1
|
Lotfalizadeh M, Khadem N, Sadeghi T, Jahanpak N, Mahmoudinia M, Faraji P, Zakerinasab F, Mahmoudinia M. The effect of intravenous calcium gluconate on the prevention of ovarian hyperstimulation syndrome. (A randomized clinical trial). J Gynecol Obstet Hum Reprod 2024; 53:102850. [PMID: 39293587 DOI: 10.1016/j.jogoh.2024.102850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/15/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE Oral Cabergoline and intravenous Calcium have the potential to prevent Ovarian Hyperstimulation Syndrome (OHSS) in assisted reproductive technology by regulating the activity of the Vascular Endothelial Growth Factor (VEGF) receptor. The purpose of this study was to examine the effect of Cabergoline with intravenous Calcium versus oral Cabergoline alone on the overall rate of OHSS. METHODS This study is a randomized clinical trial which was carried out in Milad Infertility Center affiliated with Mashhad University of Medical Sciences, Mashhad, Iran between April 2016 and January 2018. A total of 192 patients were randomly assigned into two groups. The control group received oral Cabergoline and the intervention group received Calcium gluconate in addition to Cabergoline. A total rate of OHSS, moderate and severe OHSS were measured in both groups. RESULTS The demographic characteristics of the participants and the types of drugs used showed homogeneity between the intervention and control groups (P > 0.05). Furthermore, there was no significant difference between the two groups in terms of the number of the follicle, oocytes obtained, metaphase II oocytes, the number of embryos, and the rate of fertilization. Regarding the incidence of OHSS, 26.2 % of participants in the control group experienced OHSS, while the occurrence rate was 15.7 % in the intervention group (P = 0.401). The incidence of severe OHSS in the control group and intervention group was 7.1 % and 3.6 %, respectively. CONCLUSION Intravenous injection of Calcium gluconate can be effective in preventing Ovarian Hyperstimulation Syndrome.
Collapse
Affiliation(s)
- Marzieh Lotfalizadeh
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nayereh Khadem
- Associate Professor, Fellowship of Infertility, Supporting the Family and the Youth of Population Research Core, Department of Obstetrics and Gynecology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Tahere Sadeghi
- Nursing and Midwifery Care Research Center, Clinical Research Development Unit of Akbar Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | - Pardis Faraji
- Mashhad University of Medical Sciences. Mashhad, Iran.
| | - Faezeh Zakerinasab
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Malihe Mahmoudinia
- Associate Professor, Fellowship of Infertility, Supporting the Family and the Youth of Population Research Core, Department of Obstetrics and Gynecology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Gullo G, Cucinella G, Stojanovic V, Stojkovic M, Bruno C, Streva AV, Lopez A, Perino A, Marinelli S. Ovarian Hyperstimulation Syndrome (OHSS): A Narrative Review and Legal Implications. J Pers Med 2024; 14:915. [PMID: 39338169 PMCID: PMC11433561 DOI: 10.3390/jpm14090915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Infertility is a highly meaningful issue with potentially life-changing consequences, and its incidence has been growing worldwide. Assisted reproductive technology (ART) has made giant strides in terms of treating many infertility conditions, despite the risk of developing ovarian hyperstimulation syndrome (OHSS), a potentially life-threatening complication. METHODS This narrative review draws upon scientific articles found in the PubMed database. The search spanned the 1990-2024 period. Search strings used included "OHSS" or "ovarian hyperstimulation" and "IVF" and "GnRH" and "hCG"; 1098 results were retrieved and were ultimately narrowed down to 111 suitable sources, i.e., relevant articles dealing with the condition's underlying dynamics, management pathways, and evidence-based criteria and guidelines, crucial both from a clinical perspective and from the standpoint of medicolegal tenability. RESULTS The following features constitute OHSS risk factors: young age, low body weight, and polycystic ovarian syndrome (PCOS), among others. GnRH antagonist can substantially lower the risk of severe OHSS, compared to the long protocol with a gonadotropin-releasing hormone (GnRH) agonist. However, a mild or moderate form of OHSS is also possible if the antagonist protocol is used, especially when hCG is used for the final maturation of oocytes. For women at risk of OHSS, GnRH agonist trigger and the freeze-all strategy is advisable. OHSS is one of the most frequent complications, with a 30% rate in IVF cycles. CONCLUSION Providing effective care for OHSS patients begins with early diagnosis, while also evaluating for comorbidities and complications. In addition to that, we should pay more attention to the psychological component of this complication and of infertility as a whole. Compliance with guidelines and evidence-based best practices is essential for medicolegal tenability.
Collapse
Affiliation(s)
- Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Gaspare Cucinella
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Vukasin Stojanovic
- Emergency Medicine Center of Montenegro, Faculty of Medicine, University of Montenegro, 81000 Podgorica, Montenegro;
| | - Mirjana Stojkovic
- Clinic of Endocrinology, Diabetes and Metabolic Disorders, University Clinical Center of Serbia, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Carmine Bruno
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Istituto Dermopatico dell’Immacolata (IDI IRCCS), 00167 Rome, Italy
| | - Adriana Vita Streva
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Alessandra Lopez
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Antonio Perino
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Susanna Marinelli
- School of Law, Polytechnic University of Marche, 60121 Ancona, Italy;
| |
Collapse
|
3
|
Moon SY, Kim HJ, Kim JK, Kim J, Choi JS, Won SY, Park K, Lee SHS. An examination of the mechanisms driving the therapeutic effects of an AAV expressing a soluble variant of VEGF receptor-1. PLoS One 2024; 19:e0305466. [PMID: 38990973 PMCID: PMC11239064 DOI: 10.1371/journal.pone.0305466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
In previous animal model studies, we demonstrated the potential of rAAV2-sVEGFRv-1, which encodes a truncated variant of the alternatively spliced soluble version of VEGF receptor-1 (VEGFR1), as a human gene therapy for age-related macular degeneration (AMD) and diabetic retinopathy (DR). Here, we elucidate in vitro some of the mechanisms by which rAAV2-sVEGFRv-1 exerts its therapeutic effects. Human umbilical vein endothelial cells (HUVECs) were infected with rAAV2-sVEGFRv-1 or a control virus vector in the presence of members of the VEGF family to identify potential binding partners via ELISA, which showed that VEGF-A, VEGF-B, and placental growth factor (PlGF) are all ligands of its transgene product. In order to determine the effects of rAAV2-sVEGFRv-1 on cell proliferation and permeability, processes that are important to the progression AMD and DR, HUVECs were infected with the therapeutic virus vector under the stimulation of VEGF-A, the major driver of the neovascularization that characterizes the forms of these conditions most associated with vision loss. rAAV2-sVEGFRv-1 treatment, as a result, markedly reduced the extent to which these processes occurred, with the latter determined by measuring zonula occludens 1 expression. Finally, the human microglial HMC3 cell line was used to show the effects of the therapeutic virus vector upon inflammatory processes, another major contributor to angiogenic eye disease pathophysiology, with rAAV2-sVEGFRv-1 reducing therein the secretion of pro-inflammatory cytokines interleukin (IL)-1β and IL-6. Combined with our previously published in vivo data, the in vitro activity of the expressed transgene here further demonstrates the great promise of rAAV2-sVEGFRv-1 as a potential human gene therapeutic for addressing angiogenic ocular conditions.
Collapse
Affiliation(s)
- Seo Yun Moon
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - Hee Jong Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - Jin Kwon Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - Jin Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - Jun-Sub Choi
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - So-Yoon Won
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - Keerang Park
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| | - Steven Hyun Seung Lee
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul, Korea
- CdmoGen Co., Ltd., Cheongju, Korea
| |
Collapse
|
4
|
Bedoschi G, Ingold C, Navarro PA. Fertility Preservation and Ovarian Hyperstimulation Syndrome Management in Cancer Care: A Pathophysiological Perspective on Gonadotropin-Releasing Hormone Agonists and Antagonists. PATHOPHYSIOLOGY 2024; 31:288-297. [PMID: 38921726 PMCID: PMC11206524 DOI: 10.3390/pathophysiology31020021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/02/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
This narrative review delves into the evolving landscape of fertility preservation techniques, with a particular focus on their use in patients undergoing oncology treatment that carries a risk of ovarian insufficiency. Advances in established methods such as cryopreservation of oocytes and embryos are highlighted, and the increasing use of gonadotropin-releasing hormone (GnRH) agonists is discussed. The review also addresses the complexities and controversies associated with these approaches, such as the 'flare-up' effect associated with GnRH agonists and the potential of GnRH antagonists to reduce the risk of ovarian hyperstimulation syndrome. Despite advances in fertility preservation, the report highlights the challenges we face, including the need for personalized treatment protocols and the management of associated risks. It calls for continued research and collaboration between healthcare professionals to refine these techniques and ultimately improve reproductive outcomes for patients facing the prospect of fertility-impairing treatment.
Collapse
Affiliation(s)
- Giuliano Bedoschi
- Department of Gynecology and Obstetrics, Reproductive Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil;
| | - Caroline Ingold
- Department of Collective Health, Sexual, Reproductive Health and Population Genetics, Faculdade de Medicina do ABC, Santo André 09060-870, Brazil;
| | - Paula Andrea Navarro
- Department of Gynecology and Obstetrics, Reproductive Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil;
| |
Collapse
|
5
|
Mahajan D, Sambyal V, Uppal MS, Sudan M, Guleria K. VEGF-2578C/A, -460T/C Polymorphisms and Gastrointestinal Tract Cancer Risk: An Updated Meta-Analysis. Genet Test Mol Biomarkers 2024; 28:176-188. [PMID: 38597641 DOI: 10.1089/gtmb.2023.0628] [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] [Indexed: 04/11/2024] Open
Abstract
Functional polymorphisms in the vascular endothelial growth factor (VEGF) alter the susceptibility toward different gastrointestinal tract (GIT) cancers. In this study, we explored the association of VEGF-2578C/A and VEGF-460T/C polymorphisms with esophageal cancer (EC) risk. In total, 330 patients with EC and 330 controls for VEGF-2578C/A polymorphism and 316 patients with EC and 316 controls for VEGF-460T/C polymorphism were genotyped. AA genotype (p = 0.01) and A allele (p = 0.02) of VEGF-2578C/A and CC genotype (p = 0.04) and C allele (p = 0.04) of VEGF-460T/C polymorphism were significantly associated with an increased risk of EC. VEGF-2578C/A and VEGF-460T/C polymorphisms have been studied in different GIT cancers, but results are inconclusive. Therefore, we performed a meta-analysis to assess the association of these polymorphisms with the risk of GIT cancers. The PubMed, ScienceDirect, and Google Scholar databases were used to search the articles. Twenty-one studies on VEGF-2578C/A and 20 studies on VEGF-460T/C polymorphism were included in this meta-analysis. VEGF-2578C/A polymorphism was associated with the decreased risk of GIT cancer in the overall population under the overdominant model (p = 0.009). A significant association of VEGF-2578C/A polymorphism with GIT cancer risk has been observed in the middle easterners, Caucasians, and Asians under different genetic models. VEGF-460T/C polymorphism was significantly associated with an increased risk of GIT cancers in Caucasians. Stratification of the data on the basis of cancer type showed a significant association of VEGF-2578C/A polymorphism with the risk of gallbladder cancer, whereas VEGF-460T/C polymorphism was associated with the risk of hepatocellular cancer, gastric cancer, and colorectal cancer. Our meta-analysis suggested that VEGF-2578C/A and VEGF-460T/C polymorphisms were associated with GIT cancer risk.
Collapse
Affiliation(s)
- Deepanshi Mahajan
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vasudha Sambyal
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Manjit Singh Uppal
- Department of Surgery, Sri Guru Ram Das Institute of Medical Sciences and Research, Amritsar, Punjab, India
| | - Meena Sudan
- Department of Radiotherapy, Sri Guru Ram Das Institute of Medical Sciences and Research, Amritsar, Punjab, India
| | - Kamlesh Guleria
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
6
|
Choi YK. Detrimental Roles of Hypoxia-Inducible Factor-1α in Severe Hypoxic Brain Diseases. Int J Mol Sci 2024; 25:4465. [PMID: 38674050 PMCID: PMC11050730 DOI: 10.3390/ijms25084465] [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: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Hypoxia stabilizes hypoxia-inducible factors (HIFs), facilitating adaptation to hypoxic conditions. Appropriate hypoxia is pivotal for neurovascular regeneration and immune cell mobilization. However, in central nervous system (CNS) injury, prolonged and severe hypoxia harms the brain by triggering neurovascular inflammation, oxidative stress, glial activation, vascular damage, mitochondrial dysfunction, and cell death. Diminished hypoxia in the brain improves cognitive function in individuals with CNS injuries. This review discusses the current evidence regarding the contribution of severe hypoxia to CNS injuries, with an emphasis on HIF-1α-mediated pathways. During severe hypoxia in the CNS, HIF-1α facilitates inflammasome formation, mitochondrial dysfunction, and cell death. This review presents the molecular mechanisms by which HIF-1α is involved in the pathogenesis of CNS injuries, such as stroke, traumatic brain injury, and Alzheimer's disease. Deciphering the molecular mechanisms of HIF-1α will contribute to the development of therapeutic strategies for severe hypoxic brain diseases.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
7
|
Yang Y, Li C, Yang S, Zhang Z, Bai X, Tang H, Huang J. Cepharanthine maintains integrity of the blood-brain barrier (BBB) in stroke via the VEGF/VEGFR2/ZO-1 signaling pathway. Aging (Albany NY) 2024; 16:5905-5915. [PMID: 38517394 PMCID: PMC11042958 DOI: 10.18632/aging.205678] [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: 07/16/2023] [Accepted: 11/21/2023] [Indexed: 03/23/2024]
Abstract
Dysfunction of tight junctions such as zonula occludens protein-1 (ZO-1)-associated aggravation of blood-brain barrier (BBB) permeability plays an important role in the progression of stroke. Cepharanthine (CEP) is an extract from the plant Stephania cepharantha. However, the effects of CEP on stroke and BBB dysfunction have not been previously reported. In this study, we report that CEP improved dysfunction in neurological behavior in a middle cerebral artery occlusion (MCAO) mouse model. Importantly, CEP suppressed blood-brain barrier (BBB) hyperpermeability by increasing the expression of ZO-1. Notably, we found that CEP inhibited the expression of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) in the cortex of MCAO mice. Additionally, the results of in vitro experiments demonstrate that treatment with CEP ameliorated cytotoxicity of human bEnd.3 brain microvascular endothelial cells against hypoxia/reperfusion (H/R). Also, CEP attenuated H/R-induced aggravation of endothelial permeability in bEND.3 cells by restoring the expression of ZO-1. Further study proved that the protective effects of CEP are mediated by inhibition of VEGF-A and VEGFR2. Based on the results, we conclude that CEP might possess a therapeutic prospect in stroke through protecting the integrity of the BBB mediated by the VEGF/VEGFR2/ZO-1 axis.
Collapse
Affiliation(s)
- Yunfang Yang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Changjiang Li
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Sijin Yang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacology, Southwest Medical University, Luzhou 646099, Sichuan, China
| | - Xue Bai
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Hongmei Tang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| |
Collapse
|
8
|
Osipova NA, Panova AY, Efremov AM, Lozinskaya NA, Beznos OV, Katargina LA. Melatonin and its bioisosteres as potential therapeutic agents for the treatment of retinopathy of prematurity. Chem Biol Drug Des 2024; 103:e14504. [PMID: 38480485 DOI: 10.1111/cbdd.14504] [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: 11/14/2023] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
We conducted a study on the impact of intraperitoneal injections of melatonin and its three bioisosteres (compounds 1-3) on the development of oxygen-induced retinopathy in newborn rats during a 21-day experiment. It was demonstrated that melatonin and its analogues 1-3 effectively reduce the total protein concentration in the vitreous body of rat pups, decrease concentration of VEGF-A, and lower the level of oxidative stress (as indicated by normalization of antioxidant activity in the vitreous body). Melatonin and its analogues 1-3 equally normalize the level of VEGF-A. Analogues 1 and 2 even exceed melatonin in their ability to reduce protein influx into the vitreous body. However, analogue 2 had no effect on antioxidant activity, while analogues 1 and 3 caused a significant increase in this parameter, with analogue 3 even slightly exceeding melatonin. Thus, it can be concluded that analogues 1-3 are comparable to melatonin and can be utilized as potential therapeutic agents for the treatment of retinopathy of prematurity.
Collapse
Affiliation(s)
- N A Osipova
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| | - A Y Panova
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| | - A M Efremov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - N A Lozinskaya
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - O V Beznos
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| | - L A Katargina
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| |
Collapse
|
9
|
Zhao G, Xue L, Weiner AI, Gong N, Adams-Tzivelekidis S, Wong J, Gentile ME, Nottingham AN, Basil MC, Lin SM, Niethamer TK, Diamond JM, Bermudez CA, Cantu E, Han X, Cao Y, Alameh MG, Weissman D, Morrisey EE, Mitchell MJ, Vaughan AE. TGF-βR2 signaling coordinates pulmonary vascular repair after viral injury in mice and human tissue. Sci Transl Med 2024; 16:eadg6229. [PMID: 38295183 DOI: 10.1126/scitranslmed.adg6229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024]
Abstract
Disruption of pulmonary vascular homeostasis is a central feature of viral pneumonia, wherein endothelial cell (EC) death and subsequent angiogenic responses are critical determinants of the outcome of severe lung injury. A more granular understanding of the fundamental mechanisms driving reconstitution of lung endothelium is necessary to facilitate therapeutic vascular repair. Here, we demonstrated that TGF-β signaling through TGF-βR2 (transforming growth factor-β receptor 2) is activated in pulmonary ECs upon influenza infection, and mice deficient in endothelial Tgfbr2 exhibited prolonged injury and diminished vascular repair. Loss of endothelial Tgfbr2 prevented autocrine Vegfa (vascular endothelial growth factor α) expression, reduced endothelial proliferation, and impaired renewal of aerocytes thought to be critical for alveolar gas exchange. Angiogenic responses through TGF-βR2 were attributable to leucine-rich α-2-glycoprotein 1, a proangiogenic factor that counterbalances canonical angiostatic TGF-β signaling. Further, we developed a lipid nanoparticle that targets the pulmonary endothelium, Lung-LNP (LuLNP). Delivery of Vegfa mRNA, a critical TGF-βR2 downstream effector, by LuLNPs improved the impaired regeneration phenotype of EC Tgfbr2 deficiency during influenza injury. These studies defined a role for TGF-βR2 in lung endothelial repair and demonstrated efficacy of an efficient and safe endothelial-targeted LNP capable of delivering therapeutic mRNA cargo for vascular repair in influenza infection.
Collapse
Affiliation(s)
- Gan Zhao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron I Weiner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephanie Adams-Tzivelekidis
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanna Wong
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria E Gentile
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ana N Nottingham
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan M Lin
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Terren K Niethamer
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua M Diamond
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christian A Bermudez
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Cantu
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yaqi Cao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [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: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
11
|
Kim CH, Kim HY, Nah SY, Choi YK. The effects of Korean Red Ginseng on heme oxygenase-1 with a focus on mitochondrial function in pathophysiologic conditions. J Ginseng Res 2023; 47:615-621. [PMID: 37720574 PMCID: PMC10499582 DOI: 10.1016/j.jgr.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/21/2023] [Accepted: 04/07/2023] [Indexed: 09/19/2023] Open
Abstract
Korean Red Ginseng (KRG) plays a key role in heme oxygenase (HO)-1 induction under physical and moderate oxidative stress conditions. The transient and mild induction of HO-1 is beneficial for cell protection, mitochondrial function, regeneration, and intercellular communication. However, chronic HO-1 overexpression is detrimental in severely injured regions. Thus, in a chronic pathological state, diminishing HO-1-mediated ferroptosis is beneficial for a therapeutic approach. The molecular mechanisms by which KRG protects various cell types in the central nervous system have not yet been established, especially in terms of HO-1-mediated mitochondrial functions. Therefore, in this review, we discuss the multiple roles of KRG in the regulation of astrocytic HO-1 under pathophysiological conditions. More specifically, we discuss the role of the KRG-mediated astrocytic HO-1 pathway in regulating mitochondrial functions in acute and chronic neurodegenerative diseases as well as physiological conditions.
Collapse
Affiliation(s)
- Chang-Hee Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Republic of Korea
| | - Hahn Young Kim
- Department of Neurology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yoon Kyung Choi
- Bio/Molecular Informatics Center, Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
12
|
González López R, Contreras Caro Del Castillo DA, Valdez Magaña G, Sarmiento Silva RE, Martínez Castañeda FE, Trujillo Ortega ME. Expression and localization of vascular endothelial growth factor and its receptors in the pig uterus during peri-implantation and determination of the in vitro effect of the angiogenesis inhibitor SU5416 on VEGF system expression. Theriogenology 2023; 207:49-60. [PMID: 37269596 DOI: 10.1016/j.theriogenology.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 06/05/2023]
Abstract
The aim of this work was to determine endometrial mRNA expression and uterine protein localization of vascular endothelial growth factor (VEGF) and its receptors VEGFR1 and VEGFR2 during the estrous cycle and peri-implantation period in sows. Uterine tissues were collected from pregnant sows on days 12, 14, 16, and 18 after artificial insemination and from non-pregnant animals on days 2 and 12 of the estrous cycle (day 0 = day of estrus). Using immunohistochemistry, a positive signal for VEGF and its receptor VEGFR2 was found in uterine luminal epithelial cells, endometrial glands, stroma, blood vessels, and myometrium. A VEGFR1 signal was only found in endometrial and myometrial blood vessels and stroma. By day 18 of gestation, the mRNA expression levels of VEGF, VEGFR1, and VEGFR2 were higher than those observed on days 2 and 12 of the estrous cycle and on days 12, 14, and 16 of gestation. Then, a primary culture of sow endometrial epithelial cells was established to define the potential of the selective inhibition of VEGFR2 after treatment with inhibitor SU5416 and determine its effects on the expression pattern of the VEGF system. The endometrial epithelial cells treated with SU5416 showed a dose-dependent decrease in VEGFR1 and VEGFR2 mRNA expression. The present study provides additional evidence on the importance of the VEGF system during peri-implantation, as well as on the specific inhibitory activity of SU5416 in epithelial cells, which, as demonstrated, express the protein and mRNA of VEGF and its receptors VEGFR1 and VEGFR2.
Collapse
Affiliation(s)
- Rodrigo González López
- Universidad Nacional Autónoma de México, Facultad de Medicina Veterinaria y Zootecnia, 04510, Ciudad de México, Mexico
| | | | - Griselda Valdez Magaña
- Universidad Nacional Autónoma de México, Facultad de Medicina Veterinaria y Zootecnia, 04510, Ciudad de México, Mexico
| | - Rosa Elena Sarmiento Silva
- Universidad Nacional Autónoma de México, Facultad de Medicina Veterinaria y Zootecnia, 04510, Ciudad de México, Mexico
| | | | - María Elena Trujillo Ortega
- Universidad Nacional Autónoma de México, Facultad de Medicina Veterinaria y Zootecnia, 04510, Ciudad de México, Mexico.
| |
Collapse
|
13
|
Ahmad A, Nawaz MI. Molecular mechanism of VEGF and its role in pathological angiogenesis. J Cell Biochem 2022; 123:1938-1965. [PMID: 36288574 DOI: 10.1002/jcb.30344] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/12/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022]
Abstract
Over the last seven decades, a significant scientific contribution took place in the delineation of the implications of vascular endothelial-derived growth factor (VEGF) in the processes of angiogenesis. Under pathological conditions, mainly in response to hypoxia or ischemia, elevated VEGF levels promote vascular damage and the growth of abnormal blood vessels. Indeed, the development of VEGF biology has revolutionized our understanding of its role in pathological conditions. Hence, targeting VEGF or VEGF-mediated molecular pathways could be an excellent therapeutic strategy for managing cancers and intraocular neovascular disorders. Although anti-VEGF therapies, such as monoclonal antibodies and small-molecule tyrosine kinase inhibitors, have limited clinical efficacy, they can still significantly improve the overall survival rate. This thus demands further investigation through the development of alternative strategies in the management of VEGF-mediated pathological angiogenesis. This review article focuses on the recent developments toward the delineation of the functional biology of VEGF and the role of anti-VEGF strategies in the management of tumor and eye pathologies. Moreover, therapeutic angiogenesis, an exciting frontier for the treatment of ischemic disorders, is highlighted in this review, including wound healing.
Collapse
Affiliation(s)
- Ajmal Ahmad
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology, Abdulaziz University Hospital, Riyadh, Saudi Arabia
| | - Mohd Imtiaz Nawaz
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology, Abdulaziz University Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Bilister Egilmez C, Koyu A, Azak Pazarlar B, Keselik E, Oz Oyar E, Kurus M. Effects of intracerebroventricular and intravenous administration of Kisspeptin-54 and Gonadotropin-releasing hormone agonist in rats with ovarian hyperstimulation. Neuropeptides 2022; 96:102298. [PMID: 36367482 DOI: 10.1016/j.npep.2022.102298] [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: 07/25/2022] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE We aim to determine the effect of local and systemic administration of kisspeptin-54 on ovarian hyperstimulation. METHODS Immature female rats were used. In order to generate the ovarian hyperstimulation model, 50 IU PMSG was administered for 4 consecutive days and a single dose of 25 IU hCG was administered to all groups except for the sham group. To synchronize the sham group, a single dose of 10 IU PMSG followed by 10 IU hCG (48 h later) was applied. Kisspeptin-54 and gonadotropin-releasing hormone (GnRH) agonists were administered 48 h after hCG injection. While intracerebroventricular injection is performed with stereotaxic surgery, Intravenous administration was from the tail vein. Ovarian weights were measured. FSH, LH, estrogen and progesterone hormones were detected in serum by ELISA. VEGFa, IL-1β, TNF-α, MCP-1 immunohistochemical staining was performed on the ovaries and hypothalamus and their optical densities were determined with Image J. Kiss1R mRNA expression was determined by qRT-PCR. RESULTS Ovarian weights increased significantly in the OHSS group and the systemic GnRH agonist group. The optical densities of VEGFa, IL-1β, TNF- α and MCP-1 immunoreactivity showed us that both local and systemic applied kisspeptin-54 attenuates the level of investigated inflammation parameters in the ovaries. Moreover, local administration of kisspeptin-54 has been shown to enhance the level of Kiss1R mRNA in both the ovaries and the hypothalamus. CONCLUSION(S) Local and systemic administration of Kisspeptin-54 as a post-treatment reduces inflammation parameters in the ovaries. These findings promote the potential use of kisspeptin-54 on OHSS.
Collapse
Affiliation(s)
- Cansu Bilister Egilmez
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey.
| | - Ahmet Koyu
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Burcu Azak Pazarlar
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Erdi Keselik
- Department of Histology and Embryology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Eser Oz Oyar
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Meltem Kurus
- Department of Histology and Embryology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| |
Collapse
|
15
|
Phoenix KN, Yue Z, Yue L, Cronin CG, Liang BT, Hoeppner LH, Claffey KP. PLCβ2 Promotes VEGF-Induced Vascular Permeability. Arterioscler Thromb Vasc Biol 2022; 42:1229-1241. [PMID: 35861069 PMCID: PMC9492642 DOI: 10.1161/atvbaha.122.317645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Regulation of vascular permeability is critical to maintaining tissue metabolic homeostasis. VEGF (vascular endothelial growth factor) is a key stimulus of vascular permeability in acute and chronic diseases including ischemia reperfusion injury, sepsis, and cancer. Identification of novel regulators of vascular permeability would allow for the development of effective targeted therapeutics for patients with unmet medical need. METHODS In vitro and in vivo models of VEGFA-induced vascular permeability, pathological permeability, quantitation of intracellular calcium release and cell entry, and phosphatidylinositol 4,5-bisphosphate levels were evaluated with and without modulation of PLC (phospholipase C) β2. RESULTS Global knock-out of PLCβ2 in mice resulted in blockade of VEGFA-induced vascular permeability in vivo and transendothelial permeability in primary lung endothelial cells. Further work in an immortalized human microvascular cell line modulated with stable knockdown of PLCβ2 recapitulated the observations in the mouse model and primary cell assays. Additionally, loss of PLCβ2 limited both intracellular release and extracellular entry of calcium following VEGF stimulation as well as reduced basal and VEGFA-stimulated levels of phosphatidylinositol 4,5-bisphosphate compared to control cells. Finally, loss of PLCβ2 in both a hyperoxia-induced lung permeability model and a cardiac ischemia:reperfusion model resulted in improved animal outcomes when compared with wild-type controls. CONCLUSIONS The results implicate PLCβ2 as a key positive regulator of VEGF-induced vascular permeability through regulation of both calcium flux and phosphatidylinositol 4,5-bisphosphate levels at the cellular level. Targeting of PLCβ2 in a therapeutic setting may provide a novel approach to regulating vascular permeability in patients.
Collapse
Affiliation(s)
- Kathryn N. Phoenix
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT
| | - Zhichao Yue
- Pat and Jim Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, CT
| | - Lixia Yue
- Pat and Jim Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, CT
| | - Chunxia G. Cronin
- Pat and Jim Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, CT
| | - Bruce T. Liang
- Pat and Jim Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, CT
| | - Luke H. Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Kevin P. Claffey
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
16
|
Analysis of Tumor-Infiltrating T-Cell Transcriptomes Reveal a Unique Genetic Signature across Different Types of Cancer. Int J Mol Sci 2022; 23:ijms231911065. [PMID: 36232369 PMCID: PMC9569723 DOI: 10.3390/ijms231911065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
CD8+ and CD4+ T-cells play a key role in cellular immune responses against cancer by cytotoxic responses and effector lineages differentiation, respectively. These subsets have been found in different types of cancer; however, it is unclear whether tumor-infiltrating T-cell subsets exhibit similar transcriptome profiling across different types of cancer in comparison with healthy tissue-resident T-cells. Thus, we analyzed the single cell transcriptome of five tumor-infiltrating CD4-T, CD8-T and Treg cells obtained from different types of cancer to identify specific pathways for each subset in malignant environments. An in silico analysis was performed from single-cell RNA-sequencing data available in public repositories (Gene Expression Omnibus) including breast cancer, melanoma, colorectal cancer, lung cancer and head and neck cancer. After dimensionality reduction, clustering and selection of the different subpopulations from malignant and nonmalignant datasets, common genes across different types of cancer were identified and compared to nonmalignant genes for each T-cell subset to identify specific pathways. Exclusive pathways in CD4+ cells, CD8+ cells and Tregs, and common pathways for the tumor-infiltrating T-cell subsets were identified. Finally, the identified pathways were compared with RNAseq and proteomic data obtained from T-cell subsets cultured under malignant environments and we observed that cytokine signaling, especially Th2-type cytokine, was the top overrepresented pathway in Tregs from malignant samples.
Collapse
|
17
|
Turco C, Esposito G, Iaiza A, Goeman F, Benedetti A, Gallo E, Daralioti T, Perracchio L, Sacconi A, Pasanisi P, Muti P, Pulito C, Strano S, Ianniello Z, Fatica A, Forcato M, Fazi F, Blandino G, Fontemaggi G. MALAT1-dependent hsa_circ_0076611 regulates translation rate in triple-negative breast cancer. Commun Biol 2022; 5:598. [PMID: 35710947 PMCID: PMC9203778 DOI: 10.1038/s42003-022-03539-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
Vascular Endothelial Growth Factor A (VEGFA) is the most commonly expressed angiogenic growth factor in solid tumors and is generated as multiple isoforms through alternative mRNA splicing. Here, we show that lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) and ID4 (inhibitor of DNA-binding 4) protein, previously referred to as regulators of linear isoforms of VEGFA, induce back-splicing of VEGFA exon 7, producing circular RNA circ_0076611. Circ_0076611 is detectable in triple-negative breast cancer (TNBC) cells and tissues, in exosomes released from TNBC cells and in the serum of breast cancer patients. Circ_0076611 interacts with a variety of proliferation-related transcripts, included MYC and VEGFA mRNAs, and increases cell proliferation and migration of TNBC cells. Mechanistically, circ_0076611 favors the expression of its target mRNAs by facilitating their interaction with components of the translation initiation machinery. These results add further complexity to the multiple VEGFA isoforms expressed in cancer cells and highlight the relevance of post-transcriptional regulation of VEGFA expression in TNBC cells. The circular isoform of VEGFA mRNA (circ_0076611), associated with size and pathogenesis of triple-negative breast tumors, is produced via back splicing of exon-7 by a RNP complex comprising lncRNA-MALAT1, ID4 and SRSF1, and secreted through exosomes.
Collapse
Affiliation(s)
- Chiara Turco
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gabriella Esposito
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessia Iaiza
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Frauke Goeman
- UOSD SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Benedetti
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Enzo Gallo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Theodora Daralioti
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Letizia Perracchio
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Sacconi
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizia Pasanisi
- Unit of Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paola Muti
- Department of Health Research Methods, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.,Department of Biomedical, Surgical and Dental Sciences, "Università degli Studi di Milano", Milan, Italy
| | - Claudio Pulito
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sabrina Strano
- UOSD SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Zaira Ianniello
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Alessandro Fatica
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Mattia Forcato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Giulia Fontemaggi
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
18
|
Tang Y, Yu Z, Lu X, Fan Q, Huang W. Overcoming Vascular Barriers to Improve the Theranostic Outcomes of Nanomedicines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103148. [PMID: 35246962 PMCID: PMC9069202 DOI: 10.1002/advs.202103148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/16/2022] [Indexed: 05/04/2023]
Abstract
Nanotheranostics aims to utilize nanomaterials to prevent, diagnose, and treat diseases to improve the quality of patients' lives. Blood vessels are responsible to deliver nutrients and oxygen to the whole body, eliminate waste, and provide access for patrolling immune cells for healthy tissues. Meanwhile, they can also nourish disease tissues, spread disease factors or cells into other healthy tissues, and deliver nanotheranostic agents to cover all the regions of a disease tissue. Thus, blood vessels are the first and the most important barrier for highly efficient nanotheranostics. Here, the structure and function of blood vessels are explored and how these characteristics affect nanotheranostics is discussed. Moreover, new mechanisms and related strategies about overcoming vascular obstacles for improved nanotheranostic outcomes are critically summarized, and their merits and demerits of each strategy are analyzed. Moreover, the present challenges to completely exhibit the potential of overcoming vascular barriers to improve the theranostic outcomes of nanomedicines in life science are also discussed. Finally, the future perspective is further discussed.
Collapse
Affiliation(s)
- Yufu Tang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
| | - Zhongzheng Yu
- School of Chemical and Biomedical EngineeringNanyang Technological UniversitySingapore637459Singapore
| | - Xiaomei Lu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
- Shaanxi Institute of Flexible Electronics (SIFE)Northwestern Polytechnical University (NPU)Xi'an710072China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
- Shaanxi Institute of Flexible Electronics (SIFE)Northwestern Polytechnical University (NPU)Xi'an710072China
| |
Collapse
|
19
|
He L, Pi Y, Li Y, Wu Y, Jiang J, Rong X, Cai J, Yue Z, Cheng J, Li H, Chua MLK, Simone CB, Aronow WS, Lattanzi S, Palmer JD, Gaertner J, Glass J, Chen P, Tang Y. Efficacy and safety of apatinib in radiation-induced brain injury among head and neck cancer: an open-label, single-arm, phase 2 study. Int J Radiat Oncol Biol Phys 2022; 113:796-804. [PMID: 35378217 DOI: 10.1016/j.ijrobp.2022.03.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The treatment of radiation-induced brain injury (RI) caused by radiotherapy for head and neck cancer is challenging. Antiangiogenic therapy is a promising treatment. Apatinib is an oral tyrosine kinase inhibitor (TKI) that selectively inhibits vascular endothelial growth factor receptor (VEGFR) 2. We aimed to assess the efficacy and safety of apatinib in patients with RI. PATIENTS AND METHODS In this phase 2, open-label, single-arm, prospective study, we recruited patients aged 35-80 years with prior radiotherapy history for head and neck cancer who had newly diagnosed RI at the Sun Yat-sen Memorial Hospital, China. Apatinib was administered at a dosage of 250 mg once daily orally for 4 weeks. A Simon's mini-max two-stage design was performed. The primary outcome was the proportion of patients with an overall clinical efficacy defined as radiographic response ≥ 25% reduction in baseline brain edema volume on magnetic resonance (MR) fluid attenuated inversion recovery (FLAIR) images at week 4. Secondary endpoints were overall improvement rate of brain necrosis, neurological function, and safety. RESULTS We screened 37 patients, 36 of whom were enrolled between October 17, 2019 and August 3, 2020. At the cutoff date, 36 patients were assessed for efficacy and safety (19 to stage 1 and 17 to stage 2). Of the 36 patients evaluated for overall clinical efficacy, 22 patients (61.1%; 95%CI 43.5-76.9%) achieved the primary endpoint at week 4. Among the 31 patients with brain necrosis lesions, 19 patients (61.3%; 95%CI 42.2%-78.2%) showed improvement of brain necrosis. The most common grade 1 to 2 adverse events were hand-foot syndrome, fatigue and hypertension There were no treatment-related grade 4-5 toxicities. CONCLUSION Oral apatinib shows promising efficacy and is well-tolerated in patients with RI. Further randomized controlled studies are warranted. TRIAL REGISTRATION ClinicalTrials.gov (ID: NCT04152681).
Collapse
Affiliation(s)
- Lei He
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yaxuan Pi
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Li
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Wu
- Department of Biostatistics, Southern Medical University, Guangzhou, China
| | - Jingru Jiang
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Rong
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinhua Cai
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zongwei Yue
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinping Cheng
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Honghong Li
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Melvin Lee Kiang Chua
- Division of Radiation Oncology and Medical Sciences, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore
| | - Charles B Simone
- Department of Radiation Oncology, New York Proton Center and Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wilbert S Aronow
- New York Medical College/Westchester Medical Center, Valhalla, NY, USA
| | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Joshua D Palmer
- Departments of Radiation Oncology and Neurosurgery, The James Cancer Center at The Ohio State University, Columbus, OH, USA
| | - Jan Gaertner
- Palliative Care Center Hildegard, Basel, Switzerland, University of Basel, Switzerland
| | - Jon Glass
- Departments of Neurology and Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Pingyan Chen
- Department of Biostatistics, Southern Medical University, Guangzhou, China
| | - Yamei Tang
- Department of Neurology,Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | | |
Collapse
|
20
|
Miller B, Sewell-Loftin MK. Mechanoregulation of Vascular Endothelial Growth Factor Receptor 2 in Angiogenesis. Front Cardiovasc Med 2022; 8:804934. [PMID: 35087885 PMCID: PMC8787114 DOI: 10.3389/fcvm.2021.804934] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelial cells that compose the vascular system in the body display a wide range of mechanotransductive behaviors and responses to biomechanical stimuli, which act in concert to control overall blood vessel structure and function. Such mechanosensitive activities allow blood vessels to constrict, dilate, grow, or remodel as needed during development as well as normal physiological functions, and the same processes can be dysregulated in various disease states. Mechanotransduction represents cellular responses to mechanical forces, translating such factors into chemical or electrical signals which alter the activation of various cell signaling pathways. Understanding how biomechanical forces drive vascular growth in healthy and diseased tissues could create new therapeutic strategies that would either enhance or halt these processes to assist with treatments of different diseases. In the cardiovascular system, new blood vessel formation from preexisting vasculature, in a process known as angiogenesis, is driven by vascular endothelial growth factor (VEGF) binding to VEGF receptor 2 (VEGFR-2) which promotes blood vessel development. However, physical forces such as shear stress, matrix stiffness, and interstitial flow are also major drivers and effectors of angiogenesis, and new research suggests that mechanical forces may regulate VEGFR-2 phosphorylation. In fact, VEGFR-2 activation has been linked to known mechanobiological agents including ERK/MAPK, c-Src, Rho/ROCK, and YAP/TAZ. In vascular disease states, endothelial cells can be subjected to altered mechanical stimuli which affect the pathways that control angiogenesis. Both normalizing and arresting angiogenesis associated with tumor growth have been strategies for anti-cancer treatments. In the field of regenerative medicine, harnessing biomechanical regulation of angiogenesis could enhance vascularization strategies for treating a variety of cardiovascular diseases, including ischemia or permit development of novel tissue engineering scaffolds. This review will focus on the impact of VEGFR-2 mechanosignaling in endothelial cells (ECs) and its interaction with other mechanotransductive pathways, as well as presenting a discussion on the relationship between VEGFR-2 activation and biomechanical forces in the extracellular matrix (ECM) that can help treat diseases with dysfunctional vascular growth.
Collapse
Affiliation(s)
- Bronte Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
21
|
Kamaev A, Bulatov V, Vakhratyan P, Volkov A, Volkov A, Gavrilov E, Golovina V, Efremova O, Ivanov O, Ilyukhin E, Katorkin S, Konchugova T, Kravtsov P, Maksimov S, Mzhavanadze N, Pikhanova Z, Pryadko S, Smirnov A, Sushkov S, Chabbarov R, Shimanko A, Yakushkin S, Apkhanova T, Derkachev S, Zolotukhin I, Kalinin R, Kirienko A, Kulchitskaya D, Pelevin A, Petrikov A, Rachin A, Seliverstov E, Stoyko Y, Suchkov I. Varicose Veins. FLEBOLOGIIA 2022; 16:41. [DOI: 10.17116/flebo20221601141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
|
22
|
Kim M, Mok H, Yeo WS, Ahn JH, Choi YK. Role of ginseng in the neurovascular unit of neuroinflammatory diseases focused on the blood-brain barrier. J Ginseng Res 2021; 45:599-609. [PMID: 34803430 PMCID: PMC8587512 DOI: 10.1016/j.jgr.2021.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/28/2020] [Accepted: 02/17/2021] [Indexed: 12/18/2022] Open
Abstract
Ginseng has long been considered as an herbal medicine. Recent data suggest that ginseng has anti-inflammatory properties and can improve learning- and memory-related function in the central nervous system (CNS) following the development of CNS neuroinflammatory diseases such as Alzheimer's disease, cerebral ischemia, and other neurological disorders. In this review, we discuss the role of ginseng in the neurovascular unit, which is composed of endothelial cells surrounded by astrocytes, pericytes, microglia, neural stem cells, oligodendrocytes, and neurons, especially their blood-brain barrier maintenance, anti-inflammatory effects and regenerative functions. In addition, cell-cell communication enhanced by ginseng may be attributed to regeneration via induction of neurogenesis and angiogenesis in CNS diseases. Thus, ginseng may have therapeutic potential to exert cognitive improvement in neuroinflammatory diseases such as stroke, traumatic brain injury, multiple sclerosis, Parkinson's disease, and Alzheimer's disease.
Collapse
Affiliation(s)
- Minsu Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Woon-Seok Yeo
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Joong-Hoon Ahn
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Karabulut S, Korkmaz O, Erdem Altun C, Keskin I. A Histopathological Evaluation of Ovarian Hyperstimulation Syndrome on Reproductive and Vital Organs and the Role of the VEGF-PKA Pathway in a Mouse Model. Cells Tissues Organs 2021; 210:218-238. [PMID: 34320509 DOI: 10.1159/000517424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/14/2021] [Indexed: 11/19/2022] Open
Abstract
Ovarian hyperstimulation syndrome (OHSS) is one of the most common and iatrogenic complications of in vitro fertilization therapy, which is an exaggerated response to excess hormones resulting in the development of a large number of maturing follicles. Although the complications of and reasons for the condition are well known, the overall histopathological effects on systemic organs and the extent of the damage have not been fully elucidated. Besides, the mechanism that underlies the situation is not very well known. The aim of the present work was to analyse the histopathological effects of OHSS on reproductive (uterus and ovary) and vital organs (liver and kidney) and the possible role of the VEGF-PKA pathway in triggering the condition. Balb/c mice were used to establish an OHSS model. The OHSS group were injected with overdose PMSG while the normal responder group were injected with an optimal dose. Histopathological evaluation was utilised in the liver, kidney, ovary, and uterus stained with hematoxylin and eosin, Masson's trichrome, and periodic acid-Schiff stain. The expression profiles of VEGF (vascular endothelial growth factor), PKA (protein kinase A), and p-PKA (an activated form of PKA) were detected with immunohistochemistry and Western blotting. OHSS was demonstrated to have a negative histopathological effect on all of the organs analysed. These effects were associated with an overall increase in the expression levels of VEGF, PKA, and p-PKA. OHSS has a serious histopathological negative effect on the systemic and reproductive organs and is proven to affect overall health, and thus should be considered a dangerous complication during ART techniques. The activation of the VEGF-PKA pathway, which is indicated by the expression levels of VEGF, PKA, and p-PKA, is demonstrated to accompany this complication, which should be further elucidated to understand the mechanisms underlying the condition.
Collapse
Affiliation(s)
- Seda Karabulut
- Department of Histology and Embryology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Health Science and Technologies Research Institute (SABITA), Istanbul, Turkey
| | - Oya Korkmaz
- Department of Histology and Embryology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Health Science and Technologies Research Institute (SABITA), Istanbul, Turkey
| | - Ceren Erdem Altun
- Department of Histology and Embryology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ilknur Keskin
- Department of Histology and Embryology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Health Science and Technologies Research Institute (SABITA), Istanbul, Turkey
| |
Collapse
|
24
|
Yeo KP, Lim HY, Angeli V. Leukocyte Trafficking via Lymphatic Vessels in Atherosclerosis. Cells 2021; 10:cells10061344. [PMID: 34072313 PMCID: PMC8229118 DOI: 10.3390/cells10061344] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 02/03/2023] Open
Abstract
In recent years, lymphatic vessels have received increasing attention and our understanding of their development and functional roles in health and diseases has greatly improved. It has become clear that lymphatic vessels are critically involved in acute and chronic inflammation and its resolution by supporting the transport of immune cells, fluid, and macromolecules. As we will discuss in this review, the involvement of lymphatic vessels has been uncovered in atherosclerosis, a chronic inflammatory disease of medium- and large-sized arteries causing deadly cardiovascular complications worldwide. The progression of atherosclerosis is associated with morphological and functional alterations in lymphatic vessels draining the diseased artery. These defects in the lymphatic vasculature impact the inflammatory response in atherosclerosis by affecting immune cell trafficking, lymphoid neogenesis, and clearance of macromolecules in the arterial wall. Based on these new findings, we propose that targeting lymphatic function could be considered in conjunction with existing drugs as a treatment option for atherosclerosis.
Collapse
|
25
|
Munoz K, Wasnik S, Abdipour A, Bi H, Wilson SM, Tang X, Ghahramanpouri M, Baylink DJ. The Effects of Insulin-Like Growth Factor I and BTP-2 on Acute Lung Injury. Int J Mol Sci 2021; 22:ijms22105244. [PMID: 34063554 PMCID: PMC8170877 DOI: 10.3390/ijms22105244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury (ALI) afflicts approximately 200,000 patients annually and has a 40% mortality rate. The COVID-19 pandemic has massively increased the rate of ALI incidence. The pathogenesis of ALI involves tissue damage from invading microbes and, in severe cases, the overexpression of inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). This study aimed to develop a therapy to normalize the excess production of inflammatory cytokines and promote tissue repair in the lipopolysaccharide (LPS)-induced ALI. Based on our previous studies, we tested the insulin-like growth factor I (IGF-I) and BTP-2 therapies. IGF-I was selected, because we and others have shown that elevated inflammatory cytokines suppress the expression of growth hormone receptors in the liver, leading to a decrease in the circulating IGF-I. IGF-I is a growth factor that increases vascular protection, enhances tissue repair, and decreases pro-inflammatory cytokines. It is also required to produce anti-inflammatory 1,25-dihydroxyvitamin D. BTP-2, an inhibitor of cytosolic calcium, was used to suppress the LPS-induced increase in cytosolic calcium, which otherwise leads to an increase in proinflammatory cytokines. We showed that LPS increased the expression of the primary inflammatory mediators such as toll like receptor-4 (TLR-4), IL-1β, interleukin-17 (IL-17), TNF-α, and interferon-γ (IFN-γ), which were normalized by the IGF-I + BTP-2 dual therapy in the lungs, along with improved vascular gene expression markers. The histologic lung injury score was markedly elevated by LPS and reduced to normal by the combination therapy. In conclusion, the LPS-induced increases in inflammatory cytokines, vascular injuries, and lung injuries were all improved by IGF-I + BTP-2 combination therapy.
Collapse
Affiliation(s)
- Kevin Munoz
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - Samiksha Wasnik
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - Amir Abdipour
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Division of Nephrology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Hongzheng Bi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China;
| | - Sean M. Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA;
| | - Xiaolei Tang
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY 11548, USA
| | - Mahdis Ghahramanpouri
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - David J. Baylink
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Correspondence: ; Tel.: +909-558-4000-49796; Fax: +(909)-558-0428
| |
Collapse
|
26
|
Merkus D, Muller-Delp J, Heaps CL. Coronary microvascular adaptations distal to epicardial artery stenosis. Am J Physiol Heart Circ Physiol 2021; 320:H2351-H2370. [PMID: 33961506 DOI: 10.1152/ajpheart.00992.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Until recently, epicardial coronary stenosis has been considered the primary outcome of coronary heart disease, and clinical interventions have been dedicated primarily to the identification and removal of flow-limiting stenoses. However, a growing body of literature indicates that both epicardial stenosis and microvascular dysfunction contribute to damaging myocardial ischemia. In this review, we discuss the coexistence of macro- and microvascular disease, and how the structure and function of the distal microcirculation is impacted by the hemodynamic consequences of an epicardial, flow-limiting stenosis. Mechanisms of endothelial dysfunction as well as alterations of smooth muscle function in the coronary microcirculation distal to stenosis are discussed. Risk factors including diabetes, metabolic syndrome, and aging exacerbate microvascular dysfunction in the myocardium distal to a stenosis, and our current understanding of the role of these factors in limiting collateralization and angiogenesis of the ischemic myocardium is presented. Importantly, exercise training has been shown to promote collateral growth and improve microvascular function distal to stenosis; thus, the current literature reporting the mechanisms that underlie the beneficial effects of exercise training in the microcirculation distal to epicardial stenosis is reviewed. We also discuss recent studies of therapeutic interventions designed to improve microvascular function and stimulate angiogenesis in clinically relevant animal models of epicardial stenosis and microvascular disease. Finally, microvascular adaptation to removal of epicardial stenosis is considered.
Collapse
Affiliation(s)
- Daphne Merkus
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine (WBex), University Clinic, LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Munich, Germany.,Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Judy Muller-Delp
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Cristine L Heaps
- Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas.,Michael E. DeBakey Institute for Comparative Cardiovascular Science and Biomedical Devices, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
27
|
Chi Y, Liu X, Chai J. A narrative review of changes in microvascular permeability after burn. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:719. [PMID: 33987417 PMCID: PMC8106041 DOI: 10.21037/atm-21-1267] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective We aimed to review and discuss some of the latest research results related to post-burn pathophysiological changes and provide some clues for future study. Background Burns are one of the most common and serious traumas and consist of a series of pathophysiological changes of thermal injury. Accompanied by thermal damage to skin and soft tissues, inflammatory mediators are released in large quantities. Changes in histamine, bradykinin, and cytokines such as vascular endothelial growth factor (VEGF), metabolic factors such as adenosine triphosphate (ATP), and activated neutrophils all affect the body’s vascular permeability. Methods We searched articles with subject words “microvascular permeability”, “burn” “endothelium”, and “endothelial barrier” in PubMed in English published from the beginning of database to Dec, 2020. Conclusions The essence of burn shock is the rapid and extensive fluid transfer in burn and non-burn tissue. After severe burns, the local and systemic vascular permeability increase, causing intravascular fluid extravasation, leading to a progressive decrease in effective circulation volume, an increase in systemic vascular resistance, a decrease in cardiac output, peripheral tissue edema, multiple organ failure, and even death. There are many cells, tissues, mediators and structures involved in the pathophysiological process of the damage to vascular permeability. Ulinastatin is a promising agent for this problem.
Collapse
Affiliation(s)
- Yunfei Chi
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Xiangyu Liu
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Jiake Chai
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| |
Collapse
|
28
|
Xin C, Zhu C, Jin Y, Li H. Discovering the role of VEGF signaling pathway in mesendodermal induction of human embryonic stem cells. Biochem Biophys Res Commun 2021; 553:58-64. [PMID: 33756346 DOI: 10.1016/j.bbrc.2021.03.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/06/2021] [Indexed: 11/28/2022]
Abstract
Human embryonic stem cells (hESCs) have the unique feature of unlimited self-renewal and differentiation into derivatives of all three germ layers in human body, providing a powerful in vitro model for studying cell differentiation. FGF2, BMP4 and TGF-β signaling have been shown to play crucial roles in mesendodermal differentiation of hESCs. However, their underlying molecular mechanisms and other signaling pathways potentially involved in mesendodermal differentiation of hESCs remain to be further investigated. In this study, we uncover that VEGF signaling pathway plays a critical role in the mesendodermal induction of hESCs. Treating hESCs with Lenvatinib, a pan-inhibitor of VEGF receptors (VEGFRs), impedes their mesendodermal induction. Conversely, overexpression of VEGFA165, a major human VEGF isoform, promotes the mesendodermal differentiation. Similar to the VEGFR inhibitor, MEK inhibitor PD0325901 hinders mesendodermal induction of hESCs. In contrast, overexpression of ERK2GOF, an intrinsically active ERK2 mutant, markedly reduces the inhibitory effect of the VEGFR inhibitor. Thus, the MEK-ERK cascade plays an important role for the function of VEGF signaling pathway in the mesendodermal induction of hESCs. All together, this study identifies the critical role of VEGF signaling pathway as well as potential crosstalk of VEGF signaling pathway with other known signaling pathways in mesendodermal differentiation of hESCs.
Collapse
Affiliation(s)
- Chenge Xin
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaonan Zhu
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Jin
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Basic Clinical Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Hui Li
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Basic Clinical Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
29
|
Haron A, Ruzal M, Shinder D, Druyan S. Hypoxia during incubation and its effects on broiler's embryonic development. Poult Sci 2021; 100:100951. [PMID: 33652530 PMCID: PMC7936217 DOI: 10.1016/j.psj.2020.12.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/06/2020] [Accepted: 12/15/2020] [Indexed: 12/03/2022] Open
Abstract
In all vertebrates, hypoxia plays an important role in fetal development, driving vasculogenesis, angiogenesis, hematopoiesis, and chondrogenesis. Therefore, the ability to sense and respond to changes in the availability of oxygen (O2) is crucial for normal embryonic development as well as for developmental plasticity. Moderate levels of hypoxia trigger a regulated process which leads to adaptive responses. Regulation of angiogenesis by hypoxia is an important component of homeostatic control mechanisms that link the cardio-pulmonary-vascular O2 supply to metabolic demands in local tissues. Hypoxia leads to the activation of genes that are important for cell and tissue adaptation to low O2 conditions, such as hypoxia-inducible factor 1. Previous studies have shown a dose-response effect to hypoxia in chicken embryos, with lower and/or prolonged O2 levels affecting multiple mechanisms and providing a spectrum of responses that facilitate the ability to maintain O2 demand despite environmental hypoxia. In chicken embryos, mild to extreme hypoxia during embryogenesis improves chorioallantoic membrane and cardiovascular development, resulting in an increase in O2 carrying capacity and leading to developmental plasticity that may affect post-hatch chick performance and improve adaptation to additional environmental stresses at suboptimal environmental conditions.
Collapse
Affiliation(s)
- Amit Haron
- Institute of Animal Science, Agricultural Research Organization (ARO), The Volcani Center, Rishon Le Ziyyon 15159, Israel; Faculty of Agriculture Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Mark Ruzal
- Institute of Animal Science, Agricultural Research Organization (ARO), The Volcani Center, Rishon Le Ziyyon 15159, Israel
| | - Dmitry Shinder
- Institute of Animal Science, Agricultural Research Organization (ARO), The Volcani Center, Rishon Le Ziyyon 15159, Israel
| | - Shelly Druyan
- Institute of Animal Science, Agricultural Research Organization (ARO), The Volcani Center, Rishon Le Ziyyon 15159, Israel.
| |
Collapse
|
30
|
WANG KAI, ZHANG FENGTIAN, WEN CHANGLONG, HUANG ZHIHUA, HU ZHIHAO, ZHANG YUWEN, HU FUQIANG, WEN LIJUAN. Regulation of pathological blood-brain barrier for intracranial enhanced drug delivery and anti-glioblastoma therapeutics. Oncol Res 2021. [DOI: 10.32604/or.2022.025696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
|
31
|
Taskaeva I, Bgatova N. Microvasculature in hepatocellular carcinoma: An ultrastructural study. Microvasc Res 2021; 133:104094. [PMID: 33011171 DOI: 10.1016/j.mvr.2020.104094] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most vascularized tumor types, and is characterized by development of heterogeneous immature vessels with increased permeability. Here, we analyzed morphology and vascular permeability-related structures in endothelial cells of HCC microvessels. METHODS Small (Type I) and large (Type II) peritumoral blood microvessels were assessed in HCC-bearing mice. By transmission electron microscopy, endothelial cell cytoplasm area, free transport vesicles, vesiculo-vacuolar organelles and clathrin-coated vesicles were measured. RESULTS The phenotypic changes in the HCC microvessels included presence of sinusoidal capillarization, numerous luminal microprocesses and abnormal luminal channels, irregular dilatations of interendothelial junctions, local detachment of basement membranes and widened extracellular space. Endothelial cells Type I microvessels showed increased vesicular trafficking-related structures. CONCLUSION Ultrastructural characteristics of microvessels Type I can associate with HCC new-formed microvessels. The morphological changes observed in HCC microvessels might explain the increased transcellular and paracellular permeability in HCC endothelial cells.
Collapse
Affiliation(s)
- Iuliia Taskaeva
- Laboratory of Ultrastructural research, Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia; Laboratory of Boron-Neutron Capture Therapy, Department of Physics, Novosibirsk State University, Novosibirsk, Russia.
| | - Nataliya Bgatova
- Laboratory of Ultrastructural research, Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
32
|
Guo C, Chen M, Ma W, Cai B, Xu Y, Zhong Y, Zhou C. Growth differentiation factor 9 inhibits vascular endothelial growth factor expression in human granulosa cells. Gynecol Endocrinol 2020; 36:907-911. [PMID: 31996061 DOI: 10.1080/09513590.2020.1718640] [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] [Indexed: 10/25/2022] Open
Abstract
In aortic endothelial cells, the TGFβ signaling pathway is involved in the regulation of vascular endothelial growth factor (VEGF), which encodes a potent angiogenic factor crucial for the development of ovarian hyperstimulation syndrome. Growth differentiation factor 9 (GDF9) is a member of the TGFβ family and its effect on VEGF expression in human granulosa cells is unknown. In this study, human granulosa cells were collected from patients during the course of oocyte retrieval for in vitro fertilization and were cultured in vitro. After the first 48 h of culture, cells were treated with GDF9 with or without SB431542 (an ALK5 inhibitor) at various doses. The medium was then collected to determine the concentration of VEGF by ELISA. Cellular RNA was collected and extracted for quantification by real-time quantitative fluorescence PCR. Our study showed that GDF9 suppressed VEGF release from human granulosa cells in a dose-dependent manner and also downregulated VEGF mRNA levels in these cells. Furthermore, SB431542 antagonized the suppression of VEGF mRNA by GDF9 and diminished the inhibitory effect of GDF9 on VEGF release by human granulosa cells. Our results indicated that GDF9 can inhibit VEGF expression in human granulosa cells and ALK5 might mediate this process.
Collapse
Affiliation(s)
- Congcong Guo
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Reproductive Medicine Center, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, China
| | - Minghui Chen
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenmin Ma
- Reproductive Medicine Center, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, China
| | - Bing Cai
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanwen Xu
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yiping Zhong
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Canquan Zhou
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Microenvironment remodeled by tumor and stromal cells elevates fibroblast-derived COL1A1 and facilitates ovarian cancer metastasis. Exp Cell Res 2020; 394:112153. [PMID: 32589888 DOI: 10.1016/j.yexcr.2020.112153] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/08/2020] [Accepted: 06/21/2020] [Indexed: 01/25/2023]
Abstract
Wide peritoneal metastasis is the cause of the highest lethality of ovarian cancer in gynecologic malignancies. Ascites play a key role in ovarian cancer metastasis, but involved mechanism is uncertain. Here, we performed a quantitative proteomics of ascites, and found that collagen type I alpha 1 (COL1A1) was notably elevated in ascites from epithelial ovarian cancer patients compared to normal peritoneal fluids, and verified that elevated COL1A1 was mainly originated from fibroblasts. COL1A1 promoted migration and invasion of ovarian cancer cells, but such effects were partially eliminated by COL1A1 antibodies. Intraperitoneally injected COL1A1 accelerated intraperitoneal metastasis of ovarian cancer xenograft in NOD/SCID mice. Further, COL1A1 activated downstream AKT phosphorylation by binding to membrane surface receptor integrin β1 (ITGB1). Knockdown or blockage of ITGB1 reversed COL1A1 enhanced migration and invasion in ovarian cancer cells. Conversely, ovarian cancer ascites and fibrinogen promoted fibroblasts to secrete COL1A1. Elevated fibrinogen in ascites might be associated with increased vascular permeability induced by ovarian cancer. Our findings suggest that microenvironment remodeled by tumor cells and stromal cells promotes fibroblasts to secrete COL1A1 and facilitates the metastasis of ovarian cancer, which may provide a new approach for ovarian cancer therapeutics.
Collapse
|
34
|
Genova T, Gaglioti D, Munaron L. Regulation of Vessel Permeability by TRP Channels. Front Physiol 2020; 11:421. [PMID: 32431625 PMCID: PMC7214926 DOI: 10.3389/fphys.2020.00421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
The vascular endothelium constitutes a semi-permeable barrier between blood and interstitial fluids. Since an augmented endothelial permeability is often associated to pathological states, understanding the molecular basis for its regulation is a crucial biomedical and clinical challenge. This review focuses on the processes controlling paracellular permeability that is the permeation of fluids between adjacent endothelial cells (ECs). Cytosolic calcium changes are often detected as early events preceding the alteration of the endothelial barrier (EB) function. For this reason, great interest has been devoted in the last decades to unveil the molecular mechanisms underlying calcium fluxes and their functional relationship with vessel permeability. Beyond the dicotomic classification between store-dependent and independent calcium entry at the plasma membrane level, the search for the molecular components of the related calcium-permeable channels revealed a difficult task for intrinsic and technical limitations. The contribution of redundant channel-forming proteins including members of TRP superfamily and Orai1, together with the very complex intracellular modulatory pathways, displays a huge variability among tissues and along the vascular tree. Moreover, calcium-independent events could significantly concur to the regulation of vascular permeability in an intricate and fascinating multifactorial framework.
Collapse
Affiliation(s)
- Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Deborah Gaglioti
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| |
Collapse
|
35
|
Peng J, Liang T, Chen C, Zhang Q, Xu Y, Liu J, Zhao P. Subretinal injection of ranibizumab in advanced pediatric vasoproliferative disorders with total retinal detachments. Graefes Arch Clin Exp Ophthalmol 2020; 258:1005-1012. [PMID: 32043167 DOI: 10.1007/s00417-020-04600-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/20/2019] [Accepted: 01/04/2020] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To describe the surgical procedures, outcomes, and complications of a novel technique of subretinal injection of ranibizumab (SRR). METHODS Between September 2012 and September 2018, 37 eyes of 26 consecutive children with vascularly active total retinal detachments in 1 or both eyes treated with SRR as primary treatment were included in this retrospective study. All included eyes received subretinal injection of ranibizumab (0.25 mg/ 0.025 ml). Data included demographics, ocular examination, and anatomic outcomes, following treatment and complications of eyes after SRR were collected. RESULTS Eleven patients had bilateral SRR injections and 15 had monocular SRR injection. Thirteen patients were diagnosed as retinopathy of prematurity. Of all patients, the mean gestational age was 34.5 ± 5.1 weeks (range: 29.6~40.7 weeks), and birth weight was 2328.1 ± 1083.9 g (range: 940~3900 g). On 1-week postoperative follow-up, vascular activity decreased in all 37 eyes (100%). On the 1-month postoperative follow-up, vascular activity decreased but remained in 24 eyes (24/35, 68.6%) of 16 patients and vanished in 11 eyes (11/35, 31.4%) of 9 patients. No eye needed a secondary anti-VEGF therapy. Local subconjunctival hemorrhage was noted in two eyes (2/37, 5.4%). Localized wound leakage of subretinal fluid was also noted in one eye (1/37, 2.7%). CONCLUSIONS In this very limited study, we showed that SRR in vascularly active advanced pediatric vasoproliferative disorders with total retinal detachments is effective and promising, although more extensive controlled trials will be needed to confirm its safety and efficacy.
Collapse
Affiliation(s)
- Jie Peng
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingyi Liang
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunli Chen
- Department of ophthalmology, Tianjin Medical University Eye Hospital & Tianjin Medical University Eye Institute, Tianjin, China.,Department of ophthalmology, Shengli Oilfield Central Hospital, Shan dong Province, China
| | - Qi Zhang
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingjing Liu
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiquan Zhao
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Padhi S, Behera A. Nanotechnology Based Targeting Strategies for the Delivery of Camptothecin. SUSTAINABLE AGRICULTURE REVIEWS 2020. [DOI: 10.1007/978-3-030-41842-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
Evaluation of Vascular Endothelial Growth Factor A and Selected Parameters of Coagulation and Fibrinolysis in a Group of Patients with Subarachnoid Haemorrhage. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8759231. [PMID: 31360727 PMCID: PMC6644279 DOI: 10.1155/2019/8759231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 11/21/2022]
Abstract
Introduction. Subarachnoid hemorrhage (SAH) is currently one of the most serious diseases of the central nervous system. To reduce the negative consequences of SAH and help clinicians to assess the patient's condition, there are attempts to search for new diagnostic markers, which quickly and accurately allow for the proper diagnosis. The aim of this research was the concentration and activity of Vascular Endothelial Growth Factor A (VEGF-A) and selected parameters of coagulation and fibrinolysis in the blood of patients with SAH. Serum levels of VEGF-A in patients diagnosed with SAH are measured to assess the correlation between VEGF-A and the clinical condition of patient. This may help with proper therapeutics and better prognosis. Methods. The study involved 85 patients with subarachnoid hemorrhage. The control group consisted of 45 healthy subjects, sex and age matched. The following parameters were determined: APTT (Activated Partial Thromboplastin Time), INR (International Normalized Ratio), D-dimers and fibrinogen concentration, and the concentration of VEGF-A by ELISA (R&D USA). Results. The average concentration of VEGF-A in the study group was significantly lower compared to the control group. The D-dimer concentration was higher in patients with SAH but the difference was not significant. Coagulation parameters such as INR, APTT, and fibrinogen did not show significant differences between investigated groups. Conclusions. VEGF-A cannot be an independent marker of SAH. Selected parameters of coagulation and fibrinolysis such as D-dimers, INR, APTT, and fibrinogen should not be used as markers of SAH.
Collapse
|
38
|
Wang Q, Deng Y, Huang L, Zeng W, Chen S, Lv B, Jiang W, Han Y, Ding H, Wen M, Zeng H. Hypertonic saline downregulates endothelial cell-derived VEGF expression and reduces blood-brain barrier permeability induced by cerebral ischaemia via the VEGFR2/eNOS pathway. Int J Mol Med 2019; 44:1078-1090. [PMID: 31524227 PMCID: PMC6657973 DOI: 10.3892/ijmm.2019.4262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/28/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to explore the possible mechanisms by which hypertonic saline (HS) effectively ameliorates cerebral oedema via the vascular endothelial growth factor receptor 2 (VEGFR2)-mediated endothelial nitric oxide synthase (eNOS) pathway of endothelial cells in rats. A middle cerebral artery occlusion (MCAO) model in Sprague-Dawley rats and an oxygen-glucose deprivation (OGD) model in cells were used in the present study. Evans blue (EB) staining and a horseradish peroxidase flux assay were performed to evaluate the protective effect of 10% HS on the blood-brain barrier (BBB). The expression levels of vascular endothelial growth factor (VEGF), VEGFR2, zonula occludens 1 (ZO1) and occludin were quantified. The results demonstrated that 10% HS effectively reduced EB extravasation in the peri-ischaemic brain tissue. At 24 h after MCAO, the protein expression levels of VEGF and VEGFR2 in the peri-ischaemic brain tissue were downregulated following treatment with 10% HS. In vitro experiments demonstrated that the permeability of a monolayer endothelial cell barrier was decreased significantly following HS treatment. In addition, VEGF and VEGFR2 protein expression levels were increased in endothelial cells under hypoxic conditions, but that effect was suppressed by HS treatment. Furthermore, HS inhibited the downregulation of ZO1 and occludin effectively, possibly through the VEGFR2/phospholipase C γ1 (PLCγ1)/eNOS signalling pathway. In conclusion, 10% HS may alleviate cerebral oedema through reducing ischaemia-induced BBB permeability, as a consequence of inhibiting VEGFR2/PLCγ1/eNOS-mediated downregulation of ZO1 and occludin.
Collapse
Affiliation(s)
- Qiaosheng Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yiyu Deng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Linqiang Huang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Wenxin Zeng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Shenglong Chen
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Bo Lv
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Wenqiang Jiang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yongli Han
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Hongguang Ding
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Miaoyun Wen
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Hongke Zeng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
39
|
Modern Concepts in Regenerative Therapy for Ischemic Stroke: From Stem Cells for Promoting Angiogenesis to 3D-Bioprinted Scaffolds Customized via Carotid Shear Stress Analysis. Int J Mol Sci 2019; 20:ijms20102574. [PMID: 31130624 PMCID: PMC6566983 DOI: 10.3390/ijms20102574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is associated with a tremendous economic and societal burden, and only a few therapies are currently available for the treatment of this devastating disease. The main therapeutic approaches used nowadays for the treatment of ischemic brain injury aim to achieve reperfusion, neuroprotection and neurorecovery. Therapeutic angiogenesis also seems to represent a promising tool to improve the prognosis of cerebral ischemia. This review aims to present the modern concepts and the current status of regenerative therapy for ischemic stroke and discuss the main results of major clinical trials addressing the effectiveness of stem cell therapy for achieving neuroregeneration in ischemic stroke. At the same time, as a glimpse into the future, this article describes modern concepts for stroke prevention, such as the implantation of bioprinted scaffolds seeded with stem cells, whose 3D geometry is customized according to carotid shear stress.
Collapse
|
40
|
Gaafar S, El-Gezary D, El Maghraby HA. Early onset of cabergoline therapy for prophylaxis from ovarian hyperstimulation syndrome (OHSS): A potentially safer and more effective protocol. Reprod Biol 2019; 19:145-148. [PMID: 31133458 DOI: 10.1016/j.repbio.2019.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/20/2019] [Accepted: 03/28/2019] [Indexed: 01/11/2023]
Abstract
Vascular endothelial growth factor (VEGF) is the most important angiogenic mediator in ovarian hyperstimulation syndrome OHSS. Studies proved that cabergoline administration blocks the increase in vascular permeability via dephosphorylation of VEGF receptors and hence can be used as prophylactic agent against OHSS. This study aimed at evaluating the effectiveness of early administration of cabergoline in the prevention of OHSS in high risk cases prepared for ICSI. This case series study was conducted on 126 high risk patients prepared for ICSI using the fixed antagonist protocol. High risk patients were defined as having more than 20 follicles >12 mm in diameter, and/or E2 more than 3000 pg/ml when the size of the leading follicle is more than 15 mm. When the size of the leading follicle reached 15 mm, cabergoline was administered (0.5 mg/day) for 8 days. Patients were followed up clinically, ultrasonographically and hematologically. The final E2 was 6099.5 ± 2730 and the mean number of retrieved oocytes was 19.7 ± 7.8. The clinical pregnancy rate was 62/126 (49.2%). There were no significant changes (p > 0.05) comparing hematological parameters, renal function tests and liver function tests between the day of HCG and the day of blastocyst transfer. The incidence of severe OHSS in this group was 1/126 (0.9%), while moderate OHSS was 12 (9.5%) and there were no cases of critical OHSS. We concluded that early administration of cabergoline is a safe and potentially more effective approach for prophylaxis against OHSS in high risk cases.
Collapse
Affiliation(s)
- Sherif Gaafar
- Department of Obstetrics, Gynecology and Reproductive Medicine, El Shatby University Hospital, Alexandria University, Egypt.
| | - Dalal El-Gezary
- Clinical Pathology Department, Faculty of Medicine, Alexandria University, Egypt.
| | - Hassan A El Maghraby
- Department of Obstetrics, Gynecology and Reproductive Medicine, El Shatby University Hospital, Alexandria University, Egypt.
| |
Collapse
|
41
|
Ovarian hyperstimulation syndrome: A review for emergency clinicians. Am J Emerg Med 2019; 37:1577-1584. [PMID: 31097257 DOI: 10.1016/j.ajem.2019.05.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/07/2019] [Accepted: 05/07/2019] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION A great deal of literature has recently evaluated the prevention and management of ovarian hyperstimulation syndrome (OHSS) in the outpatient setting, but there remains a dearth of research evaluating OHSS in the emergency department (ED) and its management. OBJECTIVE This narrative review evaluates the underlying pathophysiology and clinical manifestations of OHSS and discusses approaches to patient care in the ED based on current literature. DISCUSSION OHSS is an iatrogenic complication caused by an excessive response to controlled ovarian stimulation during assisted reproductive cycles (ART). OHSS complicates up to 30% of ART cycles, and many of these patients seek initial care in the ED. Risk factors for the development of OHSS include age < 35, history of polycystic ovarian syndrome or previous OHSS, and pregnancy. Emergency physicians will be faced with several complications including ascites, abdominal compartment syndrome, renal dysfunction, acute respiratory distress syndrome, thromboembolic disease, and hemodynamic instability. Critical patients should be evaluated in the resuscitation bay, and consultation with the primary obstetrics/gynecology team is needed, which improves patient outcomes. This review provides several guiding principles for management of OHSS and associated complications. CONCLUSIONS OHSS occurs in up to 30% of IVF cycles and carries a high morbidity. Effective care of the OHSS patient begins with early diagnosis while evaluating for other diseases and complications. Understanding these complications and an approach to the management of OHSS is essential to optimizing patient care.
Collapse
|
42
|
Xiang Z, Jiang G, Yang X, Fan D, Nan X, Li D, Hu Z, Fang Q. Peptosome Coadministration Improves Nanoparticle Delivery to Tumors through NRP1-Mediated Co-Endocytosis. Biomolecules 2019; 9:biom9050172. [PMID: 31060320 PMCID: PMC6572427 DOI: 10.3390/biom9050172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Improving the efficacy of nanoparticles (NPs) delivery to tumors is critical for cancer diagnosis and therapy. In our previous work, amphiphilic peptide APPA self-assembled nanocarriers were designed and constructed for cargo delivery to tumors with high efficiency. In this study, we explore the use of APPA self-assembled peptosomes as a nanoparticle adjuvant to enhance the delivery of nanoparticles and antibodies to integrin αvβ3 and neuropilin-1 (NRP1) positive tumors. The enhanced tumor delivery of coadministered NPs was confirmed by better magnetosome (Mag)-based T2-weighted magnetic resonance imaging (MRI), liposome-based fluorescence imaging, as well as the improved anti-tumor efficacy of monoclonal antibodies (trastuzumab in this case) and doxorubicin (DOX)-containing liposomes. Interestingly, the improvement is most significant for the delivering of compounds that have active or passive tumor targeting ability, such as antibodies or NPs that have enhanced permeability and retention (EPR) effect. However, for non-targeting small molecules, the effect is not significant. In vitro and in vivo studies suggest that both peptosomes and the coadministered compounds might be internalized into cells through a NRP1 mediated co-endocytosis (CoE) pathway. The improved delivery of coadministered NPs and antibodies to tumors suggests that the coadministration with APPA self-assembled peptosomes could be a valuable approach for advancing αvβ3 and NRP1 positive tumors diagnosis and therapy.
Collapse
Affiliation(s)
- Zhichu Xiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Gexuan Jiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiaoliang Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Di Fan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiaohui Nan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Dan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
- Sino-Danish Center for Education and Research, Beijing 101408, China.
| | - Qiaojun Fang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
- Sino-Danish Center for Education and Research, Beijing 101408, China.
- Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Beijing 100190, China.
| |
Collapse
|
43
|
Bekes I, Löb S, Holzheu I, Janni W, Baumann L, Wöckel A, Wulff C. Nectin-2 in ovarian cancer: How is it expressed and what might be its functional role? Cancer Sci 2019; 110:1872-1882. [PMID: 30843637 PMCID: PMC6549928 DOI: 10.1111/cas.13992] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 02/20/2019] [Accepted: 02/28/2019] [Indexed: 12/14/2022] Open
Abstract
Nectin‐2 is an adhesion molecule that has been reported to play a role in tumor growth, metastasis and tumor angiogenesis. Herein, we investigated Nectin‐2 in ovarian cancer patients and in cell culture. Tumor as well as peritoneal biopsies of 60 ovarian cancer patients and 22 controls were dual stained for Nectin‐2 and CD31 using immunohistochemistry. Gene expression of Nectin‐2 was quantified by real‐time PCR and differences analyzed in relation to various tumor characteristics. In the serum of patients, vascular endothelial growth factor (VEGF) was quantified by ELISA. Effect of VEGF on Nectin‐2 expression as well as permeability was investigated in HUVEC. In tumor biopsies, Nectin‐2 protein was mainly localized in tumor cells, whereas in peritoneal biopsies, clear colocalization was found in the vasculature. T3 patients had a significantly higher percentage of positive lymph nodes and this correlated with survival. Nectin‐2 was significantly upregulated in tumor biopsies in patients with lymph node metastasis and with residual tumor >1 cm after surgery. Nectin‐2 expression was significantly suppressed in the peritoneal endothelium of patients associated with significantly increased VEGF serum levels. In cell culture, VEGF stimulation led to a significant downregulation of Nectin‐2 which was reversed by VEGF‐inhibition. In addition, Nectin‐2 knockdown in endothelial cells was associated with significantly increased endothelial permeability. Nectin‐2 expression in ovarian cancer may support tumor cell adhesion, leading to growth and lymph node metastasis. In addition, VEGF‐induced Nectin‐2 suppression in peritoneal endothelium may support an increase in vascular permeability leading to ascites production.
Collapse
Affiliation(s)
- Inga Bekes
- Department of Obstetrics and Gynecology, University of Ulm, Ulm, Germany
| | - Sanja Löb
- Department of Obstetrics and Gynecology, University of Würzburg, Würzburg, Germany
| | - Iris Holzheu
- Department of Obstetrics and Gynecology, University of Ulm, Ulm, Germany
| | - Wolfgang Janni
- Department of Obstetrics and Gynecology, University of Ulm, Ulm, Germany
| | - Lisa Baumann
- Department of Pathology, University of Ulm, Ulm, Germany
| | - Achim Wöckel
- Department of Obstetrics and Gynecology, University of Würzburg, Würzburg, Germany
| | - Christine Wulff
- Department of Obstetrics and Gynecology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
44
|
Abstract
A wide variety of pathogens reach the circulatory system during viral, parasitic, fungal, and bacterial infections, causing clinically diverse pathologies. Such systemic infections are usually severe and frequently life-threatening despite intensive care, in particular during the age of antibiotic resistance. Because of its position at the interface between the blood and the rest of the organism, the endothelium plays a central role during these infections. Using several examples of systemic infections, we explore the diversity of interactions between pathogens and the endothelium. These examples reveal that bacterial pathogens target specific vascular beds and affect most aspects of endothelial cell biology, ranging from cellular junction stability to endothelial cell proliferation and inflammation.
Collapse
|
45
|
Park KS, Park DH. The effect of Korean Red Ginseng on full-thickness skin wound healing in rats. J Ginseng Res 2019; 43:226-235. [PMID: 30976160 PMCID: PMC6438823 DOI: 10.1016/j.jgr.2017.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 12/05/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Panax ginseng is regarded as one of the best compounds for promoting health, and it has been used traditionally as a medicinal herb. Recently, Korean Red Ginseng (RG) has been shown to protect skin from aging and wrinkling; it can also relieve atopic dermatitis and allergy symptoms. This study aimed to evaluate RG's effects on the regeneration of the full-thickness skin wounds in rat. METHODS Full-thickness skin wounds were generated in rats, and then RG was administered either orally or topically. The wound-healing effects of RG were investigated by assessing wound size, mRNA expression patterns of genes related to wound healing, histological staining, and measurements of lipid, moisture, and elasticity in skin tissues. RESULTS The wound size was smaller, and tissue regeneration rate was faster in the RG-treated group than that in the control group on days 15 and 20 after initiating treatment. On postoperative day 20, skin lipid and moisture content had increased significantly in the RG-treated group. Significant increases in the gene expression levels of transforming growth factor-β1 and vascular endothelial growth factor were found in the RG group during the early stages of wound healing. Matrix metalloproteinase-1 and matrix metalloproteinase-9 showed significant increases in gene expression levels on day 20. CONCLUSION The results suggested that RG may promote healing of full-thickness skin wounds in rats. They also provided basic insights into the effects of RG on skin regeneration, supporting its use as a dressing material for wound treatment and its development as a functional food.
Collapse
Affiliation(s)
| | - Dae-Hwan Park
- Department of Plastic and Reconstructive Surgery, College of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
46
|
Modulation of Receptor Tyrosine Kinase Activity through Alternative Splicing of Ligands and Receptors in the VEGF-A/VEGFR Axis. Cells 2019; 8:cells8040288. [PMID: 30925751 PMCID: PMC6523102 DOI: 10.3390/cells8040288] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/14/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) signaling is essential for physiological and pathological angiogenesis. Alternative splicing of the VEGF-A pre-mRNA gives rise to a pro-angiogenic family of isoforms with a differing number of amino acids (VEGF-Axxxa), as well as a family of isoforms with anti-angiogenic properties (VEGF-Axxxb). The biological functions of VEGF-A proteins are mediated by a family of cognate protein tyrosine kinase receptors, known as the VEGF receptors (VEGFRs). VEGF-A binds to both VEGFR-1, largely suggested to function as a decoy receptor, and VEGFR-2, the predominant signaling receptor. Both VEGFR-1 and VEGFR-2 can also be alternatively spliced to generate soluble isoforms (sVEGFR-1/sVEGFR-2). The disruption of the splicing of just one of these genes can result in changes to the entire VEGF-A/VEGFR signaling axis, such as the increase in VEGF-A165a relative to VEGF-A165b resulting in increased VEGFR-2 signaling and aberrant angiogenesis in cancer. Research into this signaling axis has recently focused on manipulating the splicing of these genes as a potential therapeutic avenue in disease. Therefore, further research into understanding the mechanisms by which the splicing of VEGF-A/VEGFR-1/VEGFR-2 is regulated will help in the development of drugs aimed at manipulating splicing or inhibiting specific splice isoforms in a therapeutic manner.
Collapse
|
47
|
Poret JM, Battle C, Mouton AJ, Gaudet DA, Souza-Smith F, Gardner JD, Braymer HD, Harrison-Bernard L, Primeaux SD. The prevalence of cardio-metabolic risk factors is differentially elevated in obesity-prone Osborne-Mendel and obesity-resistant S5B/Pl rats. Life Sci 2019; 223:95-101. [PMID: 30872180 DOI: 10.1016/j.lfs.2019.03.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 01/08/2023]
Abstract
AIMS Individual susceptibility to develop obesity may impact the development of cardio-metabolic risk factors that lead to obesity-related comorbid conditions. Obesity-prone Osborne-Mendel (OM) rats expressed higher levels of visceral adipose inflammation than obesity-resistant, S5B/Pl (S5B) rats. However, the consumption of a high fat diet (HFD) differentially affected OM and S5B rats and induced an increase in visceral adipose inflammation in S5B rats. The current study examined the effects of HFD consumption on cardio-metabolic risk factors in OM and S5B rats. MATERIALS & METHODS Glucose regulation and circulating levels of lipids, adiponectin and C-reactive protein were assessed following 8 weeks of HFD or low fat diet (LFD) consumption. Left ventricle hypertrophy and mRNA expression of cardiovascular disease biomarkers were also quantified in OM and S5B rats. KEY FINDINGS Circulating levels of triglycerides were higher, while HDL cholesterol, adiponectin and glycemic control were lower in OM rats, compared to S5B rats. In the left ventricle, BNP and CTGF mRNA expression were higher in OM rats and IL-6, IL-1β, VEGF, and iNOS mRNA expression were higher in S5B rats. SIGNIFICANCE These findings support the hypothesis that cardio-metabolic risk factors are increased in obesity-prone individuals, which may increase the risk for the development of obesity-related comorbidities. In the current models, obesity-resistant S5B rats also exhibited cardiovascular risk factors supporting the importance of monitoring cardiovascular health in individuals characterized as obesity-resistant.
Collapse
Affiliation(s)
- Jonquil M Poret
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - Claire Battle
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - Alan J Mouton
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - Darryl A Gaudet
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - Flavia Souza-Smith
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - Jason D Gardner
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - H Douglas Braymer
- Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, LA 70808, United States of America
| | - Lisa Harrison-Bernard
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America
| | - Stefany D Primeaux
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, United States of America; Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, LA 70808, United States of America.
| |
Collapse
|
48
|
Xiang Z, Yang X, Jiang G, Fan D, Geng L, Wang H, Hu Z, Fang Q. Design of a Simple and Practical Nanosystem Coordinates Tumor Targeting and Penetration for Improved Theranostics. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Zhichu Xiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Xiaoliang Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Gexuan Jiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Di Fan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Lingling Geng
- Xuanwu Hospital; Capital Medical University; Beijing 100053 China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
- Sino-Danish Center for Education and Research; Beijing 101408 China
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
- Sino-Danish Center for Education and Research; Beijing 101408 China
| | - Qiaojun Fang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; Beijing 100190 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology; Beijing 100190 China
- University of Chinese Academy of Sciences; Beijing 100049 China
- Sino-Danish Center for Education and Research; Beijing 101408 China
- Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques; National Center for Nanoscience and Technology; Beijing 100190 China
| |
Collapse
|
49
|
Mugisho OO, Green CR, Zhang J, Acosta ML, Rupenthal ID. Connexin43 hemichannels: A potential drug target for the treatment of diabetic retinopathy. Drug Discov Today 2019; 24:1627-1636. [PMID: 30690195 DOI: 10.1016/j.drudis.2019.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 02/07/2023]
Abstract
Diabetic retinopathy (DR) is a chronic vascular disease of the retina that causes vision loss in patients with type 1 and type 2 diabetes, and is associated with vascular dysfunction and occlusion, retinal oedema, haemorrhage and inadequate growth of new blood vessels. Current DR therapies primarily target downstream, later-stage vascular defects with a significant proportion of diabetic macular oedema patients being non-responders. Moreover, other evidence suggests that prolonged use of therapies targeting vascular endothelial growth factor (VEGF) might be associated with increased onset of geographic atrophy and retinal ganglion cell death. It is therefore highly desirable to prevent the onset of DR or arrest its progression at a stage preceding the appearance of more-advanced pathology by targeting upstream disease mechanisms. Connexin43 hemichannels play a part in the pathogenesis of chronic inflammatory diseases, including inflammasome pathway activation; and hemichannel block has been shown to alleviate vascular leak and inflammation. This review discusses the inflammatory changes occurring in DR as well as current therapies and their limitations. It then focuses on the role of connexin43 in DR, providing evidence for the utility of connexin43 hemichannel blockers as novel therapeutics for DR treatment.
Collapse
Affiliation(s)
- Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Jie Zhang
- Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Monica L Acosta
- School of Optometry and Vision Science and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
50
|
Benelhaj NE, Maraveyas A, Featherby S, Collier MEW, Johnson MJ, Ettelaie C. Alteration in endothelial permeability occurs in response to the activation of PAR2 by factor Xa but not directly by the TF-factor VIIa complex. Thromb Res 2019; 175:13-20. [PMID: 30677622 DOI: 10.1016/j.thromres.2019.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 01/09/2023]
Abstract
Alterations in the endothelial permeability occur in response to the activation of coagulation mechanisms in order to control clot formation. The activation of the protease activated receptors (PAR) can induce signals that regulate such cellular responses. PAR2 is a target for the coagulation factor Xa (fXa) and tissue factor-factor VIIa (TF-fVIIa) complex. By measuring the permeability of dextran blue across endothelial monolayer, we examined the mechanisms linking coagulation and endothelial permeability. Activation of PAR2 using the agonist peptide (PAR2-AP) resulted in increased permeability across the monolayer and was comparable to that obtained with VEGF at 60 min. Incubation of cells with activated factor Xa (fXa) resulted in an initial decrease in permeability by 30 min, but then significantly increased at 60 min. These responses required fXa activity, and were abrogated by incubation of the cells with a PAR2-blocking antibody (SAM11). Activation of PAR2 alone, or inhibition of PAR1, abrogated the initial reduction in permeability. Additionally, inclusion of Rivaroxaban (0.6 μg/ml) significantly inhibited the response to fXa. Finally, incubation of the endothelial monolayers up to 2 h with TF-containing microvesicles derived from MDA-MB-231 cells, in the presence or absence of fVIIa, did not influence the permeability across the monolayers. In conclusion, fXa but not TF-fVIIa is a noteworthy mediator of endothelial permeability. The rapid initial decrease in permeability requires PAR2 and PAR1 which may act to constrain bleeding. The longer-term response is mediated by PAR2 with increased permeability, presumably to enhance clot formation at the site of damage.
Collapse
Affiliation(s)
- Naima E Benelhaj
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Anthony Maraveyas
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Sophie Featherby
- Biomedical Section, School of Life Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Mary E W Collier
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester LE3 9QP, UK
| | - Miriam J Johnson
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Camille Ettelaie
- Biomedical Section, School of Life Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK.
| |
Collapse
|