1
|
Cicinelli E, Nicolì P, Vimercati A, Cicinelli R, Marinaccio M, Matteo M, de Ziegler D, Vitagliano A. High prevalence of chronic endometritis in women with nonstructural abnormal uterine bleeding and benefits of antimicrobial treatment on blood loss pattern: A prospective, observational study. Int J Gynaecol Obstet 2025. [PMID: 40249452 DOI: 10.1002/ijgo.70115] [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: 11/21/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVE The causal link between chronic endometritis (CE) and nonstructural abnormal uterine bleeding (AUB) has been poorly investigated and requires further clarification. This study aimed to evaluate the prevalence of CE in women with nonstructural AUB and to assess the effect of CE cure on the menstrual blood loss pattern. METHODS This prospective study was conducted between 2022 and 2024 at the Department of Obstetrics and Gynecology, University of Bari, Italy. Women aged 20-45 undergoing hysteroscopy for AUB with confirmation of nonstructural causes were evaluated for CE and considered affected when both hysteroscopic and histologic/immunohistochemical (HIS/IHC) criteria were met. Women with CE were treated with specific culture-guided therapy. Cure ascertainment was confirmed by triple negativity at hysteroscopy-HIS/IHC-culture (Group A). In cases of a positive test, a second course of therapy, up to three courses, was repeated before declaring a persistent condition (Group B). At enrollment and at end-of-treatment evaluation, all participants completed a questionnaire to describe the bleeding characteristics and the Pictorial Blood Assessment Chart (PBAC). Serum hemoglobin and ferritin were also assessed. RESULTS Chronic endometritis was diagnosed in 102 of 145 (70.3%) women with nonstructural AUB enrolled in the study. Out of 102 CE patients, 81 (79.4%) patients showed CE resolution after therapy (group A), while in 21 patients (20.6%) CE was persistent (group B). The duration of heavy bleeding before treatment (baseline) was similar in both groups (P = ns). In contrast, at the end of treatment, days of heavy bleeding, days of spotting, and PBAC scores decreased significantly in group A compared with both same-group baseline assessment and group B. Serum hemoglobin and ferritin levels in cured women (A) were significantly higher than in those with persistent CE (B). PBAC scores in group A at 3 and 6 months after the end of treatment showed persistence of cure benefits on AUB. Finally, among patients with CE diagnosis, both univariate and multivariate regressions showed a significant association between cure of CE and reduced bleeding. CONCLUSION Chronic endometritis was highly prevalent in women with nonstructural AUB in our cohort. Cure of CE by targeted antimicrobial therapy led to significant improvements in bleeding patterns.
Collapse
Affiliation(s)
- Ettore Cicinelli
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro," Policlinico of Bari, Bari, BA, Italy
| | - Pierpaolo Nicolì
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro," Policlinico of Bari, Bari, BA, Italy
| | - Antonella Vimercati
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro," Policlinico of Bari, Bari, BA, Italy
| | - Rossana Cicinelli
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Hospital Foch, the University of Paris Ouest (UVSQ), Paris, France
| | - Marco Marinaccio
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro," Policlinico of Bari, Bari, BA, Italy
| | - Maria Matteo
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Dominique de Ziegler
- Fertility Clinic of Cambodia, Phnom Penh, Kingdom of Cambodia
- Department of Obstetrics and Gynecology, Hopital Foch-Université de Paris Ouest (UVSQ), Paris, France
| | - Amerigo Vitagliano
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro," Policlinico of Bari, Bari, BA, Italy
| |
Collapse
|
2
|
Peng H, Du Z, Li J, Wang W, Li Z, Ru S. The sprouting angiogenesis and vascular dysfunction triggered by bisphenol S and tetrabromobisphenol S through disrupting vascular endothelial-cadherin in zebrafish. ENVIRONMENTAL RESEARCH 2025; 278:121632. [PMID: 40246265 DOI: 10.1016/j.envres.2025.121632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Exogenous chemical toxicants may be important inducers of pathological angiogenesis diseases. However, few studies have investigated the associations between pathological angiogenesis diseases and chemical toxicant exposures, and the specific mechanism by which chemical toxicants induce sprouting angiogenesis is unclear. In this study, zebrafish were exposed to bisphenol S (BPS, 1-100 μg/L) and tetrabromobisphenol S (TBBPS, 0.1 and 10 μg/L) from the embryonic stage to the larval stage to investigate how pollutants interfere with angiogenesis and the function of ectopic sprouting vessels. The results showed that BPS and TBBPS promoted ectopic sprouting angiogenesis in different types of vascular plexuses, including the posterior cardinal vein (PCV) and superficial choroidal vessels (SOVs), at different developmental time points. Proteomic analyses of eGFP-positive endothelial cells (ECs) isolated from Tg(flk1: eGFP) zebrafish revealed that both BPS and TBBPS induced ectopic angiogenesis by acting on vascular endothelial-cadherin (VE-cadherin) and activating downstream proangiogenic signaling. In ectopic sprouting vessels induced by BPS and TBBPS, increased endothelial permeability resulted in white blood cell recruitment. Human oxidized lipids also tended to deposit in these ectopic vessels following BPS and TBBPS exposure. These findings suggest that chemical toxicant-induced ectopic angiogenesis is an important cause of vascular dysfunction and related diseases.
Collapse
Affiliation(s)
- Hongyuan Peng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Zehui Du
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Jiali Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| | - Ze Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
3
|
Severino P, D'Amato A, Prosperi S, Myftari V, Germanò R, Marek-Iannucci S, De Prisco A, Mariani MV, Marchiori L, Battaglia C, Tabacco L, Segato C, Mancone M, Fedele F, Vizza CD. Coronary microcirculation in myocardial ischemia: A genetic perspective. J Mol Cell Cardiol 2025; 203:67-75. [PMID: 40220989 DOI: 10.1016/j.yjmcc.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Coronary microvascular dysfunction (CMD) is a major contributor to ischemic heart disease (IHD), acting both independently and together with atherosclerosis. CMD encompasses structural and functional microcirculatory changes that result in dysregulated coronary blood flow. Structural abnormalities include microvascular remodeling, resulting in arteriolar and capillary narrowing, perivascular fibrosis and capillary rarefaction. Endothelial dysfunction and smooth muscle cell hyperactivity further impair microcirculation. Genetic factors may play a crucial role in the pathophysiology of CMD, mainly due to single nucleotide polymorphisms (SNPs) in genes that regulate coronary blood flow and microcirculation structural modifications. This manuscript aims to review the genetic determinants of CMD, with particular focus on ion channels, microRNAs (miRNAs), and proteins involved in the endothelial environment. The improving knowledge about genetic aspects of CMD opens the possibility to have new biomarkers, improving diagnosis and the development of targeted treatments in light of an even more patient-tailored approach.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea D'Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Rosanna Germanò
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefanie Marek-Iannucci
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea De Prisco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Valerio Mariani
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ludovica Marchiori
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Corinne Battaglia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Leonardo Tabacco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Camilla Segato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | | | - Carmine Dario Vizza
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
4
|
Liu ZN, Shen YL, Dong HJ, Tan KX, Li J, Peng YM, Cui HJ. Management of Regorafenib-Induced Hand-Foot Skin Reaction with Topical Chinese Medicine and Urea Ointment: A Case Report and Literature Review. Onco Targets Ther 2025; 18:509-519. [PMID: 40225901 PMCID: PMC11994073 DOI: 10.2147/ott.s510766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Regorafenib, a multikinase inhibitor, frequently induces severe hand-foot skin reactions (HFSR), often requiring dose reduction or discontinuation. This case report demonstrates the successful management of HFSR in a patient with fibromyxoid sarcoma using topical Chinese medicine "Shouzuping" soaking combined with urea ointment. It suggests the unique advantages of integrated traditional Chinese and Western medicine in managing HFSR. This article further reviews the clinical characteristics, pathogenesis, prevention and treatment strategies of HFSR caused by targeted therapies, with a view to providing valuable clinical insights.
Collapse
Affiliation(s)
- Zhe-Ning Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Yu-Lei Shen
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Hui-Jing Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Ke-Xin Tan
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Jia Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Yan-Mei Peng
- Oncology Department, Beijing Fangshan District Traditional Chinese Medicine Hospital, Beijing, 102401, People’s Republic of China
| | - Hui-Juan Cui
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| |
Collapse
|
5
|
Shi H, Yang H, Wu C, Wang S, He S, Chen L, Chan YK, Lai S, Liang K, Deng Y. Glucose-triggered NO-evolving coating bestows orthopedic implants with enhanced anti-bacteria and angiectasis for safeguarding diabetic osseointegration. Biomaterials 2025; 321:123334. [PMID: 40239593 DOI: 10.1016/j.biomaterials.2025.123334] [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: 02/19/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
As a common chronic metabolic disease, diabetes mellitus (DM) features a hyperglycemic micromilieu around implants, resulting in the critical implantation failure and high complications such as peri-implantitis and angiectasis disorder. To address the plaguing issue, we devise and develop a glucose-unlocked NO-evolving orthopedic implant consisted of polyetheretherketone (PEEK), glucose oxidase (GOx) and l-arginine (Arg) with enhanced angiogenesis for boosting diabetic osseointegration. Upon hyperglycemic niche, GOx on implants catalytically exhaust glucose to H2O2, which immediately reacts with Arg to in situ liberate nitric oxide (NO), resulting in enhanced angiogenesis and angiectasis around PEEK implant. Besides, the engineered implant exhibits great anti-bacterial properties against both Gram-positive and Gram-negative bacteria, as well as fortifies osteogenicity of osteoblasts in terms of cell proliferation, alkaline phosphatase activity and calcium matrix mineralization. Intriguingly, in vivo evaluations utilizing diabetic infectious bone defect models of rat further authenticate that the engineered implants substantially augment bone remodeling and osseointegration at weeks 4 and 8 through dampening pathogens, anti-inflammatory as well as promoting angiectasis. Altogether, this work proposed a new tactic to remedy stalled diabetic osseointegration with hyperglycemic micromilieu-responsive therapeutic gas-evolving orthopedic implants.
Collapse
Affiliation(s)
- Hongxing Shi
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Hao Yang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Chao Wu
- Institute of Digital Medicine, Zigong Academy of Big Data for Medical Science and Artificial Intelligence, Department of Orthopedics, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Song Wang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Shuai He
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Lin Chen
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Shuangquan Lai
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Kunneng Liang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China; Department of Cardiology and Endodontics, State Key Laboratory of Oral Disease, West China Hospital, Sichuan University, Chengdu, 610065, China.
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China; National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China; Department of Mechanical Engineering, The University of Hong Kong, 999077, Hong Kong, China.
| |
Collapse
|
6
|
Elhemiely AA, Elesawy WH. Modulation of VEGF/eNOS/TGF-β Axis by Piracetam as a New Avenue to Ameliorate Valproic Acid-Induced Placental Toxicity and Teratogenicity in Rats. J Biochem Mol Toxicol 2025; 39:e70266. [PMID: 40226982 DOI: 10.1002/jbt.70266] [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: 02/26/2024] [Revised: 02/19/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
Valproic acid (VPA) is a very effective therapy used to treat generalized epilepsy, but it must be avoided during pregnancy as it leads to a high risk of teratogenesis. Its teratogenic effect is believed to be due to its placental toxic effect, altering angiogenesis and inducing oxidative stress. Piracetam (PIRA) is a derivative of the neurotransmitter γ-aminobutyric acid (GABA) and has anti-oxidative and pro-angiogenic features. However, its effects against Valproic acid-evoked placental toxicity and abnormal fetal development have not been mechanistically examined. Herein, the present study targets angiogenesis and oxidative stress by Piracetam to investigate the potential modulation of Valproic acid-induced placental toxicity and abnormal fetal development in rats. After administration of Valproic acid (500 mg/kg/day, orally) and/or piracetam (500 mg/kg/day, orally) from the 6th to 15th of gestation, fetuses and placenta were obtained for analysis. The present findings revealed that Piracetam improved the histopathological lesions in the placenta and restored the labyrinth zone area percent. Moreover, it improved the intra-uterine growth retardation (IUGR) via restoring fetal body weight and length and also ameliorated all external malformations (subcutaneous hemorrhage, fore limb, and hind limb anomalies) and additionally amended the skeletal lack of ossification. These favorable effects of Piracetam were mediated by the enhancement of placental angiogenesis via the VEGF/eNOS/TGF-β pathway and attenuating placental oxidative stress, which appeared as decreased MDA content and increased GSH and TAC levels. In conclusion, activation of placental angiogenesis via the VEGF/eNOS/TGF-β axis alongside inhibition of oxidative stress by Piracetam can ameliorate Valproic acid-evoked placental toxicity and, subsequently, fetal malformations in rats.
Collapse
Affiliation(s)
- Alzahraa A Elhemiely
- Department of Pharmacology, Egyptian Drug Authority, EDA, Formerly NODCAR, Giza, Egypt
| | - Wessam H Elesawy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| |
Collapse
|
7
|
Rhim WK, Woo J, Kim JY, Lee EH, Cha SG, Kim DS, Baek SW, Park CG, Kim BS, Kwon TG, Han DK. Multiplexed PLGA scaffolds with nitric oxide-releasing zinc oxide and melatonin-modulated extracellular vesicles for severe chronic kidney disease. J Adv Res 2025; 69:75-89. [PMID: 38537702 PMCID: PMC11954823 DOI: 10.1016/j.jare.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION With prevalence of chronic kidney disease (CKD) in worldwide, the strategies to recover renal function via tissue regeneration could provide alternatives to kidney replacement therapies. However, due to relatively low reproducibility of renal basal cells and limited bioactivities of implanted biomaterials along with the high probability of substance-inducible inflammation and immunogenicity, kidney tissue regeneration could be challenging. OBJECTIVES To exclude various side effects from cell transplantations, in this study, we have induced extracellular vesicles (EVs) incorporated cell-free hybrid PMEZ scaffolds. METHODS Hybrid PMEZ scaffolds incorporating essential bioactive components, such as ricinoleic acid grafted Mg(OH)2 (M), extracellular matrix (E), and alpha lipoic acid-conjugated ZnO (Z) based on biodegradable porous PLGA (P) platform was successfully manufactured. Consecutively, for functional improvements, melatonin-modulated extracellular vesicles (mEVs), derived from the human umbilical cord MSCs in chemically defined media without serum impurities, were also loaded onto PMEZ scaffolds to construct the multiplexed PMEZ/mEV scaffold. RESULTS With functionalities of Mg(OH)2 and extracellular matrix-loaded PLGA scaffolds, the continuous nitric oxide-releasing property of modified ZnO and remarkably upregulated regenerative functionalities of mEVs showed significantly enhanced kidney regenerative activities. Based on these, the structural and functional restoration has been practically achieved in 5/6 nephrectomy mouse models that mimicked severe human CKD. CONCLUSION Our study has proved the combinatory bioactivities of the biodegradable PLGA-based multiplexed scaffold for kidney tissue regeneration in 5/6 nephrectomy mouse representing a severe CKD model. The optimal microenvironments for the morphogenetic formations of renal tissues and functional restorations have successfully achieved the combinatory bioactivities of remarkable components for PMEZ/mEV, which could be a promising therapeutic alternative for CKD treatment.
Collapse
Affiliation(s)
- Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jiwon Woo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Tae Gyun Kwon
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea.
| |
Collapse
|
8
|
Warden C, Zubieta D, Brantley MA. Citrulline Plus Arginine Induces an Angiogenic Response and Increases Permeability in Retinal Endothelial Cells via Nitric Oxide Production. Int J Mol Sci 2025; 26:2080. [PMID: 40076703 PMCID: PMC11900006 DOI: 10.3390/ijms26052080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
We previously observed elevated plasma levels of citrulline and arginine in diabetic retinopathy patients compared to diabetic controls. We tested our hypothesis that citrulline plus arginine induces angiogenesis and increases permeability in retinal endothelial cells. Human retinal microvascular endothelial cells (HRMECs) were treated with citrulline, arginine, or citrulline + arginine, and angiogenesis was measured with cell proliferation, migration, and tube formation assays. Permeability was measured in HRMEC monolayers via trans-endothelial electrical resistance (TEER) and FITC-labeled dextran. We also measured arginase activity, arginase-1 and arginase-2 expression, protein expression and phosphorylation of endothelial nitric oxide synthase (eNOS), and nitric oxide (NO) production. Citrulline + arginine induced endothelial cell proliferation (p = 0.018), migration (p = 0.011), and tube formation (p = 0.0042). Citrulline + arginine also increased FITC-dextran flow-through (p = 1.5 × 10-5) and decreased TEER (p = 0.010). Citrulline + arginine had no effect on arginase activity, but it increased eNOS (p = 6.3 × 10-4) and phosphorylated eNOS (p = 0.029), as well as NO production (p = 0.025). Inhibiting eNOS prevented the increase in NO (p = 0.0092), inhibited citrulline + arginine-induced cell migration (p = 0.0080) and tube formation (p = 0.0092), and blocked citrulline + arginine-related alterations in FITC-dextran flow-through (p = 3.6 × 10-4) and TEER (p = 3.9 × 10-4). These data suggest that citrulline + arginine treatment induces angiogenesis and increases permeability in retinal endothelial cells by activating eNOS and increasing NO production.
Collapse
Affiliation(s)
| | | | - Milam A. Brantley
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.W.); (D.Z.)
| |
Collapse
|
9
|
Wayne N, Singamneni VS, Venkatesh R, Cherlin T, Verma SS, Guerraty MA. Genetic Insights Into Coronary Microvascular Disease. Microcirculation 2025; 32:e12896. [PMID: 39755372 DOI: 10.1111/micc.12896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Coronary microvascular disease (CMVD) affects the coronary pre-arterioles, arterioles, and capillaries and can lead to blood supply-demand mismatch and cardiac ischemia. CMVD can present clinically as ischemia or myocardial infarction with no obstructive coronary arteries (INOCA or MINOCA, respectively). Currently, therapeutic options for CMVD are limited, and there are no targeted therapies. Genetic studies have emerged as an important tool to gain rapid insights into the molecular mechanisms of human diseases. For example, coronary artery disease (CAD) genome-wide association studies (GWAS) have enrolled hundreds of thousands of patients and have identified > 320 loci, elucidating CAD pathogenic pathways and helping to identify therapeutic targets. Here, we review the current landscape of genetic studies of CMVD, consisting mostly of genotype-first approaches. We then present the hypothesis that CAD GWAS have enrolled heterogenous populations and may be better characterized as ischemic heart disease (IHD) GWAS. We discuss how several of the genetic loci currently associated with CAD may be involved in the pathogenesis of CMVD. Genetic studies could help accelerate progress in understanding CMVD pathophysiology and identifying putative therapeutic targets. Larger phenotype-first genomic studies into CMVD with adequate sex and ancestry representation are needed. Given the extensive CAD genetic and functional validation data, future research should leverage these loci as springboards for CMVD genomic research.
Collapse
Affiliation(s)
- Nicole Wayne
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Venkata S Singamneni
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rasika Venkatesh
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tess Cherlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Marie A Guerraty
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Yu J, Li Y, Hu J, Wang Y. Interleukin-33 induces angiogenesis after myocardial infarction via AKT/eNOS signaling pathway. Int Immunopharmacol 2024; 143:113433. [PMID: 39486188 DOI: 10.1016/j.intimp.2024.113433] [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: 08/08/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality and morbidity worldwide. MI-damaged vascular structures are difficult to completely restore due to the heart's low regenerative capacity. Given interleukin-33 (IL-33) as a potent endothelial activator promoting angiogenesis, this study investigated the role of IL-33 in angiogenesis and cardiac repair after MI. A mouse model of MI was established. IL-33 improved cardiac function and induced an increase in vascular density after MI. Besides, IL-33 promoted human endothelial cells proliferation, migration, and differentiation under both normoxic and hypoxic conditions, consistently with increased angiogenesis in vivo. Mechanistic studies demonstrated that IL-33 could promote angiogenesis by activating eNOS and AKT, and stimulating NO production in vivo and in vitro. Given that injection of exogenous IL-33 induced an inflammatory response, we employed a multifunctional biomimetic nanoparticle drug delivery system to deliver IL-33, thereby enhancing its targeting to the heart for fibrotic therapy and reducing inflammation. In conclusion, our results indicate that IL-33 promotes endothelial angiogenesis after MI through AKT/eNOS/NO signaling pathway. PM&EM/IL-33 nanoparticles may hold promising therapeutic potential for protecting cardiac ischemic injury and mitigating inflammation.
Collapse
Affiliation(s)
- Jiaqi Yu
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| | - Yuyu Li
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| |
Collapse
|
11
|
Villadangos L, Serrador JM. Subcellular Localization Guides eNOS Function. Int J Mol Sci 2024; 25:13402. [PMID: 39769167 PMCID: PMC11678294 DOI: 10.3390/ijms252413402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide synthases (NOS) are enzymes responsible for the cellular production of nitric oxide (NO), a highly reactive signaling molecule involved in important physiological and pathological processes. Given its remarkable capacity to diffuse across membranes, NO cannot be stored inside cells and thus requires multiple controlling mechanisms to regulate its biological functions. In particular, the regulation of endothelial nitric oxide synthase (eNOS) activity has been shown to be crucial in vascular homeostasis, primarily affecting cardiovascular disease and other pathophysiological processes of importance for human health. Among other factors, the subcellular localization of eNOS plays an important role in regulating its enzymatic activity and the bioavailability of NO. The aim of this review is to summarize pioneering studies and more recent publications, unveiling some of the factors that influence the subcellular compartmentalization of eNOS and discussing their functional implications in health and disease.
Collapse
Affiliation(s)
| | - Juan M. Serrador
- Interactions with the Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas (CSIC)—Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
12
|
Kong AM, Idris ZA, Urrutia-Cabrera D, Lees JG, Phang RJ, Mitchell GM, Wong RC, Lim SY. NOS3 regulates angiogenic potential of human induced pluripotent stem cell-derived endothelial cells. Biochem Biophys Rep 2024; 40:101876. [PMID: 39634339 PMCID: PMC11616527 DOI: 10.1016/j.bbrep.2024.101876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
Incorporation of blood capillaries in engineered tissues and their functional connection to host blood vessels is essential for success in engineering vascularized tissues, a process which involves spatial patterning of endothelial cells (ECs) to form functional and integrated vascular networks. Different types of ECs have been employed for vascular network formation and each source offers advantages and disadvantages. While ECs derived from induced pluripotent stem cells (iPSC-ECs) offer advantages over primary ECs in that they can be generated in large quantities for autologous applications, their suitability for disease modelling and cell replacement therapies is not well-explored. The present study compares the angiogenic capacity of iPSC-ECs and primary ECs (cardiac microvascular ECs and lymphatic microvascular ECs) using an in vitro tubulogenesis assay, revealing comparable performance in forming a pseudo-capillary network on Matrigel. Analysis of genes encoding angiogenic factors (VEGFA, VEGFC, VEGFD and ANG), endothelial cell markers (PECAM1, PCDH12 and NOS3) and proliferation markers (AURKB and MKI67) indicates a significant positive correlation between NOS3 mRNA expression levels and various tubulogenic parameters. Further experimentation using a CRISPR activation system demonstrates a positive impact of NOS3 on tubulogenic activity of ECs, suggesting that iPSC-ECs can be enhanced with endogenous NOS3 activation. Collectively, these findings highlight the potential of iPSC-ECs in generating vascularized tissues and advancing therapeutic vascularization.
Collapse
Affiliation(s)
- Anne M. Kong
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
| | - Zulhusni A. Idris
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- School of Engineering, University of Melbourne, VIC, Australia
| | - Daniel Urrutia-Cabrera
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Jarmon G. Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
| | - Ren Jie Phang
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
| | - Geraldine M. Mitchell
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Faculty of Health Sciences, Australian Catholic University, VIC, Australia
| | - Raymond C.B. Wong
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Shiang Y. Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Victoria, Monash University, Australia
- National Heart Research Institute Singapore, National Heart Centre, Singapore
| |
Collapse
|
13
|
Rabadi MM, Verde MR, Camilliere M, Vecchio N, Kandhi S, Sekulic M, Wolin MS, Ratliff BB. Renal and Vascular Functional Decline in Aged Low Birth Weight Murine Adults. Kidney Blood Press Res 2024; 49:1075-1090. [PMID: 39571568 DOI: 10.1159/000542141] [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: 05/14/2024] [Accepted: 10/16/2024] [Indexed: 12/19/2024] Open
Abstract
INTRODUCTION Maternal undernutrition (MUN)-induced low birth weight (LBW) neonates are susceptible to the development of high blood pressure and kidney disease later in life, although the underlying pathological causes remain unclear. The study here investigated the role of renal oxidative stress, impairment of vascular function, and altered sensitivity to angiotensin II (Ang II) as factors that contribute to these pathologies in aged LBW mice. METHODS LBW offspring were generated using a combined protein and caloric restricted MUN mouse model. The resulting LBW offspring were examined 1 year after birth for mean arterial blood pressure (MABP) (carotid artery catheterization), renal blood flow (RBF) (laser Doppler flowmetry), glomerular filtration rate (GFR) (sinistrin clearance), vasoreactivity (myograph), renal vascular density (CD31 staining), and reactive oxygen species (ROS) (ROS probes). Immunoblotting examined Ang II type 1 receptor (AT1R), soluble guanylate cyclase (sGC), and antioxidant systems. Pharmacological agents delivered to animals included the sGC stimulator δ-aminolevulinic acid (ALA), the AT1R inhibitor losartan, the antioxidant ethyl pyruvate (EP), and the toll-like receptor 4 inhibitor TAK242. RESULTS After 1 year, MABP was increased, while RBF, GFR, vascular reactivity, renal vascular density, and sGC were all reduced in the LBW aged adult. All four pharmacological agents improved MABP, RBF, GFR, vascular density, and vascular reactivity. Renal ROS was increased in the LBW adult but was reduced by ALA, EP, and TAK242 treatment. AT1R was upregulated in the LBW adult, while sGC was decreased, an effect reversed by ALA treatment. Endogenous antioxidant systems, including SOD1, catalase, and glutathione were downregulated in the LBW adult. CONCLUSION MUN-induced LBW mice experience increased Ang II sensitivity and oxidative stress. The increased Ang II sensitivity and ROS generation influences vascular density and reactivity, which drive an increase in MABP, and a concomitantly decrease in RBF and glomerular filtration. Pharmacological intervention that inhibits AT1R, enhances levels of sGC, reduces ROS, or inhibits toll-like receptor 4 improves vascular and renal function in the LBW adult.
Collapse
Affiliation(s)
- May M Rabadi
- Department of Medicine, New York Medical College, Valhalla, New York, USA
| | - Marella R Verde
- Department of Physiology, New York Medical College, Valhalla, New York, USA
| | - Mia Camilliere
- Department of Pathology, New York Medical College, Valhalla, New York, USA
| | - Nicholas Vecchio
- Department of Medicine, New York Medical College, Valhalla, New York, USA
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, New York, USA
| | - Miroslav Sekulic
- Department of Pathology and Cell Biology, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, New York, USA
| | - Brian B Ratliff
- Department of Medicine, New York Medical College, Valhalla, New York, USA
- Department of Physiology, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
14
|
Alhusban S, Nofal M, Kovacs-Kasa A, Kress TC, Koseoglu MM, Zaied AA, Belin de Chantemele EJ, Annex BH. Glucosamine-Mediated Hexosamine Biosynthesis Pathway Activation Uses ATF4 to Promote "Exercise-Like" Angiogenesis and Perfusion Recovery in PAD. Circulation 2024; 150:1702-1719. [PMID: 39253813 PMCID: PMC11955094 DOI: 10.1161/circulationaha.124.069580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Endothelial cells (ECs) use glycolysis to produce energy. In preclinical models of peripheral arterial disease, further activation of EC glycolysis was ineffective or deleterious in promoting hypoxia-dependent angiogenesis, whereas pentose phosphate pathway activation was effective. Hexosamine biosynthesis pathway, pentose phosphate pathway, and glycolysis are closely linked. Glucosamine directly activates hexosamine biosynthesis pathway. METHODS Hind-limb ischemia in endothelial nitric oxide synthase knockout (eNOS-/-) and BALB/c mice was used. Glucosamine (600 μg/g per day) was injected intraperitoneally. Blood flow recovery was assessed using laser Doppler perfusion imaging and angiogenesis was studied by CD31 immunostaining. In vitro, human umbilical vein ECs and mouse microvascular ECs with glucosamine, L-glucose, or vascular endothelial growth factor (VEGF165a) were tested under hypoxia and serum starvation. Cell Counting Kit-8, tube formation, intracellular reactive oxygen species, electric cell-substrate impedance sensing, and fluorescein isothiocyanate dextran permeability were assessed. Glycolysis and oxidative phosphorylation were assessed by seahorse assay. Gene expression was assessed using RNA sequencing, real-time quantitative polymerase chain reaction, and Western blot. Human muscle biopsies from patients with peripheral arterial disease were assessed for EC O-GlcNAcylation before and after supervised exercise versus standard medical care. RESULTS On day 3 after hind-limb ischemia, glucosamine-treated versus control eNOS-/- mice had less necrosis (n=4 or 5 per group). Beginning on day 7 after hind-limb ischemia, glucosamine-treated versus control BALB/c mice had higher blood flow, which persisted to day 21, when ischemic muscles showed greater CD31 staining per muscle fiber (n=8 per group). In vitro, glucosamine versus L-glucose ECs showed improved survival (n=6 per group) and tube formation (n=6 per group). RNA sequencing of glucosamine versus L-glucose ECs showed increased amino acid metabolism (n=3 per group). That resulted in increased oxidative phosphorylation (n=8-12 per group) and serine biosynthesis pathway without an increase in glycolysis or pentose phosphate pathway genes (n=6 per group). This was associated with better barrier function (n=6-8 per group) and less reactive oxygen species (n=7 or 8 per group) compared with activating glycolysis by VEGF165a. These effects were mediated by activating transcription factor 4, a driver of exercise-induced angiogenesis. In muscle biopsies from humans with peripheral arterial disease, EC/O-GlcNAcylation was increased by 12 weeks of supervised exercise versus standard medical care (n=6 per group). CONCLUSIONS In cells, mice, and humans, activation of hexosamine biosynthesis pathway by glucosamine in peripheral arterial disease induces an "exercise-like" angiogenesis and offers a promising novel therapeutic pathway to treat this challenging disorder.
Collapse
Affiliation(s)
- Suhib Alhusban
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Mohamed Nofal
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Anita Kovacs-Kasa
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Taylor C Kress
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - M Murat Koseoglu
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Abdelrahman A Zaied
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
- Department of Medicine (A.A.Z., B.H.A.), Medical College of Georgia at Augusta University
| | - Eric J Belin de Chantemele
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Brian H Annex
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
- Department of Medicine (A.A.Z., B.H.A.), Medical College of Georgia at Augusta University
| |
Collapse
|
15
|
Eltanahy AM, Aupetit A, Buhr ED, Van Gelder RN, Gonzales AL. Light-sensitive Ca 2+ signaling in the mammalian choroid. Proc Natl Acad Sci U S A 2024; 121:e2418429121. [PMID: 39514305 PMCID: PMC11573543 DOI: 10.1073/pnas.2418429121] [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: 09/12/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
The choroid is the thin, vasculature-filled layer of the eye situated between the sclera and the retina, where it serves the metabolic needs of the light-sensing photoreceptors in the retina. Illumination of the interior surface of the back of the eye (fundus) is a critical regulator of subretinal fluid homeostasis, which determines the overall shape of the eye, but it is also important for choroidal perfusion. Noted for having some of the highest blood flow rates in the body, the choroidal vasculature has been reported to lack intrinsic, intravascular pressure-induced (myogenic) autoregulatory mechanisms. Here, we ask how light directly regulates choroid perfusion and ocular fluid homeostasis, testing the hypothesis that light facilitates ocular fluid absorption by directly increasing choroid endothelial permeability and decreasing choroid perfusion. Utilizing ex vivo pressurized whole-choroid and whole-eye preparations from mice expressing cell-specific Ca2+ indicators, we found that the choroidal vasculature has two intrinsically light-sensitive Ca2+-signaling mechanisms: One increases Ca2+-dependent production of nitric oxide in choroidal endothelial cells; the other promotes vasoconstriction through Ca2+ elevation in vascular smooth muscle cells. In addition, we found that choroidal flow, or pressure, modulates endothelial and smooth muscle photosensitivity and trans-retinal absorption of fluid into the choroid. These results collectively suggest that the choroid vasculature exhibits an inverted form of autoregulatory control, where pressure- and light-induced mechanisms work in opposition to regulate blood flow and maintain fluid balance in response to changes in light and dark, aligning with the metabolic needs of photoreceptors.
Collapse
Affiliation(s)
- Ahmed M Eltanahy
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318
| | - Alex Aupetit
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318
| | - Ethan D Buhr
- Department of Ophthalmology, University of Washington, Seattle, WA 98104
- Roger and Angie Karalis Retina Center, Department of Ophthalmology, University of Washington, Seattle, WA 98104
| | - Russell N Van Gelder
- Department of Ophthalmology, University of Washington, Seattle, WA 98104
- Roger and Angie Karalis Retina Center, Department of Ophthalmology, University of Washington, Seattle, WA 98104
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA 98104
| | - Albert L Gonzales
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318
| |
Collapse
|
16
|
Wesson T, Zhang L, Morrison RA, Brookes S, Calcagno H, Finnegan P, Voytik-Harbin S, Halum S. Tissue-Engineered Implant for Hemilaryngectomy Reconstruction with Recurrent Laryngeal Nerve Injury. Laryngoscope 2024; 134:4604-4613. [PMID: 38989732 PMCID: PMC11529076 DOI: 10.1002/lary.31616] [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: 03/18/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE Laryngeal cancer resections often require excision of portions of the larynx along with sacrifice of the ipsilateral recurrent laryngeal nerve (RLN). In such cases, there are no reconstructive options that reliably restore laryngeal function, rendering patients with severe functional impairment. To address this unmet clinical need, we extend our evaluation of a 3-implant mucosal, muscle, cartilage reconstruction approach aimed at promoting functional laryngeal restoration in a porcine hemilaryngectomy model with ipsilateral RLN transection. METHODS Six Yucatan mini-pigs underwent full-thickness hemilaryngectomies with RLN transection followed by transmural reconstruction using fabricated collagen polymeric mucosal, muscle, and cartilage replacements. To determine the effect of adding therapeutic cell populations, subsets of animals received collagen muscle implants containing motor-endplate-expressing muscle progenitor cells (MEEs) and/or collagen cartilage implants containing adipose stem cell (ASC)-derived chondrocyte-like cells. Acoustic vocalization and laryngeal electromyography (L-EMG) provided functional assessments and histopathological analysis with immunostaining was used to characterize the tissue response. RESULTS Five of six animals survived the 4-week postoperative period with weight gain, airway maintenance, and audible phonation. No tracheostomy or feeding tube was required. Gross and histological assessments of all animals revealed implant integration and regenerative remodeling of airway mucosa epithelium, muscle, and cartilage in the absence of a material-mediated foreign body reaction or biodegradation. Early voice and L-EMG data were suggestive of positive functional outcomes. CONCLUSION Laryngeal reconstruction with collagen polymeric mucosa, muscle, and cartilage replacements may provide effective restoration of function after hemilaryngectomy with RLN transection. Future preclinical studies should focus on long-term functional outcomes. LEVEL OF EVIDENCE NA Laryngoscope, 134:4604-4613, 2024.
Collapse
Affiliation(s)
- Troy Wesson
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine
| | - Lujuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine
| | | | - Sarah Brookes
- Weldon School of Biomedical Engineering, Purdue University
| | - Haley Calcagno
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine
| | - Patrick Finnegan
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine
| | | | - Stacey Halum
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine
- Department of Speech, Language, and Hearing Sciences, Purdue University
| |
Collapse
|
17
|
Wazan LE, Widhibrata A, Liu GS. Soluble FLT-1 in angiogenesis: pathophysiological roles and therapeutic implications. Angiogenesis 2024; 27:641-661. [PMID: 39207600 DOI: 10.1007/s10456-024-09942-8] [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: 06/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Fine-tuning angiogenesis, the development of new blood vessels, is essential for maintaining a healthy circulatory and lymphatic system. The small glycoprotein vascular endothelial growth factors (VEGF) are the key mediators in this process, binding to their corresponding membrane-bound VEGF receptors (VEGFRs) to activate angiogenesis signaling pathways. These pathways are crucial throughout human life as they are involved in lymphatic and vascular endothelial cell permeability, migration, proliferation, and survival. Neovascularization, the formation of abnormal blood vessels, occurs when there is a dysregulation of angiogenesis and can result in debilitating disease. Hence, VEGFRs have been widely studied to understand their role in disease-causing angiogenesis. VEGFR1, also known as Fms-like tyrosine kinase-1 (FLT-1), is also found in a soluble form, soluble FLT-1 or sFLT-1, which is known to act as a VEGF neutralizer. It is incorporated into anti-VEGF therapy, designed to treat diseases caused by neovascularization. Here we review the journey of sFLT-1 discovery and delve into the alternative splicing mechanism that creates the soluble receptor, its prevalence in disease states, and its use in current and future potential therapies.
Collapse
Affiliation(s)
- Layal Ei Wazan
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Level 7, 32 Gisborne Street, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia
| | - Ariel Widhibrata
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Level 7, 32 Gisborne Street, East Melbourne, VIC, 3002, Australia
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Level 7, 32 Gisborne Street, East Melbourne, VIC, 3002, Australia.
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| |
Collapse
|
18
|
Shabanian K, Shabanian T, Karsai G, Pontiggia L, Paneni F, Ruschitzka F, Beer JH, Saeedi Saravi SS. AQP1 differentially orchestrates endothelial cell senescence. Redox Biol 2024; 76:103317. [PMID: 39180980 PMCID: PMC11388013 DOI: 10.1016/j.redox.2024.103317] [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: 03/18/2024] [Revised: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Accumulation of senescent endothelial cells (ECs) with age is a pivotal driver of cardiovascular diseases in aging. However, little is known about the mechanisms and signaling pathways that regulate EC senescence. In this report, we delineate a previously unrecognized role of aquaporin 1 (AQP1) in orchestrating extracellular hydrogen peroxide (H2O2)-induced cellular senescence in aortic ECs. Our findings underscore AQP1's differential impact on senescence hallmarks, including cell-cycle arrest, senescence-associated secretory phenotype (SASP), and DNA damage responses, intricately regulating angiogenesis. In proliferating ECs, AQP1 is crucial for maintaining angiogenic capacity, whereas disruption of AQP1 induces morphological and mitochondrial alterations, culminating in senescence and impaired angiogenesis. Conversely, Aqp1 knockdown or selective blockade of AQP1 in senescent ECs rescues the excess H2O2-induced cellular senescence phenotype and metabolic dysfunction, thereby ameliorating intrinsic angiogenic incompetence. Mechanistically, AQP1 facilitates H2O2 transmembrane transport, exacerbating oxidant-sensitive kinases CaMKII-AMPK. This process suppresses HDAC4 translocation, consequently de-repressing Mef2A-eNOS signaling in proliferating ECs. However, in senescent ECs, AQP1 overexpression is linked to preserved HDAC4-Mef2A complex and downregulation of eNOS signaling. Together, our studies identify AQP1 as a novel epigenetic regulator of HDAC4-Mef2A-dependent EC senescence and angiogenic potential, highlighting its potential as a therapeutic target for antagonizing age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Khatereh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Taraneh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich, 8952, Schlieren, Switzerland
| | - Luca Pontiggia
- Tissue Biology Research Unit, University Children's Hospital Zurich, 8952, Schlieren, Switzerland; Children's Research Center, University Children's Hospital Zurich, 8032, Zurich, Switzerland; Faculty of Medicine, University of Zurich, 8032, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404, Baden, Switzerland.
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Liu S, Zhang Y, Ma X, Zhan C, Ding N, Shi M, Zhang W, Yang S. Protective effects of engineered Lactobacillus crispatus strains expressing G-CSF on thin endometrium of mice. Hum Reprod 2024; 39:2305-2319. [PMID: 39178354 DOI: 10.1093/humrep/deae190] [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: 11/18/2023] [Revised: 06/26/2024] [Indexed: 08/25/2024] Open
Abstract
STUDY QUESTION Does recombinant Lactobacillus expressing granulocyte colony-stimulating factor (G-CSF) have a better protective effect than the current treatment of thin endometrium (TE)? SUMMARY ANSWER This study suggested that the intrauterine injection of Lactobacillus crispastus (L. crispastus)-pPG612-G-CSF has a positive effect on preventing TE induced by 95% alcohol in mice. WHAT IS KNOWN ALREADY TE has a negative impact on the success rate of ART in patients, and is usually caused by intrauterine surgery, endometrial infection, or hormone drugs. Exogenous G-CSF can promote endometrial vascular remodelling and increase endometrial receptivity and the embryo implantation rate. Moreover, Lactobacillus plays a crucial role in maintaining and regulating the local microecological balance of the reproductive tract, and it could be a delivery carrier of the endometrial repair drug G-CSF. STUDY DESIGN, SIZE, DURATION We constructed engineered L. crispastus strains expressing G-CSF. The mice were divided into five groups: (i) Control group (C, n = 28), uteri were treated with preheated saline solution via intrauterine injection on the third and sixth day of oestrus; (ii) Model group (M, n = 35), where uteri were treated with 95% alcohol on the third day of oestrus and preheated saline solution on the sixth day of oestrus via intrauterine injection; (iii) L. crispatus-pPG612-treatment group (L, n = 45), where uteri were treated with 95% alcohol on the third day of oestrus and 0.1 ml × 108 CFU/ml L. crispatus-pPG612 on the sixth day of oestrus via intrauterine injection; (iv) L. crispatus-pPG612-treatment group (LG, n = 45), where uteri were treated with 95% alcohol on the third day of oestrus and 0.1 ml × 108 CFU/ml L. crispatus-pPG612-G-CSF on the sixth day of oestrus via intrauterine injection; (v) G-CSF-treatment group (G, n = 52), where uteri were treated with 95% alcohol on the third day of oestrus and 30 µg/kg G-CSF on the sixth day of oestrus via intrauterine injection. Then, we compared the effects of L. crispastus, L. crispatus-pPG612-G-CSF and G-CSF on endometrial thickness, angiogenesis, fibrosis, and inflammation in the TE mouse. PARTICIPANTS/MATERIALS, SETTING, METHODS We collected uterine tissues for haematoxylin-eosin staining, immunohistochemical staining, Western blot and RT-PCR, as well as serum for ELISA and uterine flushing solution for high-throughput sequencing. MAIN RESULTS AND THE ROLE OF CHANCE Compared with those in the M group (the mice of the group were intrauterine injected 95% alcohol and treated with saline solution), the L. crispatus-pPG612-G-CSF strain increased the thickness of the endometrium (P < 0.001) and the number of blood vessels and glands (both P < 0.001), enhanced the expression of cytokeratin 19 (CK19) (P < 0.001), vimentin (Vim) (P < 0.001), vascular endothelial growth factor-A (P < 0.001), and CD34 (P < 0.001), and decreased fibrosis levels (P = 0.004). In addition, the high-throughput sequencing results indicated that the L. crispatus-pPG612-G-CSF strain could decrease the abundance of Pseudomonas (P = 0.044) and Actinomyces spp. (P = 0.094) in TE mice and increased the average number of embryos (P = 0.036). Finally, the L. crispatus-pPG612-G-CSF strain was preliminarily confirmed to activate the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signalling pathway and enhance the mRNA expression of hypoxia-inducible factor-1α (P < 0.001), vascular endometrial growth factor (P = 0.003), and endothelial cell nitric oxide synthase (P = 0.003) in mouse uterine tissue. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Therapy with the L. crispatus-pPG612-G-CSF strain has tremendous potential to accelerate the reparative processes of TE. However, we have reported only the expression of genes and proteins related to the PI3K/AKT pathway, and numerous other mechanisms may also be involved in the restoration of the endometrium by L. crispatus-pPG612-G-CSF. WIDER IMPLICATIONS OF THE FINDINGS The results from the study provide new ideas and suggest new methods for TE treatment. STUDY FUNDING/COMPETING INTEREST(S) This work was financially supported by the Project of Science and Technology Development Plan of Jilin Province (grant number 20210101232JC), the Science and Technology Plan Item of Jilin Provincial Education Department (grant number JT53101022010), and the Doctoral Research Start-up Fund of Jilin Medical University (grant numbers JYBS2021014LK and 2022JYBS006KJ). The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.
Collapse
Affiliation(s)
- Shuang Liu
- Reproductive Immunology Laboratory, Basic Medical College, Jilin Medical University, Jilin, China
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yingnan Zhang
- Chronic Disease Laboratory, School of Public Health, Jilin Medical University, Jilin, China
- Department of Biology, College of Life Science, Changchun Sci-Tech University, Changchun, China
| | - Xin Ma
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chenglin Zhan
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Ning Ding
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Mai Shi
- Chronic Disease Laboratory, School of Public Health, Jilin Medical University, Jilin, China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jilin Medical University, Jilin, China
| | - Shubao Yang
- Reproductive Immunology Laboratory, Basic Medical College, Jilin Medical University, Jilin, China
| |
Collapse
|
20
|
Faustino TG, da Rosa Filho RR, Francischini MCP, Brito MM, Angrimani DSR, Vannucchi CI. In Situ Uterine Artery Prostaglandin E 2 and Nitric Oxide in Open-Cervix Pyometra and Medically Treated Bitches. J Vet Pharmacol Ther 2024. [PMID: 39287059 DOI: 10.1111/jvp.13482] [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: 02/13/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
Uterine vascular alterations take place in pyometra bitches speculatively influenced by prostaglandin and nitric oxide pathways. However, no causative effect of nitric oxide on endometrial vascularization was proved elsewhere for medically treated pyometra bitches. This study aimed to identify the main in situ uterine artery vasodilation pathway in pyometra bitches medically treated with antigestagen solely or coupled with prostaglandin. Pyometra bitches were enrolled into groups: Ovariohysterectomy at diagnosis (Control-OHE; n = 7), Antigestagen (10 mg/kg aglepristone on Days 1, 2, and 8 after diagnosis; n = 5), and Antigestagen + luteolytic (aglepristone plus 1 μg/kg of cloprostenol from Days 1-7; n = 5). Treated bitches were ovariohysterectomized after 8 days of treatment. Uterine artery fragments from all bitches were collected for tissue nitric oxide and prostaglandin E2 assays. Control-OHE group had lower uterine artery concentration of nitric oxide compared to treated bitches (Antigestagen and Antigestagen + luteolytic groups). No significant difference was verified between the medical treated groups. Uterine artery concentration of prostaglandin E2 was not different between control and treated bitches, as well as between both treated groups. In conclusion, nitric oxide and prostaglandin E2 are not directly involved in vascular modulation of the uterine artery, albeit pyometra medical treatment influences nitric oxide concentration in the uterine artery.
Collapse
Affiliation(s)
- Thaís Gomes Faustino
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Roberto Rodrigues da Rosa Filho
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Maíra Morales Brito
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Daniel Souza Ramos Angrimani
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Camila Infantosi Vannucchi
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Saw EL, Fronius M, Katare R, Kakinuma Y. Mini Review: the non-neuronal cardiac cholinergic system in type-2 diabetes mellitus. Front Cardiovasc Med 2024; 11:1425534. [PMID: 39314774 PMCID: PMC11417620 DOI: 10.3389/fcvm.2024.1425534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Diabetic heart disease remains the leading cause of death in individuals with type-2 diabetes mellitus (T2DM). Both insulin resistance and metabolic derangement, hallmark features of T2DM, develop early and progressively impair cardiovascular function. These factors result in altered cardiac metabolism and energetics, as well as coronary vascular dysfunction, among other consequences. Therefore, gaining a deeper understanding of the mechanisms underlying the pathophysiology of diabetic heart disease is crucial for developing novel therapies for T2DM-associated cardiovascular disease. Cardiomyocytes are equipped with the cholinergic machinery, known as the non-neuronal cardiac cholinergic system (NNCCS), for synthesizing and secreting acetylcholine (ACh) as well as possessing muscarinic ACh receptor for ACh binding and initiating signaling cascade. ACh from cardiomyocytes regulates glucose metabolism and energetics, endothelial function, and among others, in an auto/paracrine manner. Presently, there is only one preclinical animal model - diabetic db/db mice with cardiac-specific overexpression of choline transferase (Chat) gene - to study the effect of activated NNCCS in the diabetic heart. In this mini-review, we discuss the physiological role of NNCCS, the connection between NNCCS activation and cardiovascular function in T2DM and summarize the current knowledge of S-Nitroso-NPivaloyl-D-Penicillamine (SNPiP), a novel inducer of NNCCS, as a potential therapeutic strategy to modulate NNCCS activity for diabetic heart disease.
Collapse
Affiliation(s)
- Eng Leng Saw
- Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Martin Fronius
- Department of Physiology, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Yoshihiko Kakinuma
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
22
|
Qiu X, Lan X, Li L, Chen H, Zhang N, Zheng X, Xie X. The role of perirenal adipose tissue deposition in chronic kidney disease progression: Mechanisms and therapeutic implications. Life Sci 2024; 352:122866. [PMID: 38936605 DOI: 10.1016/j.lfs.2024.122866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.
Collapse
Affiliation(s)
- Xiang Qiu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xin Lan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Langhui Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Ningjuan Zhang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoli Zheng
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
23
|
Lim SJ, Gan SC, Ong HT, Ngeow YF. In vitro analysis of VEGF-mediated endothelial permeability and the potential therapeutic role of Anti-VEGF in severe dengue. Biochem Biophys Rep 2024; 39:101814. [PMID: 39263317 PMCID: PMC11387214 DOI: 10.1016/j.bbrep.2024.101814] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Background Vascular endothelial growth factor (VEGF) is one of the proteins involved in dengue immunopathogenesis. It is overexpressed in severe dengue and contributes to vascular permeability and plasma leakage. In this study, we investigated the effects of VEGF and anti-VEGF treatments on endothelial cells in vitro, to assess the potential use of anti-VEGF antibodies in managing severe dengue. Methods Human pulmonary microvascular endothelial cells were treated with VEGF and a VEGF/anti-VEGF combination. The effects of the treatments were studied using an endothelial permeability assay and microarray gene expression profiling. In the permeability assay, the fluorescein isothiocyanate (FITC)-dextran fluorescence signal across the endothelial monolayer was recorded, and the cells were stained with PECAM-1 to detect gap formation. RNA was extracted from treated cells for microarray gene profiling and analysis. The results were analyzed for differentially expressed genes (DEGs) and gene enrichment analysis. The DEGs were subjected to STRING to construct the protein-protein interaction network and then Cytoscape to identify the hub genes. Results VEGF-treated endothelial cells showed greater movement of FITC-dextran across the monolayer than VEGF/anti-VEGF-treated cells. There were 111 DEGs for VEGF-treated cells and 118 DEGs for VEGF/anti-VEGF-treated cells. The genes upregulated in VEGF-treated cells were enriched in inflammatory responses and regulation of the endothelial barrier, nitric oxide synthesis, angiogenesis, and the nucleotide-binding oligomerization domain-like receptor signaling pathway. Top 10 hub genes were identified from the DEGs. Conclusions VEGF treatment increased permeability across endothelial cells, while anti-VEGF reduced this leakage. Analysis of VEGF-treated endothelial cells identified hub genes implicated in severe dengue. The top 10 hub genes were TNF, IL1B, IL6, CCL2, PTGS2, ICAM1, CXCL2, CXCL1, CSF2, and TLR2. The results of this study show that using anti-VEGF antibodies to neutralize VEGF may be a promising therapy to prevent the progression of dengue to severe dengue.
Collapse
Affiliation(s)
- Sheng Jye Lim
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| | - Seng Chiew Gan
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| | - Hooi Tin Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
- Center for Cancer Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| | - Yun Fong Ngeow
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
- Centre for Research on Communicable Diseases, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
24
|
Tsuji-Tamura K, Sato M, Tamura M. Pharmacological control of angiogenesis by regulating phosphorylation of myosin light chain 2. Cell Signal 2024; 120:111223. [PMID: 38729320 DOI: 10.1016/j.cellsig.2024.111223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Control of angiogenesis is widely considered a therapeutic strategy, but reliable control methods are still under development. Phosphorylation of myosin light chain 2 (MLC2), which regulates actin-myosin interaction, is critical to the behavior of vascular endothelial cells (ECs) during angiogenesis. MLC2 is phosphorylated by MLC kinase (MLCK) and dephosphorylated by MLC phosphatase (MLCP) containing a catalytic subunit PP1. We investigated the potential role of MLC2 in the pharmacological control of angiogenesis. METHODS AND RESULTS We exposed transgenic zebrafish Tg(fli1a:Myr-mCherry)ncv1 embryos to chemical inhibitors and observed vascular development. PP1 inhibition by tautomycetin increased length of intersegmental vessels (ISVs), whereas MLCK inhibition by ML7 decreased it; these effects were not accompanied by structural dysplasia. ROCK inhibition by Y-27632 also decreased vessel length. An in vitro angiogenesis model of human umbilical vein endothelial cells (HUVECs) showed that tautomycetin increased vascular cord formation, whereas ML7 and Y-27632 decreased it. These effects appear to be influenced by regulation of cell morphology rather than cell viability or motility. Actin co-localized with phosphorylated MLC2 (pMLC2) was abundant in vascular-like elongated-shaped ECs, but poor in non-elongated ECs. pMLC2 was associated with tightly arranged actin, but not with loosely arranged actin. Moreover, knockdown of MYL9 gene encoding MLC2 reduced total MLC2 and pMLC2 protein and inhibited angiogenesis in HUVECs. CONCLUSION The present study found that MLC2 is a pivotal regulator of angiogenesis. MLC2 phosphorylation may be involved in the regulation of of cell morphogenesis and cell elongation. The functionally opposite inhibitors positively or negatively control angiogenesis, probably through the regulating EC morphology. These findings may provide a unique therapeutic target for angiogenesis.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan.
| | - Mari Sato
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan
| | - Masato Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan
| |
Collapse
|
25
|
Dussault S, Desjarlais M, Raguema N, Boilard E, Chemtob S, Rivard A. Selective Enrichment of Angiomirs in Extracellular Vesicles Released from Ischemic Skeletal Muscles: Potential Role in Angiogenesis and Neovascularization. Cells 2024; 13:1243. [PMID: 39120274 PMCID: PMC11312235 DOI: 10.3390/cells13151243] [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: 05/24/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
MicroRNAs (miRs) regulate physiological and pathological processes, including ischemia-induced angiogenesis and neovascularization. They can be transferred between cells by extracellular vesicles (EVs). However, the specific miRs that are packaged in EVs released from skeletal muscles, and how this process is modulated by ischemia, remain to be determined. We used a mouse model of hindlimb ischemia and next generation sequencing (NGS) to perform a complete profiling of miR expression and determine the effect of ischemia in skeletal muscles, and in EVs of different sizes (microvesicles (MVs) and exosomes) released from these muscles. Ischemia significantly modulated miR expression in whole muscles and EVs, increasing the levels of several miRs that can have pro-angiogenic effects (angiomiRs). We found that specific angiomiRs are selectively enriched in MVs and/or exosomes in response to ischemia. In silico approaches indicate that these miRs modulate pathways that play key roles in angiogenesis and neovascularization, including HIF1/VEGF signaling, regulation of actin cytoskeleton and focal adhesion, NOTCH, PI3K/AKT, RAS/MAPK, JAK/STAT, TGFb/SMAD signaling and the NO/cGMP/PKG pathway. Thus, we show for the first time that angiomiRs are selectively enriched in MVs and exosomes released from ischemic muscles. These angiomiRs could be targeted in order to improve the angiogenic function of EVs for potential novel therapeutic applications in patients with severe ischemic vascular diseases.
Collapse
Affiliation(s)
- Sylvie Dussault
- Department of Medicine, Centre Hospitalier de l’Université de Montréal (CHUM) Research Center, Montréal, QC H2X 0A9, Canada; (S.D.); (N.R.)
| | - Michel Desjarlais
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada; (M.D.); (S.C.)
| | - Nozha Raguema
- Department of Medicine, Centre Hospitalier de l’Université de Montréal (CHUM) Research Center, Montréal, QC H2X 0A9, Canada; (S.D.); (N.R.)
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec City, QC G1V 0A6, Canada;
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada; (M.D.); (S.C.)
| | - Alain Rivard
- Department of Medicine, Centre Hospitalier de l’Université de Montréal (CHUM) Research Center, Montréal, QC H2X 0A9, Canada; (S.D.); (N.R.)
| |
Collapse
|
26
|
Peng KW, Chang ML, Chien RN, Chen YC, Tian YC, Peng YS, Huang HC, Fang JT, Lee FY, Yang CW, Tsai MH. Pulmonary Vascular Permeability and Extravascular Lung Water Index in Patients with Liver Cirrhosis and Septic Shock. J Clin Med 2024; 13:3796. [PMID: 38999366 PMCID: PMC11242845 DOI: 10.3390/jcm13133796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/08/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Backgrounds and Aims: Patients with cirrhosis are susceptible to sepsis and septic shock. Cirrhotic patients also have increased capillary permeability and are prone to developing volume overload. Patients with septic shock may have an enhanced pulmonary vascular permeability index (PVPI) and extravascular lung water index (EVLWI), both of which are associated with an unfavorable prognosis. It is plausible that pre-existing hyperpermeability may deteriorate when cirrhotic patients develop septic shock. However, it remains unknown whether PVPI and EVLWI can predict the prognosis of cirrhotic patients with septic shock. Pulse Indicator Continuous Cardiac Output (PiCCO) is an established tool to measure PVPI and EVLWI. Therefore, we conducted this retrospective study to investigate the prognostic significance of PVPI and EVLWI in cirrhotic patients with septic shock using PiCCO monitoring. Methods: We included 83 patients with liver cirrhosis and septic shock. EVLW indexed to actual body weight (aEVLWI), EVLW indexed to predicted body weight (pEVLWI), PVPI, disease severity scores, and other biomarkers were analyzed. We collected the PiCCO data on the first 2 days. Results: The overall 28-day mortality was 43.4%. The values of PVPI, aEVLWI, and pEVLWI on day 2 (PVPID2, aEVLWID2, EVLWID2) were significantly higher in non-survivors. The discriminating power of PVPID2 and EVLWID2 to predict 28-day mortality was tested using the area under a ROC curve. The areas under ROC curves (mean ± SEM) were 0.713 ± 0.061 and 0.650 ± 0.063 for PVPID2 and pEVLWID2. In the multivariate analysis, PVPID2, bilirubin, and lactate were independent factors which predicted 28-day mortality. Conclusions: Higher levels of PVPID2 and pEVLWID2 are associated with higher 28-day mortality rates in cirrhotic patients with septic shock. PVPI and pEVLWI may be useful to guide fluid management in this clinical setting.
Collapse
Affiliation(s)
- Kang-Wei Peng
- Division of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City 333, Taiwan; (K.-W.P.); (M.-L.C.)
| | - Ming-Ling Chang
- Division of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City 333, Taiwan; (K.-W.P.); (M.-L.C.)
| | - Rong-Nan Chien
- Division of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City 333, Taiwan; (K.-W.P.); (M.-L.C.)
| | - Yung-Chang Chen
- Division of Critical Care Nephrology, Kidney Institute, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.C.); (Y.-C.T.); (C.-W.Y.)
| | - Ya-Chung Tian
- Division of Critical Care Nephrology, Kidney Institute, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.C.); (Y.-C.T.); (C.-W.Y.)
| | - Yun-Shing Peng
- Department of Internal Medicine, Chang Gung Memorial Hospital, Chia-Yi 613, Taiwan
| | - Hui-Chun Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veteran General Hospital, Faculty of Medicine, Yang-Ming University School of Medicine, Taipei 112, Taiwan; (H.-C.H.)
- Division of General Medicine, Department of Medicine, Taipei Veteran General Hospital, Faculty of Medicine, Yang-Ming University School of Medicine, Taipei 112, Taiwan
| | - Ji-Tseng Fang
- Division of Critical Care Nephrology, Kidney Institute, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.C.); (Y.-C.T.); (C.-W.Y.)
| | - Fa-Yauh Lee
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veteran General Hospital, Faculty of Medicine, Yang-Ming University School of Medicine, Taipei 112, Taiwan; (H.-C.H.)
| | - Chih-Wei Yang
- Division of Critical Care Nephrology, Kidney Institute, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.C.); (Y.-C.T.); (C.-W.Y.)
| | - Ming-Hung Tsai
- Division of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City 333, Taiwan; (K.-W.P.); (M.-L.C.)
| |
Collapse
|
27
|
Wakasugi R, Suzuki K, Kaneko-Kawano T. Molecular Mechanisms Regulating Vascular Endothelial Permeability. Int J Mol Sci 2024; 25:6415. [PMID: 38928121 PMCID: PMC11203514 DOI: 10.3390/ijms25126415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Vascular endothelial cells form a monolayer in the vascular lumen and act as a selective barrier to control the permeability between blood and tissues. To maintain homeostasis, the endothelial barrier function must be strictly integrated. During acute inflammation, vascular permeability temporarily increases, allowing intravascular fluid, cells, and other components to permeate tissues. Moreover, it has been suggested that the dysregulation of endothelial cell permeability may cause several diseases, including edema, cancer, and atherosclerosis. Here, we reviewed the molecular mechanisms by which endothelial cells regulate the barrier function and physiological permeability.
Collapse
Affiliation(s)
| | | | - Takako Kaneko-Kawano
- Graduate School of Pharmacy, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (R.W.); (K.S.)
| |
Collapse
|
28
|
Salvarredi L, Oglio RA, Rodriguez C, Navarro D, Perona M, Dagrosa MA, Juvenal GJ, Thomasz L. 2-iodohexadecanal induces autophagy during goiter involution. Prostaglandins Other Lipid Mediat 2024; 172:106819. [PMID: 38346574 DOI: 10.1016/j.prostaglandins.2024.106819] [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: 08/24/2023] [Revised: 11/03/2023] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Iodine plays an important role in thyroid physiology and biochemistry. The thyroid is capable of producing different iodolipids such as 2-iodohexadecanal (2-IHDA). Data from different laboratories have shown that 2-IHDA inhibits several thyroid parameters and it has been postulated as intermediary on the action of iodide function. OBJECTIVE To explore different mechanisms involved during the involution of the hyperplastic thyroid gland of Wistar rats towards normality induced by 2-IHDA. METHODS Goiter was induced by the administration of MMI for 10 days, then the treatment was discontinued and Wistar rats were injected with 2-IHDA or KI. RESULTS During involution, 2-IHDA treatment reduced PCNA expression compared to spontaneous involution. KI treatment caused an increase of Caspase-3 activity and TUNEL-positive cells. In contrast, 2-IHDA failed to alter this value but induced an increase of LC3B expression. KI but not 2-IHDA led to an increase in peroxides levels, catalase and glutathione peroxidase activity. CONCLUSIONS We demonstrated that 2-IHDA, in contrast to iodide, did not lead to an increase in oxidative stress or apoptosis induction, indicating that the involution triggered by 2-IHDA in Wistar rats, is primarily due to the inhibition of cell proliferation and the induction of autophagy.
Collapse
Affiliation(s)
- Leonardo Salvarredi
- Nuclear Medicine School Foundation (FUESMEN), National Commission of Atomic Energy (CNEA), Mendoza, Argentina; Instituto Balseiro, National Comission of Atomic Energy & National University of Cuyo, Mendoza, Argentina
| | - Romina A Oglio
- Department of Radiobiology (CAC), National Commission of Atomic Energy (CNEA), Buenos Aires, Argentina
| | - Carla Rodriguez
- Department of Radiobiology (CAC), National Commission of Atomic Energy (CNEA), Buenos Aires, Argentina
| | | | - Marina Perona
- Department of Radiobiology (CAC), National Commission of Atomic Energy (CNEA), Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), CABA, Argentina
| | - María A Dagrosa
- Department of Radiobiology (CAC), National Commission of Atomic Energy (CNEA), Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), CABA, Argentina
| | - Guillermo J Juvenal
- Department of Radiobiology (CAC), National Commission of Atomic Energy (CNEA), Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), CABA, Argentina
| | - Lisa Thomasz
- Department of Radiobiology (CAC), National Commission of Atomic Energy (CNEA), Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), CABA, Argentina.
| |
Collapse
|
29
|
Xie J, Liu G, Chen R, Wang D, Mai H, Zhong Q, Ning Y, Fu J, Tang Z, Xu Y, Li H, Lei M, Cheng H, Huang Y, Zhang Y. NIR-activated electrospun nanodetonator dressing enhances infected diabetic wound healing with combined photothermal and nitric oxide-based gas therapy. J Nanobiotechnology 2024; 22:232. [PMID: 38720301 PMCID: PMC11546403 DOI: 10.1186/s12951-024-02474-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/09/2024] [Indexed: 11/09/2024] Open
Abstract
Diabetic wounds pose a challenge to healing due to increased bacterial susceptibility and poor vascularization. Effective healing requires simultaneous bacterial and biofilm elimination and angiogenesis stimulation. In this study, we incorporated polyaniline (PANI) and S-Nitrosoglutathione (GSNO) into a polyvinyl alcohol, chitosan, and hydroxypropyltrimethyl ammonium chloride chitosan (PVA/CS/HTCC) matrix, creating a versatile wound dressing membrane through electrospinning. The dressing combines the advantages of photothermal antibacterial therapy and nitric oxide gas therapy, exhibiting enduring and effective bactericidal activity and biofilm disruption against methicillin-sensitive Staphylococcus aureus, methicillin-resistant Staphylococcus aureus, and Escherichia coli. Furthermore, the membrane's PTT effect and NO release exhibit significant synergistic activation, enabling a nanodetonator-like burst release of NO through NIR irradiation to disintegrate biofilms. Importantly, the nanofiber sustained a uniform release of nitric oxide, thereby catalyzing angiogenesis and advancing cellular migration. Ultimately, the employment of this membrane dressing culminated in the efficacious amelioration of diabetic-infected wounds in Sprague-Dawley rats, achieving wound closure within a concise duration of 14 days. Upon applying NIR irradiation to the PVA-CS-HTCC-PANI-GSNO nanofiber membrane, it swiftly eradicates bacteria and biofilm within 5 min, enhancing its inherent antibacterial and anti-biofilm properties through the powerful synergistic action of PTT and NO therapy. It also promotes angiogenesis, exhibits excellent biocompatibility, and is easy to use, highlighting its potential in treating diabetic wounds.
Collapse
Affiliation(s)
- Jiajun Xie
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Guihua Liu
- Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou, 516008, Guangdong, People's Republic of China
| | - Rong Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Ding Wang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Huaming Mai
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Qiang Zhong
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Yanhong Ning
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Jinlang Fu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Zinan Tang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Yixin Xu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Hao Li
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Mingyuan Lei
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Hao Cheng
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yuliang Huang
- Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou, 516008, Guangdong, People's Republic of China.
| | - Yang Zhang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
30
|
Meuten TK, Dean GA, Thamm DH. Review: The PI3K-AKT-mTOR signal transduction pathway in canine cancer. Vet Pathol 2024; 61:339-356. [PMID: 37905509 DOI: 10.1177/03009858231207021] [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: 11/02/2023]
Abstract
Tumors in dogs and humans share many similar molecular and genetic features, incentivizing a better understanding of canine neoplasms not only for the purpose of treating companion animals, but also to facilitate research of spontaneously developing tumors with similar biologic behavior and treatment approaches in an immunologically competent animal model. Multiple tumor types of both species have similar dysregulation of signal transduction through phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB; AKT), and mechanistic target of rapamycin (mTOR), collectively known as the PI3K-AKT-mTOR pathway. This review aims to delineate the pertinent aspects of the PI3K-AKT-mTOR signaling pathway in health and in tumor development. It will then present a synopsis of current understanding of PI3K-AKT-mTOR signaling in important canine cancers and advancements in targeted inhibitors of this pathway.
Collapse
|
31
|
Herrera JL, Komatsu M. Akt3 activation by R-Ras in an endothelial cell enforces quiescence and barrier stability of neighboring endothelial cells via Jagged1. Cell Rep 2024; 43:113837. [PMID: 38402584 PMCID: PMC11056028 DOI: 10.1016/j.celrep.2024.113837] [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: 07/06/2023] [Revised: 12/06/2023] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
Communication between adjacent endothelial cells is important for the homeostasis of blood vessels. We show that quiescent endothelial cells use Jagged1 to instruct neighboring endothelial cells to assume a quiescent phenotype and secure the endothelial barrier. This phenotype enforcement by neighboring cells is operated by R-Ras through activation of Akt3, which results in upregulation of a Notch ligand Jagged1 and consequential upregulation of Notch target genes, such as UNC5B, and VE-cadherin accumulation in the neighboring cells. These signaling events lead to the stable interaction between neighboring endothelial cells to continue to fortify juxtacrine signaling via Jagged1-Notch. This mode of intercellular signaling provides a positive feedback regulation of endothelial cell-cell interactions and cellular quiescence required for the stabilization of the endothelium.
Collapse
Affiliation(s)
- Jose Luis Herrera
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701, USA; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701, USA; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
32
|
Pratt HG, Ma L, Dziadowicz SA, Ott S, Whalley T, Szomolay B, Eubank TD, Hu G, Boone BA. Analysis of single nuclear chromatin accessibility reveals unique myeloid populations in human pancreatic ductal adenocarcinoma. Clin Transl Med 2024; 14:e1595. [PMID: 38426634 PMCID: PMC10905544 DOI: 10.1002/ctm2.1595] [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: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND A better understanding of the pancreatic ductal adenocarcinoma (PDAC) immune microenvironment is critical to developing new treatments and improving outcomes. Myeloid cells are of particular importance for PDAC progression; however, the presence of heterogenous subsets with different ontogeny and impact, along with some fluidity between them, (infiltrating monocytes vs. tissue-resident macrophages; M1 vs. M2) makes characterisation of myeloid populations challenging. Recent advances in single cell sequencing technology provide tools for characterisation of immune cell infiltrates, and open chromatin provides source and function data for myeloid cells to assist in more comprehensive characterisation. Thus, we explore single nuclear assay for transposase accessible chromatin (ATAC) sequencing (snATAC-Seq), a method to analyse open gene promoters and transcription factor binding, as an important means for discerning the myeloid composition in human PDAC tumours. METHODS Frozen pancreatic tissues (benign or PDAC) were prepared for snATAC-Seq using 10× Chromium technology. Signac was used for preliminary analysis, clustering and differentially accessible chromatin region identification. The genes annotated in promoter regions were used for Gene Ontology (GO) enrichment and cell type annotation. Gene signatures were used for survival analysis with The Cancer Genome Atlas (TCGA)-pancreatic adenocarcinoma (PAAD) dataset. RESULTS Myeloid cell transcription factor activities were higher in tumour than benign pancreatic samples, enabling us to further stratify tumour myeloid populations. Subcluster analysis revealed eight distinct myeloid populations. GO enrichment demonstrated unique functions for myeloid populations, including interleukin-1b signalling (recruited monocytes) and intracellular protein transport (dendritic cells). The identified gene signature for dendritic cells influenced survival (hazard ratio = .63, p = .03) in the TCGA-PAAD dataset, which was unique to PDAC. CONCLUSIONS These data suggest snATAC-Seq as a method for analysis of frozen human pancreatic tissues to distinguish myeloid populations. An improved understanding of myeloid cell heterogeneity and function is important for developing new treatment targets in PDAC.
Collapse
Affiliation(s)
- Hillary G. Pratt
- Cancer Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Li Ma
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Sebastian A. Dziadowicz
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Sascha Ott
- Warwick Medical SchoolUniversity of WarwickCoventryUK
| | | | - Barbara Szomolay
- Division of Infection and Immunity & Systems Immunity Research InstituteCardiff UniversityCardiffUK
| | - Timothy D. Eubank
- Cancer Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- In Vivo Multifunctional Magnetic Resonance CenterWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Gangqing Hu
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Brian A. Boone
- Cancer Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of SurgeryWest Virginia UniversityMorgantownWest VirginiaUSA
| |
Collapse
|
33
|
Simões JS, Rodrigues RF, Zavan B, Emídio RMP, Soncini R, Boralli VB. Endotoxin-Induced Sepsis on Ceftriaxone-Treated Rats' Ventilatory Mechanics and Pharmacokinetics. Antibiotics (Basel) 2024; 13:83. [PMID: 38247642 PMCID: PMC10812549 DOI: 10.3390/antibiotics13010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Sepsis can trigger acute respiratory distress syndrome (ARDS), which can lead to a series of physiological changes, modifying the effectiveness of therapy and culminating in death. For all experiments, male Wistar rats (200-250 g) were split into the following groups: control and sepsis-induced by endotoxin lipopolysaccharide (LPS); the control group received only intraperitoneal saline or saline + CEF while the treated groups received ceftriaxone (CEF) (100 mg/kg) IP; previously or not with sepsis induction by LPS (1 mg/kg) IP. We evaluated respiratory mechanics, and alveolar bronchial lavage was collected for nitrite and vascular endothelial growth factor (VEGF) quantification and cell evaluation. For pharmacokinetic evaluation, two groups received ceftriaxone, one already exposed to LPS. Respiratory mechanics shows a decrease in total airway resistance, dissipation of viscous energy, and elastance of lung tissues in all sepsis-induced groups compared to the control group. VEGF and NOx values were higher in sepsis animals compared to the control group, and ceftriaxone was able to reduce both parameters. The pharmacokinetic parameters for ceftriaxone, such as bioavailability, absorption, and terminal half-life, were smaller in the sepsis-induced group than in the control group since clearance was higher in septic animals. Despite the pharmacokinetic changes, ceftriaxone showed a reduction in resistance in the airways. In addition, CEF lowers nitrite levels in the lungs and acts on their adverse effects, reflecting pharmacological therapy of the disease.
Collapse
Affiliation(s)
- Juliana Savioli Simões
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (J.S.S.); (R.F.R.)
| | - Rafaela Figueiredo Rodrigues
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (J.S.S.); (R.F.R.)
| | - Bruno Zavan
- Insituto de Ciências da Natureza, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (B.Z.); (R.M.P.E.); (R.S.)
| | - Ricardo Murilo Pereira Emídio
- Insituto de Ciências da Natureza, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (B.Z.); (R.M.P.E.); (R.S.)
| | - Roseli Soncini
- Insituto de Ciências da Natureza, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (B.Z.); (R.M.P.E.); (R.S.)
| | - Vanessa Bergamin Boralli
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (J.S.S.); (R.F.R.)
| |
Collapse
|
34
|
Kaefer SL, Zhang L, Morrison RA, Brookes S, Awonusi O, Shay E, Hoilett OS, Anderson JL, Goergen CJ, Voytik-Harbin S, Halum S. Early Changes in Porcine Larynges Following Injection of Motor-Endplate Expressing Muscle Cells for the Treatment of Unilateral Vocal Fold Paralysis. Laryngoscope 2024; 134:272-282. [PMID: 37436167 PMCID: PMC11790276 DOI: 10.1002/lary.30868] [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: 03/06/2023] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023]
Abstract
OBJECTIVES No curative injectable therapy exists for unilateral vocal fold paralysis. Herein, we explore the early implications of muscle-derived motor-endplate expressing cells (MEEs) for injectable vocal fold medialization after recurrent laryngeal nerve (RLN) injury. METHODS Yucatan minipigs underwent right RLN transection (without repair) and muscle biopsies. Autologous muscle progenitor cells were isolated, cultured, differentiated, and induced to form MEEs. Three weeks after the injury, MEEs or saline were injected into the paralyzed right vocal fold. Outcomes including evoked laryngeal electromyography (LEMG), laryngeal adductor pressure, and acoustic vocalization data were analyzed up to 7 weeks post-injury. Harvested porcine larynges were examined for volume, gene expression, and histology. RESULTS MEE injections were tolerated well, with all pigs demonstrating continued weight gain. Blinded analysis of videolaryngoscopy post-injection revealed infraglottic fullness, and no inflammatory changes. Four weeks after injection, LEMG revealed on average higher right distal RLN activity retention in MEE pigs. MEE-injected pigs on average had vocalization durations, frequencies, and intensities higher than saline pigs. Post-mortem, the MEE-injected larynges revealed statistically greater volume on quantitative 3D ultrasound, and statistically increased expression of neurotrophic factors (BDNF, NGF, NTF3, NTF4, NTN1) on quantitative PCR. CONCLUSIONS Minimally invasive MEE injection appears to establish an early molecular and microenvironmental framework to encourage innate RLN regeneration. Longer follow-up is needed to determine if early findings will translate into functional contraction. LEVEL OF EVIDENCE NA Laryngoscope, 134:272-282, 2024.
Collapse
Affiliation(s)
- Samuel L Kaefer
- Indiana University School of Medicine (IUSM), Indianapolis, IN, U.S.A
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
| | - Lujuan Zhang
- IUSM Department of Otolaryngology-Head and Neck Surgery, Indianapolis, IN, U.S.A
| | - Rachel A Morrison
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
| | - Sarah Brookes
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
| | - Oluwaseyi Awonusi
- Indiana University School of Medicine (IUSM), Indianapolis, IN, U.S.A
| | - Elizabeth Shay
- IUSM Department of Otolaryngology-Head and Neck Surgery, Indianapolis, IN, U.S.A
| | - Orlando S Hoilett
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
- University of Cincinnati Department of Biomedical Engineering, Cincinnati, OH, U.S.A
| | - Jennifer L Anderson
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
| | - Craig J Goergen
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
| | - Sherry Voytik-Harbin
- Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, U.S.A
- Purdue University Department of Basic Medical Sciences, West Lafayette, IN, U.S.A
| | - Stacey Halum
- Indiana University School of Medicine (IUSM), Indianapolis, IN, U.S.A
- IUSM Department of Otolaryngology-Head and Neck Surgery, Indianapolis, IN, U.S.A
- Purdue University Department of Speech, Language, and Hearing Sciences, West Lafayette, IN, U.S.A
| |
Collapse
|
35
|
Siamwala J, Swaminathan A, Chatterjee S. Assessment of Endothelial Barrier Functions in Extra Embryonic Vasculature of Chick Embryo as an Alternative Model. Methods Mol Biol 2024; 2711:185-197. [PMID: 37776458 DOI: 10.1007/978-1-0716-3429-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
Vascular permeability, a tightly regulated process, is a direct measure of angiogenic and immune responses in the endothelium altered in several acute and chronic diseases such as sepsis, high-altitude pulmonary edema (HAPE), high-altitude cerebral edema (HACE), ischemia, and coronavirus disease 2019 (COVID-19) endotheliitis. Both endogenous and exogenous factors such as cytokines, chemokines, and hormones may affect vascular permeability. The conventional tools available for the measurement of vascular permeability in vitro and in vivo based on collagen-coated Transwell and dye-based spectrophotometric methods are indirect measures of permeability. In this chapter, we present our live in ovo protocols based on dextran-Texas red and avian chorioallantoic membrane assay developed using custom-made equipment to assess leakiness of endothelial cell barrier both in vitro and in vivo. Further, we validate this assay using different stressors such as ischemia and hypoxia known to affect endothelial barrier properties by potentiating actin stress fiber disorganization and disrupting the cell-cell junctions.
Collapse
Affiliation(s)
- Jamila Siamwala
- Department of Molecular Pharmacology, Physiology and Biotechnology, Providence, RI, USA.
- Warren Alpert Medical School of Brown University, Providence Veterans Affairs Medical Center, Providence, RI, USA.
| | - Akila Swaminathan
- Department of Biotechnology, Anna University, Chennai, India
- Department of Biotechnology, The University of Burdwan, Burdwan, India
| | - Suvro Chatterjee
- Department of Biotechnology, Anna University, Chennai, India
- Department of Biotechnology, The University of Burdwan, Burdwan, India
| |
Collapse
|
36
|
Bosma EK, Darwesh S, Habani YI, Cammeraat M, Serrano Martinez P, van Breest Smallenburg ME, Zheng JY, Vogels IMC, van Noorden CJF, Schlingemann RO, Klaassen I. Differential roles of eNOS in late effects of VEGF-A on hyperpermeability in different types of endothelial cells. Sci Rep 2023; 13:21436. [PMID: 38052807 PMCID: PMC10698188 DOI: 10.1038/s41598-023-46893-4] [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: 03/03/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Vascular endothelial growth factor (VEGF)-A induces endothelial hyperpermeability, but the molecular pathways remain incompletely understood. Endothelial nitric oxide synthase (eNOS) regulates acute effects of VEGF-A on permeability of endothelial cells (ECs), but it remains unknown whether and how eNOS regulates late effects of VEGF-A-induced hyperpermeability. Here we show that VEGF-A induces hyperpermeability via eNOS-dependent and eNOS-independent mechanisms at 2 days after VEGF-A stimulation. Silencing of expression of the eNOS gene (NOS3) reduced VEGF-A-induced permeability for dextran (70 kDa) and 766 Da-tracer in human dermal microvascular ECs (HDMVECs), but not in human retinal microvascular ECs (HRECs) and human umbilical vein ECs (HUVECs). However, silencing of NOS3 expression in HRECs increased permeability to dextran, BSA and 766 Da-tracer in the absence of VEGF-A stimulation, suggesting a barrier-protective function of eNOS. We also investigated how silencing of NOS3 expression regulates the expression of permeability-related transcripts, and found that NOS3 silencing downregulates the expression of PLVAP, a molecule associated with trans-endothelial transport via caveolae, in HDMVECs and HUVECs, but not in HRECs. Our findings underscore the complexity of VEGF-A-induced permeability pathways in ECs and the role of eNOS therein, and demonstrate that different pathways are activated depending on the EC phenotype.
Collapse
Affiliation(s)
- Esmeralda K Bosma
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Shahan Darwesh
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Yasmin I Habani
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Maxime Cammeraat
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Paola Serrano Martinez
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Mathilda E van Breest Smallenburg
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Jia Y Zheng
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ilse M C Vogels
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands.
| |
Collapse
|
37
|
Gao T, Cheng S, Lu H, Li X, Weng X, Ge J. Histidine Triad Nucleotide-Binding Protein 1 Improves Critical Limb Ischemia by Regulating Mitochondrial Homeostasis. Nutrients 2023; 15:4859. [PMID: 38068718 PMCID: PMC10708213 DOI: 10.3390/nu15234859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Critical limb ischemia (CLI) is a common complication of diabetes mellitus that typically occurs in the later stages of the disease. Vascularization is indeed an important physiological process involving the formation of new blood vessels from existing ones. It occurs in response to various normal and pathophysiological conditions, and one of its critical roles is to compensate for inadequate oxygen supply, which is often seen in situations like chronic limb ischemia (CLI). Histidine triad nucleotide-binding protein 1 (Hint1) is a member of the Hint family that has been shown to attenuate cardiac hypertrophy, but its role in vascularization still needs to be clarified. In this study, we investigated the role of Hint1 in CLI. We found that Hint1 is significantly reduced in the muscle tissue of STZ-induced diabetic mice and high-glucose (HG)-treated endothelial cells (ECs). Hint1 deletion impaired blood flow recovery and vascularization, whereas Hint1 overexpression promoted these processes. In addition, our in vitro study showed that Hint1 deficiency aggravated mitochondrial dysfunction in ECs, as evidenced by impaired mitochondrial respiration, decreased mitochondrial membrane potential, and increased reactive oxygen species. Our findings suggest that Hint1 deficiency impairs blood perfusion by damaging mitochondrial function and that Hint1 may represent a potential therapeutic target for treating CLI.
Collapse
Affiliation(s)
- Tingwen Gao
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Department of Cardiology, Rizhao International Heart Hospital, Rizhao 276825, China
| | - Shuo Cheng
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiao Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
38
|
Dürig J, Calcagni M, Buschmann J. Transition metals in angiogenesis - A narrative review. Mater Today Bio 2023; 22:100757. [PMID: 37593220 PMCID: PMC10430620 DOI: 10.1016/j.mtbio.2023.100757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
The aim of this paper is to offer a narrative review of the literature regarding the influence of transition metals on angiogenesis, excluding lanthanides and actinides. To our knowledge there are not any reviews up to date offering such a summary, which inclined us to write this paper. Angiogenesis describes the process of blood vessel formation, which is an essential requirement for human growth and development. When the complex interplay between pro- and antiangiogenic mediators falls out of balance, angiogenesis can quickly become harmful. As it is so fundamental, both its inhibition and enhancement take part in various diseases, making it a target for therapeutic treatments. Current methods come with limitations, therefore, novel agents are constantly being researched, with metal agents offering promising results. Various transition metals have already been investigated in-depth, with studies indicating both pro- and antiangiogenic properties, respectively. The transition metals are being applied in various formulations, such as nanoparticles, complexes, or scaffold materials. Albeit the increasing attention this field is receiving, there remain many unanswered questions, mostly regarding the molecular mechanisms behind the observed effects. Notably, approximately half of all the transition metals have not yet been investigated regarding potential angiogenic effects. Considering the promising results which have already been established, it should be of great interest to begin investigating the remaining elements whilst also further analyzing the established effects.
Collapse
Affiliation(s)
- Johannes Dürig
- University of Zürich, Faculty of Medicine, Pestalozzistrasse 3, 8032, Zurich, Switzerland
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Maurizio Calcagni
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johanna Buschmann
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
39
|
Chen C, Wang J, Liu C, Hu J, Liu L. Pioneering therapies for post-infarction angiogenesis: Insight into molecular mechanisms and preclinical studies. Biomed Pharmacother 2023; 166:115306. [PMID: 37572633 DOI: 10.1016/j.biopha.2023.115306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Acute myocardial infarction (MI), despite significant progress in its treatment, remains a leading cause of chronic heart failure and cardiovascular events such as cardiac arrest. Promoting angiogenesis in the myocardial tissue after MI to restore blood flow in the ischemic and hypoxic tissue is considered an effective treatment strategy. The repair of the myocardial tissue post-MI involves a robust angiogenic response, with mechanisms involved including endothelial cell proliferation and migration, capillary growth, changes in the extracellular matrix, and stabilization of pericytes for neovascularization. In this review, we provide a detailed overview of six key pathways in angiogenesis post-MI: the PI3K/Akt/mTOR signaling pathway, the Notch signaling pathway, the Wnt/β-catenin signaling pathway, the Hippo signaling pathway, the Sonic Hedgehog signaling pathway, and the JAK/STAT signaling pathway. We also discuss novel therapeutic approaches targeting these pathways, including drug therapy, gene therapy, protein therapy, cell therapy, and extracellular vesicle therapy. A comprehensive understanding of these key pathways and their targeted therapies will aid in our understanding of the pathological and physiological mechanisms of angiogenesis after MI and the development and application of new treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
40
|
Zhao Z, Shan X, Zhang H, Shi X, Huang P, Sun J, He Z, Luo C, Zhang S. Nitric oxide-driven nanotherapeutics for cancer treatment. J Control Release 2023; 362:151-169. [PMID: 37633361 DOI: 10.1016/j.jconrel.2023.08.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
Nitric oxide (NO) is a gaseous molecule endowed with diverse biological functions, offering vast potential in the realm of cancer treatment. Considerable efforts have been dedicated to NO-based cancer therapy owing to its good biosafety and high antitumor activity, as well as its efficient synergistic therapy with other antitumor modalities. However, delivering this gaseous molecule effectively into tumor tissues poses a significant challenge. To this end, nano drug delivery systems (nano-DDSs) have emerged as promising platforms for in vivo efficient NO delivery, with remarkable achievements in recent years. This review aims to provide a summary of the emerging NO-driven antitumor nanotherapeutics. Firstly, the antitumor mechanism and related clinical trials of NO therapy are detailed. Secondly, the latest research developments in the stimulation of endogenous NO synthesis are presented, including the regulation of nitric oxide synthases (NOS) and activation of endogenous NO precursors. Moreover, the emerging nanotherapeutics that rely on tumor-specific delivery of NO donors are outlined. Additionally, NO-driven combined nanotherapeutics for multimodal cancer theranostics are discussed. Finally, the future directions, application prospects, and challenges of NO-driven nanotherapeutics in clinical translation are highlighted.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinzhu Shan
- Department of State Key Laboratory of Natural and Biomimetic Drugs, College of Pharmaceutical Sciences, Peking University, Beijing 100871, PR China
| | - Hongyuan Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Peiqi Huang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shenwu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
41
|
Htun Y, Nakamura S, Nakao Y, Mitsuie T, Ohta K, Arioka M, Yokota T, Inoue E, Inoue K, Tsuchiya T, Koyano K, Konishi Y, Miki T, Ueno M, Kusaka T. Conflicting findings on the effectiveness of hydrogen therapy for ameliorating vascular leakage in a 5-day post hypoxic-ischemic survival piglet model. Sci Rep 2023; 13:10486. [PMID: 37380745 DOI: 10.1038/s41598-023-37577-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a major cause of morbidity and mortality in newborns in both high- and low-income countries. The important determinants of its pathophysiology are neural cells and vascular components. In neonatal HIE, increased vascular permeability due to damage to the blood-brain barrier is associated with seizures and poor outcomes in both translational and clinical studies. In our previous studies, hydrogen gas (H2) improved the neurological outcome of HIE and ameliorated the cell death. In this study, we used albumin immunohistochemistry to assess if H2 inhalation effectively reduced the cerebral vascular leakage. Of 33 piglets subjected to a hypoxic-ischemic insult, 26 piglets were ultimately analyzed. After the insult, the piglets were grouped into normothermia (NT), H2 ventilation (H2), therapeutic hypothermia (TH), and H2 combined with TH (H2-TH) groups. The ratio of albumin stained to unstained areas was analyzed and found to be lower in the H2 group than in the other groups, although the difference was not statistically significant. In this study, H2 therapy did not significantly improve albumin leakage despite the histological images suggesting signs of improvement. Further investigations are warranted to study the efficacy of H2 gas for vascular leakage in neonatal HIE.
Collapse
Affiliation(s)
- Yinmon Htun
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Yasuhiro Nakao
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Tsutomu Mitsuie
- Medical Engineering Equipment Management Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kenichi Ohta
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Makoto Arioka
- Maternal and Perinatal Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takayuki Yokota
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Eri Inoue
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kota Inoue
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Toi Tsuchiya
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kosuke Koyano
- Maternal and Perinatal Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan.
| |
Collapse
|
42
|
El-Tanani M, Nsairat H, Aljabali AA, Serrano-Aroca Á, Mishra V, Mishra Y, Naikoo GA, Alshaer W, Tambuwala MM. Role of mammalian target of rapamycin (mTOR) signalling in oncogenesis. Life Sci 2023; 323:121662. [PMID: 37028545 DOI: 10.1016/j.lfs.2023.121662] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/07/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023]
Abstract
The signalling system known as mammalian target of rapamycin (mTOR) is believed to be required for several biological activities involving cell proliferation. The serine-threonine kinase identified as mTOR recognises PI3K-AKT stress signals. It is well established in the scientific literature that the deregulation of the mTOR pathway plays a crucial role in cancer growth and advancement. This review focuses on the normal functions of mTOR as well as its abnormal roles in cancer development.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire BD7 1DP, United Kingdom.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001, Valencia, Spain.
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Gowhar A Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, Salalah, PC 211, Oman.
| | - Walhan Alshaer
- Cell Therapy Center, the University of Jordan, Amman 11942, Jordan
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, United Kingdom.
| |
Collapse
|
43
|
Zaied AA, Ushio‐Fukai M, Fukai T, Kovacs‐Kasa A, Alhusban S, Sudhahar V, Ganta VC, Annex BH. Pentose Pathway Activation Is Superior to Increased Glycolysis for Therapeutic Angiogenesis in Peripheral Arterial Disease. J Am Heart Assoc 2023; 12:e027986. [PMID: 36974760 PMCID: PMC10122893 DOI: 10.1161/jaha.122.027986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 03/29/2023]
Abstract
Background In endothelial cells (ECs), glycolysis, regulated by PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase, isoform-3), is the major metabolic pathway for ATP generation. In preclinical peripheral artery disease models, VEGF165a (vascular endothelial growth factor165a) and microRNA-93 both promote angiogenesis. Methods and Results Mice following hind-limb ischemia (HLI) and ECs with, and without, hypoxia and serum starvation were examined with, and without, microRNA-93 and VEGF165a. Post-HLI perfusion recovery was monitored. EC metabolism was studied using seahorse assay, and the expression and activity of major metabolism genes were assessed. Reactive oxygen species levels and EC permeability were evaluated. C57Bl/6J mice generated a robust angiogenic response to HLI, with ECs from ischemic versus nonischemic muscle demonstrating no increase in glycolysis. Balb/CJ mice generated a poor angiogenic response post-HLI; ischemic versus nonischemic ECs demonstrated significant increase in glycolysis. MicroRNA-93-treated Balb/CJ mice post-HLI showed better perfusion recovery, with ischemic versus nonischemic ECs showing no increase in glycolysis. VEGF165a-treated Balb/CJ mice post-HLI showed no improvement in perfusion recovery with ischemic versus nonischemic ECs showing significant increase in glycolysis. ECs under hypoxia and serum starvation upregulated PFKFB3. In ECs under hypoxia and serum starvation, VEGF165a versus control significantly upregulated PFKFB3 and glycolysis, whereas miR-93 versus control demonstrated no increase in PFKFB3 or glycolysis. MicroRNA-93 versus VEGF165a upregulated glucose-6-phosphate dehydrogenase expression and activity, activating the pentose phosphate pathway. MicroRNA-93 versus control increased reduced nicotinamide adenine dinucleotide phosphate and virtually eliminated the increase in reactive oxygen species. In ECs under hypoxia and serum starvation, VEGF165a significantly increased and miR-93 decreased EC permeability. Conclusions In peripheral artery disease, activation of the pentose phosphate pathway to promote angiogenesis may offer potential therapeutic advantages.
Collapse
Affiliation(s)
- Abdelrahman A. Zaied
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Masuko Ushio‐Fukai
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Tohru Fukai
- Departments of Pharmacology and ToxicologyMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Charlie Norwood Veterans Affairs Medical CenterAugustaGAUSA
| | - Anita Kovacs‐Kasa
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Suhib Alhusban
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Varadarajan Sudhahar
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Vijay C. Ganta
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Brian H. Annex
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| |
Collapse
|
44
|
Abstract
Hypertension is associated with important alterations in the morphology of small arteries and arterioles. Vascular-specific manifestations are changes in the structure and function of vascular smooth muscle cells, extracellular matrix, perivascular tissues, and endothelial cells. Arteriole and capillary remodeling and capillary rarefaction have been observed in hypertensive animals and human beings which contribute to increased vascular resistance. An impairment of different angiogenetic factors, such as VEGF (vascular endothelial growth factor), VEGFR-2 (vascular endothelial growth factor receptor-2), TIMP-1 (tissue inhibitor matrix metalloproteinases-1), and TSP-1 (thrombospondin-1), seems to be responsible for the reduction of the microvascular network. Exercise training has been shown to improve vascular structure and function in hypertension not only in the large arteries but also in the peripheral circulation. Exercise training may regress microvascular remodeling and normalize capillary density, leading to capillary growth possibly by increasing proangiogenic stimuli such as VEGF. Exercise enhances endothelium-dependent vascular relaxation through nitric oxide release increase and oxidative stress reduction. Other mechanisms include improved balance between prostacyclin and thromboxane levels, lower circulating levels of endothelin-1, attenuation of infiltration of immune cells into perivascular adipose tissue, and increase of local adiponectin secretion. In addition, exercise training favorably modulates the expression of several microRNAs leading to a positive modification in muscle fiber composition. Identifying the bioactive molecules and biological mechanisms that mediate exercise benefits through pathways that differ from those used by antihypertensive drugs may help to improve our knowledge of hypertension pathophysiology and facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Carolina De Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Italy (C.D.C., D.R.)
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Italy (C.D.C., D.R.).,Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy (D.R.)
| | - Paolo Palatini
- Department of Medicine, University of Padova, Padua, Italy (P.P.)
| |
Collapse
|
45
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
46
|
Dhahri W, Dussault S, Raguema N, Desjarlais M, Rivard A. Stimulation of soluble guanylate cyclase activity with riociguat promotes angiogenesis and improves neovascularization after limb ischemia. Atherosclerosis 2023; 372:32-40. [PMID: 37023506 DOI: 10.1016/j.atherosclerosis.2023.03.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND AND AIMS The NO-cGMP pathway is essential for angiogenesis, vasculogenesis and post-natal neovascularization. The key enzyme responsible for the synthesis of cGMP following binding of NO is soluble guanylate cyclase (sGC). Riociguat is the first member of a novel class of compounds known as sGC stimulators. We tested the hypothesis that stimulation of sGC with riociguat might improve neovascularization in response to ischemia. METHODS In vitro, the angiogenic effect of riociguat was tested in human umbilical vein endothelial cells (HUVECs). In vivo, neovascularization was investigated in a mouse model of limb ischemia. C57Bl/6 mice were treated by gavage with 3 mg/kg/day of riociguat for a total of 28 days. After two weeks of treatment, hindlimb ischemia was surgically induced by femoral artery removal. RESULTS In a matrigel assay in vitro, riociguat dose-dependently stimulates tubule formation in HUVECs. Cell migration (scratch assay) is also increased in HUVECs treated with riociguat. At the molecular level, riociguat treatment leads to rapid activation of the p44/p42 MAP kinase pathway in HUVECs. Inhibition of protein kinase G (PKG) activity supresses both p44/p42 MAP kinase activation and angiogenesis in HUVECs treated with riociguat. In vivo, treatment with riociguat improves blood flow recovery after ischemia (Laser Doppler imaging), and increases capillary density in ischemic muscles (CD31 immunostaining). Clinically, this is associated with a significant decrease of ambulatory impairment and ischemic damages. Interestingly, mice treated with riociguat also show a 94% increase in the number of bone marrow-derived pro-angiogenic cells (PACs) compared to control mice. Moreover, riociguat treatment is associated with a significant improvement of PAC functions including migratory capacity, adhesion to an endothelial monolayer, and integration into endothelial tubular networks. CONCLUSIONS The sGC stimulator riociguat promotes angiogenesis and improves neovascularization after ischemia. The mechanism involves PKG-dependent activation of p44/p42 MAP kinase pathway, together with an improvement of PAC number and functions. sGC stimulation could constitute a novel therapeutic strategy to reduce tissue ischemia in patients with severe atherosclerotic diseases.
Collapse
Affiliation(s)
- Wahiba Dhahri
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Sylvie Dussault
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Nozha Raguema
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Michel Desjarlais
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Alain Rivard
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada.
| |
Collapse
|
47
|
Machado DE, Saggioro EM, Sales Junior SF, Alessandra-Perini J, de Campos Gomes Diniz L, Coelho WS, Zancan P, Perini JA. Clotrimazole is effective, safe and tolerable for the treatment of endometriosis and functions by downregulating inducible nitric oxide synthase and modulating oxidative stress biomarkers. Mol Cell Endocrinol 2023; 564:111883. [PMID: 36736881 DOI: 10.1016/j.mce.2023.111883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/04/2023]
Abstract
This study investigated the mechanism of action of clotrimazole (CTZ) and its adverse effects in a model of endometriosis. After autologous endometrial implantation, 18 rats were randomized into two treatment groups: 200 mg/kg CTZ or vehicle for 15 consecutive days. The lesion growth, implant size, glandular atrophy, nitric oxide (NO) serum levels, number of macrophage cells and inducible nitric oxide synthase (iNOS) immunoreactivity were significantly reduced in the CTZ group compared with the control. CTZ (p < 0.05) reduced the lipid peroxidation and protein carbonylation levels in the liver but did not alter the superoxide dismutase (SOD), glutathione (GSH) or glutathione S-transferase (GST) levels in the brain; however, the drug significantly reduced SOD activity and enhanced GST activity in the liver. These results suggest that CTZ interferes with reactive nitrogen species production by downregulating iNOS expression and thus enhances the antioxidant system to promote atrophy and regression of endometriotic lesions, without adverse effects on the brain and/or liver.
Collapse
Affiliation(s)
- Daniel Escorsim Machado
- Laboratório de Pesquisa de Ciências Farmacêuticas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Enrico Mendes Saggioro
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública Sérgio Arouca, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Laboratório de Avaliação e Promoção da Saúde Ambiental, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Sidney Fernandes Sales Junior
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública Sérgio Arouca, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Jéssica Alessandra-Perini
- Laboratório de Pesquisa de Ciências Farmacêuticas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Luciana de Campos Gomes Diniz
- Laboratório de Pesquisa de Ciências Farmacêuticas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Wagner Santos Coelho
- Laboratório de Pesquisa de Ciências Farmacêuticas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Patrícia Zancan
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Jamila Alessandra Perini
- Laboratório de Pesquisa de Ciências Farmacêuticas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil.
| |
Collapse
|
48
|
Aissopou E, Protogerou A, Theodossiadis P, Sfikakis PP, Chatziralli I. Alterations in Retinal Vessel Diameters in Patients with Retinal Vein Occlusion before and after Treatment with Intravitreal Ranibizumab. J Pers Med 2023; 13:jpm13020351. [PMID: 36836585 PMCID: PMC9963538 DOI: 10.3390/jpm13020351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/09/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Purpose: To investigate the alterations of retinal vessel diameters in patients with macular edema secondary to retinal vein occlusion (RVO), before and after treatment with intravitreal ranibizumab. Methods: Digital retinal images were obtained from 16 patients and retinal vessel diameters were measured before and three months after treatment with intravitreal ranibizumab with validated software to determine central retinal arteriolar and venular equivalents, as well as arteriolar to venular ratio. Results: In 17 eyes of 16 patients with macular edema secondary to RVO (10 with branch RVO and 6 with central RVO) aged 67 ± 10.2 years, we found that diameters of both retinal arterioles and venules were significantly decreased after intravitreal ranibizumab treatment. Specifically, the central retinal arteriolar equivalent was 215.2 ± 11.2 μm at baseline and 201.2 ± 11.1 μm at month 3 after treatment (p < 0.001), while the central retinal venular equivalent was 233.8 ± 29.6 μm before treatment versus 207.6 ± 21.7 μm at month 3 after treatment (p < 0.001). Conclusions: A significant vasoconstriction in both retinal arterioles and venules in patients with RVO was found at month 3 after intravitreal ranibizumab treatment compared to baseline. This could be of clinical importance, since the degree of vasoconstriction might be an early marker of treatment efficacy, compatible with the idea that hypoxia is the major trigger of VEGF in RVO. Further studies should be conducted to confirm our findings.
Collapse
Affiliation(s)
- Evaggelia Aissopou
- 2nd Department of Ophthalmology, National and Kapodistrian University of Athens, 12462 Athens, Greece
- Correspondence:
| | - Athanasios Protogerou
- Cardiovascular Prevention Unit, Department of Pathophysiology, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiotis Theodossiadis
- 2nd Department of Ophthalmology, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Petros P. Sfikakis
- Rheumatology Unit, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Irini Chatziralli
- 2nd Department of Ophthalmology, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
49
|
Duess JW, Gosemann JH, Kaskova Gheorghescu A, Puri P, Thompson J. Y-27632 Impairs Angiogenesis on Extra-Embryonic Vasculature in Post-Gastrulation Chick Embryos. TOXICS 2023; 11:134. [PMID: 36851009 PMCID: PMC9962381 DOI: 10.3390/toxics11020134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
Y-27632 inhibits Rho-associated coiled-coil-containing protein kinase (ROCK) signaling, which is involved in various embryonic developmental processes, including angiogenesis, by controlling actin cytoskeleton assembly and cell contractility. Administration of Y-27632 impairs cytoskeletal arrangements in post-gastrulation chick embryos, leading to ventral body wall defects (VBWDs). Impaired angiogenesis has been hypothesized to contribute to VBWDs. ROCK is essential in transmitting signals downstream of vascular endothelial growth factor (VEGF). VEGF-mediated angiogenesis induces gene expressions and alterations of the actin cytoskeleton upon binding to VEGF receptors (VEGFRs). The aim of this study was to investigate effects of Y-27632 on angiogenesis in post-gastrulation chick embryos during early embryogenesis. After 60 h incubation, embryos in shell-less culture were treated with Y-27632 or vehicle for controls. Y-27632-treated embryos showed reduced extra-embryonic blood vessel formation with impaired circulation of the yolk sac, confirmed by fractal analysis. Western blot confirmed impaired ROCK downstream signaling by decreased expression of phosphorylated myosin light chain. Interestingly, RT-PCR demonstrated increased gene expression of VEGF and VEGFR-2 1 h post-treatment. Protein levels of VEGF were higher in Y-27632-treated embryos at 8 h following treatment, whereas no difference was seen in membranes. We hypothesize that administration of Y-27632 impairs vessel formation during angiogenesis, which may contribute to failure of VWB closure, causing VBWDs.
Collapse
Affiliation(s)
- Johannes W. Duess
- Department of Pediatric Surgery, University of Leipzig, 04103 Leipzig, Germany
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, 12 Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Belfield, 4 Dublin, Ireland
| | - Jan-Hendrik Gosemann
- Department of Pediatric Surgery, University of Leipzig, 04103 Leipzig, Germany
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, 12 Dublin, Ireland
| | | | - Prem Puri
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, 12 Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Belfield, 4 Dublin, Ireland
| | - Jennifer Thompson
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, 12 Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Belfield, 4 Dublin, Ireland
| |
Collapse
|
50
|
Huang W, Zhang J, Luo L, Yu Y, Sun T. Nitric Oxide and Tumors: From Small-Molecule Donor to Combination Therapy. ACS Biomater Sci Eng 2023; 9:139-152. [PMID: 36576226 DOI: 10.1021/acsbiomaterials.2c01247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
As an important endogenous signaling molecule, nitric oxide (NO) is involved in various physiological and pathological activities in living organisms. It is proved that NO plays a critical role in tumor therapy, while the extremely short half-life and nonspecific distribution of NO greatly limit its further clinical applications. Thus, the past few decades have witnessed the progress made in conquering these shortcomings, including developing innovative NO donors, especially smart and multimodal nanoplatforms. These platforms can precisely control the spatiotemporal distribution of therapeutic agents in the organism, which make big differences in tumor treatment. Here current NO therapeutic mechanisms for cancer, NO donors from small molecules to smart-responsive nanodrug delivery platforms, and NO-based combination therapy are comprehensively reviewed, emphasizing outstanding breakthroughs in these fields and hoping to bring new insights into NO-based tumor treatments.
Collapse
Affiliation(s)
- Wan Huang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Jun Zhang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Li Luo
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Yao Yu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.,State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|