of several systems including cerebral, cardio-pulmonary as well as renal microvasculature, modulating multiple visceral perfusion indices. Here we analyse the phylogenetic perspective of SARS-CoV-2 along with other strains of β-coronaviridae from a standpoint of vasculopathic derangements. Based on the existing case reports, literature and open data bases, we also analyse the differential pattern of vasculopathy related changes in COVID-19 positive patients. Besides, we debate the need of modulation in clinical approach from a hemodynamical point of view, as a measure towards reducing disease transmission, morbidity and mortality in SARS-CoV-2 affected patients.
Collapse
Affiliation(s)
- Ritwick Mondal
- Institute of Post Graduate Medical Education and Research, SSKM Hospital, 52/1A, S.N. Pandit Street, Kolkata, 700025, India
| | - Durjoy Lahiri
- Institute of Post Graduate Medical Education and Research, SSKM Hospital, 52/1A, S.N. Pandit Street, Kolkata, 700025, India.
| | - Shramana Deb
- S.N. Pradhan Centre for Neuroscience, University of Calcutta, Kolkata, India
| | - Deebya Bandyopadhyay
- Institute of Post Graduate Medical Education and Research, SSKM Hospital, 52/1A, S.N. Pandit Street, Kolkata, 700025, India
| | - Gourav Shome
- Department of Microbiology, University of Calcutta, Kolkata, India
| | - Sukanya Sarkar
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sudeb R Paria
- Institute of Post Graduate Medical Education and Research, SSKM Hospital, 52/1A, S.N. Pandit Street, Kolkata, 700025, India
| | | | - Pratibha Singla
- Gian Sagar Medical College and Hospital, Patiala, Punjab, India
| | | |
Collapse
|
26
|
Marone G, Schroeder JT, Mattei F, Loffredo S, Gambardella AR, Poto R, de Paulis A, Schiavoni G, Varricchi G. Is There a Role for Basophils in Cancer? Front Immunol 2020; 11:2103. [PMID: 33013885 PMCID: PMC7505934 DOI: 10.3389/fimmu.2020.02103] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Basophils were identified in human peripheral blood by Paul Ehrlich over 140 years ago. Human basophils represent <1% of peripheral blood leukocytes. During the last decades, basophils have been described also in mice, guinea pigs, rabbits, and monkeys. There are many similarities, but also several immunological differences between human and mouse basophils. There are currently several strains of mice with profound constitutive or inducible basophil deficiency useful to prove that these cells have specific roles in vivo. However, none of these mice are solely and completely devoid of all basophils. Therefore, the relevance of these findings to humans remains to be established. It has been known for some time that basophils have the propensity to migrate into the site of inflammation. Recent observations indicate that tissue resident basophils contribute to lung development and locally promote M2 polarization of macrophages. Moreover, there is increasing evidence that lung-resident basophils exhibit a specific phenotype, different from circulating basophils. Activated human and mouse basophils synthesize restricted and distinct profiles of cytokines. Human basophils produce several canonical (e.g., VEGFs, angiopoietin 1) and non-canonical (i.e., cysteinyl leukotriene C4) angiogenic factors. Activated human and mouse basophils release extracellular DNA traps that may have multiple effects in cancer. Hyperresponsiveness of basophils has been demonstrated in patients with JAK2V617F-positive polycythemia vera. Basophils are present in the immune landscape of human lung adenocarcinoma and pancreatic cancer and can promote inflammation-driven skin tumor growth. The few studies conducted thus far using different models of basophil-deficient mice have provided informative results on the roles of these cells in tumorigenesis. Much more remains to be discovered before we unravel the hitherto mysterious roles of basophils in human and experimental cancers.
Collapse
Affiliation(s)
- Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy.,Azienda Ospedaliera Ospedali dei Colli, Monaldi Hospital Pharmacy, Naples, Italy
| | - John T Schroeder
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University, Baltimore, MD, United States
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (CNR), Naples, Italy
| | | | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (CNR), Naples, Italy
| |
Collapse
|
27
|
Si Y, Huang J, Li X, Fu Y, Xu R, Du Y, Cheng J, Jiang H. AKT/FOXO1 axis links cross-talking of endothelial cell and pericyte in TIE2-mutated venous malformations. Cell Commun Signal 2020; 18:139. [PMID: 32867785 PMCID: PMC7457504 DOI: 10.1186/s12964-020-00606-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/29/2020] [Indexed: 01/19/2023] Open
Abstract
Background Venous malformations (VMs), most of which associated with activating mutations in the endothelial cells (ECs) tyrosine kinase receptor TIE2, are characterized by dilated and immature veins with scarce smooth muscle cells (SMCs) coverage. However, the underlying mechanism of interaction between ECs and SMCs responsible for VMs has not been fully understood. Methods Here, we screened 5 patients with TIE2-L914F mutation who were diagnosed with VMs by SNP sequencing, and we compared the expression of platelet-derived growth factor beta (PDGFB) and α-SMA in TIE2 mutant veins and normal veins by immunohistochemistry. In vitro, we generated TIE2-L914F-expressing human umbilical vein endothelial cells (HUVECs) and performed BrdU, CCK-8, transwell and tube formation experiments on none-transfected and transfected ECs. Then we investigated the effects of rapamycin (RAPA) on cellular characteristics. Next we established a co-culture system and investigated the role of AKT/FOXO1/PDGFB in regulating cross-talking of mutant ECs and SMCs. Results VMs with TIE2-L914F mutation showed lower expression of PDGFB and α-SMA than normal veins. TIE2 mutant ECs revealed enhanced cell viability and motility, and decreased tube formation, whereas these phenotypes could be reversed by rapamycin. Mechanically, RAPA ameliorated the physiological function of mutant ECs by inhibiting AKT-mTOR pathway, but also facilitated the nuclear location of FOXO1 and the expression of PDGFB in mutant ECs, and then improved paracrine interactions between ECs and SMCs. Moreover, TIE2 mutant ECs strongly accelerated the transition of SMCs from contractile phenotype to synthetic phenotype, whereas RAPA could prevent the phenotype transition of SMCs. Conclusions Our data demonstrate a previously unknown mechanistic linkage of AKT-mTOR/FOXO1 pathway between mutant ECs and SMCs in modulating venous dysmorphogenesis, and AKT/FOXO1 axis might be a potential therapeutic target for the recovery of TIE2-mutation causing VMs. Video Abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Yameng Si
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiadong Huang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiang Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yu Fu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Rongyao Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yifei Du
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hongbing Jiang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
28
|
Chen L, Li X, Chen M, Feng Y, Xiong C. The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2. Cardiovasc Res 2020; 116:1097-1100. [PMID: 32227090 PMCID: PMC7184507 DOI: 10.1093/cvr/cvaa078] [Citation(s) in RCA: 791] [Impact Index Per Article: 158.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/25/2020] [Indexed: 01/08/2023] Open
Abstract
A new type of pneumonia caused by a novel coronavirus SARS-CoV-2 outbreaks recently in China and spreads into many other countries. This disease, named as COVID-19, is similar to patients infected by SARS-CoV and MERS-CoV, and nearly 20% of patients developed severe condition. Cardiac injury is a prevalent complication of severe patients, exacerbating the disease severity in coronavirus disease 2019 (COVID-19) patients. Angiotensin-converting enzyme 2 (ACE2), the key host cellular receptor of SARS-CoV-2, has been identified in multiple organs, but its cellular distribution in human heart is not illuminated clearly. This study performed the first state-of-art single cell atlas of adult human heart, and revealed that pericytes with high expression of ACE2 might act as the target cardiac cell of SARS-CoV-2. The pericytes injury due to virus infection may result in capillary endothelial cells dysfunction, inducing microvascular dysfunction. And patients with basic heart failure disease showed increased ACE2 expression at both mRNA and protein levels, meaning that if infected by the virus these patients may have higher risk of heart attack and critically ill condition. The finding of this study explains the high rate of severe cases among COVID-19 patients with basic cardiovascular disease; and these results also perhaps provide important reference to clinical treatment of cardiac injury among severe patients infected by SARS-CoV-2.
Collapse
Affiliation(s)
- Liang Chen
- Department of Epidemiology, School of Public Health, Fudan University, NO. 130 Dong'an Road, Xuhui district, Shanghai 200032, China
| | - Xiangjie Li
- Department of Medical Biostatistics, Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing 100872, China
| | - Mingquan Chen
- Department of Emergency, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Chenglong Xiong
- Department of Epidemiology, School of Public Health, Fudan University, NO. 130 Dong'an Road, Xuhui district, Shanghai 200032, China
| |
Collapse
|