1
|
Wang L, Li X, Men X, Liu X, Luo J. Research progress on antioxidants and protein aggregation inhibitors in cataract prevention and therapy (Review). Mol Med Rep 2025; 31:22. [PMID: 39513587 PMCID: PMC11574704 DOI: 10.3892/mmr.2024.13387] [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: 06/25/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Cataracts are primarily caused by aging or gene mutations and are the leading cause of blindness globally. As the older population increases, the number of patients with a cataract is expected to grow rapidly. At present, cataract surgery to replace the lens with an artificial intraocular lens is the principal treatment method. However, surgery has several drawbacks, including economic burdens and complications such as inflammation, xerophthalmia, macular edema and posterior capsular opacification. Thus, developing an effective non‑surgical treatment strategy is beneficial to both patients and public health. Mechanistically, cataract formation may be due to various reasons but is primarily initiated and promoted by oxidative stress and is closely associated with crystallin aggregation. In the present review, the current research progress on anti‑cataract drugs, including antioxidants and protein aggregation inhibitors is examined. It summarizes strategies for preventing and treating cataract through cell apoptosis and protein aggregation inhibition while discussing their limitations and further prospects.
Collapse
Affiliation(s)
- Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xin Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xiaoju Men
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xiangyi Liu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Jinque Luo
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| |
Collapse
|
2
|
Almoallim HS, Aljawdah HM, Bharathi M, Manickam R, Abudoleh SM, Hussein-Al-Ali SH, Surya P. Fabrication of ibuprofen/naringenin-coloaded into zein/sodium caseinate nanoparticles: evaluation of antiproliferative activity and apoptosis induction in liver cancer cells. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:2703-2722. [PMID: 39217620 DOI: 10.1080/09205063.2024.2391653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Nowadays, liver cancer is one of the most disturbing types of cancer that can affect either sex. Nanoparticles (NPs) of zein/sodium caseinate incorporating ibuprofen (IBU) and naringenin (NAR) have improved bioavailability and a high encapsulation efficiency (EE%). These nanoparticles are uniformly spherical. In vitro, cytotoxicity analysis on HepG2 cell lines, which are used to study human liver cancer, shows that encapsulated drugs (86.49% ± 1.90, and 78.52% ± 1.98 for NAR and IBU, respectively) have significantly lower IC50 values than individual drugs or their combined free form. In addition, the combination indices of 0.623 and 0.155 for IBU and NAR, respectively, show that the two have joint beneficial effects. The scratch wound healing assay results also show that the free drugs and the engineered NPs have a more significant anti-migratory effect than the untreated cells. The designed nanoparticles also reduce angiogenesis and proliferation while inducing apoptosis, according to in vitro results. In conclusion, a new approach to treating liver cancer may lie in the nanoencapsulation of numerous drugs within nanoparticles.
Collapse
Affiliation(s)
- Hesham S Almoallim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Hossam M Aljawdah
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Muruganantham Bharathi
- Centre for Drug Discovery, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Raja Manickam
- Department of Chemistry, Sona College of Technology, Salem, Tamil Nadu, India
| | - Suha Mujahed Abudoleh
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Middle East University, Amman, Jordan
| | | | - Parthasarathy Surya
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
3
|
Shaikh II, Bhandari R, Singh S, Zhu X, Ali Shahzad K, Shao C, Cheng L, Xiao J. Therapeutic potential of EVs loaded with CB2 receptor agonist in spinal cord injury via the Nrf2/HO-1 pathway. Redox Rep 2024; 29:2420572. [PMID: 39466990 PMCID: PMC11520104 DOI: 10.1080/13510002.2024.2420572] [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: 10/30/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) poses a challenge due to limited treatment options. Recently, the effect and mechanism of Exo-loaded cannabinoid receptor type 2 (CB2) agonist AM1241(Exo + AM1241) have been applied in other inflammatory diseases but not in SCI. METHODS The SCI model was set up using C57BL/6 mice, followed by the treatment of Exo, AM1241, and Exo + AM1241. We assessed the effects of the following treatments on motor function recovery using BMS, and evaluated histological changes, apoptosis activity, inflammation, and oxidative stress in the SCI mice model. Additionally, the effect of following treatments on spinal cord neural stem cells (NSCs) was evaluated under lipopolysaccharides (LPS) induced inflammatory and oxidative models and, glutamate (Gluts) induced cell apoptosis models. RESULT Our results demonstrated that Exo + AM1241 treatment significantly improved motor function recovery, after SCI by decreasing proinflammatory cytokines, and suppressing astrocyte/microglia (GFAP/Iba1) activation in the injury zone. Additionally, this treatment reduces pro-apoptotic proteins (Bax and caspase 3), increases the levels of the anti-apoptotic protein Bcl-2, enhances antioxidant defenses by boosting SOD and GSH, and lowers oxidative stress markers such as MDA. It also activates the Nuclear factor erythroid-2 (Nrf2) related factor 2 signaling pathway, thereby enhancing tissue protection against damage and cell death.
Collapse
Affiliation(s)
- Imran Ibrahim Shaikh
- Lishui People's Hospital, Central Laboratory of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, People’s Republic of China
- Ministry of Education, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Shanghai, People’s Republic of China
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Ramesh Bhandari
- Shanghai Tenth Peoples Hospital, Affiliated to Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Shekhar Singh
- Shanghai Tenth Peoples Hospital, Affiliated to Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Xu Zhu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Khawar Ali Shahzad
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Chuxiao Shao
- Lishui People's Hospital, Central Laboratory of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, People’s Republic of China
| | - Liming Cheng
- Ministry of Education, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Shanghai, People’s Republic of China
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Jian Xiao
- Lishui People's Hospital, Central Laboratory of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
4
|
Abida, Altamimi ASA, Ghaboura N, Balaraman AK, Rajput P, Bansal P, Rawat S, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H, Deb PK. Therapeutic Potential of lncRNAs in Regulating Disulfidptosis for Cancer Treatment. Pathol Res Pract 2024; 263:155657. [PMID: 39437641 DOI: 10.1016/j.prp.2024.155657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Non-coding RNAs (lncRNAs) play critical roles in various cellular processes, including a novel form of regulated cell death known as disulfidptosis, characterized by accumulating protein disulfide bonds and severe endoplasmic reticulum stress. This review highlights the therapeutic potential of lncRNAs in regulating disulfidptosis for cancer treatment, emphasizing their influence on key pathway components such as GPX4, SLC7A11, and PDIA family members. Recent studies have demonstrated that targeting specific lncRNAs can sensitize cancer cells to disulfidptosis, offering a promising approach to cancer therapy. The regulation of disulfidptosis by lncRNAs involves various signaling pathways, including oxidative stress, ER stress, and calcium signaling. This review also discusses the molecular mechanisms underlying lncRNA regulation of disulfidptosis, the challenges of developing lncRNA-based therapies, and the future potential of this rapidly advancing field in cancer research.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice, Pharmacy Program, Batterjee Medical College, PO Box 6231, Jeddah 21442, Saudi Arabia
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Pranchal Rajput
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institue of Technology (BIT), Mesra, Ranchi, Jharkhand 835215, India
| |
Collapse
|
5
|
Zhao Q, Li Y, Zhao X, Zhou J, Zheng Y, Li Z. Apelin-13 alleviates intrauterine adhesion by inhibiting epithelial-mesenchymal transition of endometrial epithelial cells and promoting angiogenesis. Hum Cell 2024; 37:1613-1623. [PMID: 39158615 DOI: 10.1007/s13577-024-01117-3] [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: 01/15/2024] [Accepted: 06/01/2024] [Indexed: 08/20/2024]
Abstract
Intrauterine adhesion (IUA) is a common complication of surgical manipulation of the uterine cavity such as abortion. The pathology of IUA is characterized by fibrosis, but the pathogenesis is not fully understood. The function of Apelin-13 in IUA and related mechanisms were investigated in this study. The IUA rat model was established. The pathological changes and fibrosis degree of rat uterine tissues were detected by HE and Masson staining after intraperitoneal injection of Apelin-13. Epithelial-mesenchymal transition (EMT) of endometrial epithelial cells and endothelial-mesenchymal transition (EnMT) of vein endothelial cells were induced by TGF-β1. Tube-forming assay using HUVEC was implemented to detect the effect of Apelin-13 upon angiogenesis. IHC staining, immunofluorescence staining, and Western blot were conducted to detect the expression levels of EMT markers, angiogenesis, and key proteins of the TGF-β1/Smad signaling. Apelin-13 significantly alleviated IUA and fibrosis, and increased endometrial thickness and gland number in IUA rats. In addition, Apelin-13 significantly reversed EMT and EnMT induced by IUA modeling and TGF-β1, promoted the tube-forming ability of HUVEC, and up-regulated the expression of angiogenesis-related proteins. Mechanistically, Apelin-13 significantly suppressed smad2/3 phosphorylation and inhibited the TGF-β1/Smad signaling via its receptor APJ. Apelin-13 might alleviate IUA via repressing the TGF-β1/Smad pathway and is expected to be a potent therapeutic option for the clinical treatment of IUA.
Collapse
Affiliation(s)
- Qun Zhao
- Health Management Medicine Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Yuyan Li
- Nanchang University Queen Mary School, Nanchang, 330031, Jiangxi, China
| | - Xingping Zhao
- Department of Gynecology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Jiahui Zhou
- Department of Spinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Yifan Zheng
- Department of Spinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Zhiyue Li
- Department of Spinal Surgery, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
6
|
Molavand M, Ebrahimnezhade N, Kiani A, Yousefi B, Nazari A, Majidinia M. Regulation of autophagy by non-coding RNAs in human glioblastoma. Med Oncol 2024; 41:260. [PMID: 39375229 DOI: 10.1007/s12032-024-02513-3] [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: 05/03/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024]
Abstract
Glioblastoma, a lethal form of brain cancer, poses substantial challenges in treatment due to its aggressive nature and resistance to standard therapies like radiation and chemotherapy. Autophagy has a crucial role in glioblastoma progression by supporting cellular homeostasis and promoting survival under stressful conditions. Non-coding RNAs (ncRNAs) play diverse biological roles including, gene regulation, chromatin remodeling, and the maintenance of cellular homeostasis. Emerging evidence reveals the intricate regulatory mechanisms of autophagy orchestrated by non-coding RNAs (ncRNAs) in glioblastoma. The diverse roles of these ncRNAs in regulating crucial autophagy-related pathways, including AMPK/mTOR signaling, the PI3K/AKT pathway, Beclin1, and other autophagy-triggering system regulation, sheds light on ncRNAs biological mechanisms in the proliferation, invasion, and therapy response of glioblastoma cells. Furthermore, the clinical implications of targeting ncRNA-regulated autophagy as a promising therapeutic strategy for glioblastoma treatment are in the spotlight of ongoing studies. In this review, we delve into our current understanding of how ncRNAs regulate autophagy in glioblastoma, with a specific focus on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), and their intricate interplay with therapy response.
Collapse
Affiliation(s)
- Mehran Molavand
- Student Research Commitee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Ebrahimnezhade
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Arash Kiani
- Student Research Commite, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
- Molecular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Nazari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
- Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
7
|
Almujri SS, Almalki WH. The paradox of autophagy in cancer: NEAT1's role in tumorigenesis and therapeutic resistance. Pathol Res Pract 2024; 262:155523. [PMID: 39173466 DOI: 10.1016/j.prp.2024.155523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
Cancer remains a current active problem of modern medicine, a process during which cell growth and proliferation become uncontrolled. However, the role of autophagy in the oncological processes is counterintuitive and, at the same time, increasingly influential on the formation, development, and response to therapy of oncological diseases. Autophagy is a vital cellular process that removes defective proteins and organelles and supports cellular homeostasis. Autophagy can enhance the ability to form new tumors and suppress this formation in cancer. The dual potential of apoptosis may be the reason for this duality in either promoting or impeding the survival of cancer cells, depending on the situation, including starvation or treatment stress. Furthermore, long non-coding RNA NEAT1, which has been linked to several stages of carcinogenesis and in all forms of the illness, has drawn attention as a major player in cancer biology. NEAT1 is a structural portion of nuclear paraspeckles and has roles in deactivating expression in both transcriptional and post-transcriptional levels. NEAT1 acts in carcinogenesis in numerous ways, comprising interactions with microRNAs, the influence of gene articulation, regulation of epigenetics, and engagement in signalling cascades. In addition, the complexity of NEAT1's role in cancer occurrence is amplified by its place in regulating cancer stem cells and the tumor microenvironment. NEAT1's interaction with autophagy further complicates the already complicated function of this RNA in cancer biology. NEAT1 has been linked to autophagy in several types of cancer, influencing autophagy pathways and altering its stress response and tumor cell viability. Understanding the interrelation between NEAT1, autophagy, and cancer will enable practitioners to identify novel treatment targets and approaches to disrupt oncogenic processes, reduce the occurrence of treatment resistance, and increase patient survival rates. Specialized treatment strategies and regimens are thus achievable. In the present review, the authors analyze sophisticated relationship schemes in cancer: The NEAT1 pathway and the process of autophagy.
Collapse
Affiliation(s)
- Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia.
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
8
|
Syed RU, Alshammari MD, Banu H, Khojali WMA, Jafar M, Nagaraju P, Alshammari A. Targeting the autophagy-miRNA axis in prostate cancer: toward novel diagnostic and therapeutic strategies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7421-7437. [PMID: 38761210 DOI: 10.1007/s00210-024-03153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
Since prostate cancer is one of the leading causes of cancer-related death, a better understanding of the molecular pathways guiding its development is imperative. A key factor in prostate cancer is autophagy, a cellular mechanism that affects both cell survival and death. Autophagy is essential in maintaining cellular homeostasis. Autophagy is a physiological mechanism wherein redundant or malfunctioning cellular constituents are broken down and recycled. It is essential for preserving cellular homeostasis and is implicated in several physiological and pathological conditions, including cancer. Autophagy has been linked to metastasis, tumor development, and treatment resistance in prostate cancer. The deregulation of miRNAs related to autophagy appears to be a crucial element in the etiology of prostate cancer. These miRNAs influence the destiny of cancer cells by finely regulating autophagic mechanisms. Numerous investigations have emphasized the dual function of specific miRNAs in prostate cancer, which alter autophagy-related pathways to function as either tumor suppressors or oncogenes. Notably, miRNAs have been linked to the control of autophagy and the proliferation, apoptosis, and migration of prostate cancer cells. To create customized therapy approaches, it is imperative to comprehend the dynamic interplay between autophagy and miRNAs in prostate cancer. The identification of key miRNAs provides potential diagnostic and prognostic markers. Unraveling the complex network of lncRNAs, like PCA3, also expands the repertoire of molecular targets for therapeutic interventions. This review explores the intricate interplay between autophagy and miRNAs in prostate cancer, focusing on their regulatory roles in cellular processes ranging from survival to programmed cell death.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia.
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
| | - Humera Banu
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Weam M A Khojali
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, 14415, Sudan
| | - Mohammed Jafar
- Department of Pharmaceutics, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 34212, Saudi Arabia.
| | - Potnuri Nagaraju
- Department of Pharmaceutics, Mandesh Institute of Pharmaceutical Science and Research Center, Mhaswad, Maharashtra, India
| | - Alia Alshammari
- Department of Pharmaceutics, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
| |
Collapse
|
9
|
Tian Y, Wang X, Sun Y, Xiong X, Zeng W, Yang K, Zhao H, Deng Y, Song D. NPTX1 Mediates the Facilitating Effects of Hypoxia-Stimulated Human Adipocytes on Adipose-Derived Stem Cell Activation and Autologous Adipose Graft Survival Rate. Aesthetic Plast Surg 2024; 48:4203-4216. [PMID: 38789811 DOI: 10.1007/s00266-024-04118-7] [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: 12/05/2023] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Autologous adipose tissue is an ideal material for soft tissue filling and transplantation; however, high volumes of fat absorption over time lead to a relatively low overall survival percentage. The survival and differentiation of adipose-derived stem cells (ADSCs) in the transplanted microenvironment might improve adipose graft survival. Adipocytes have been reported to affect ADSC activation. However, its underlying mechanisms remain unclear. METHODS Human ADSCs were incubated in a culture medium supplemented with hypoxic or normoxic conditioned culture medium (CM) derived from human adipocytes. Neuronal Pentraxin 1 (NPTX1) was overexpressed or knocked down in human adipocytes using an overexpression vector (NPTX1 OE) or small interfering RNA (siRNA) transfection, respectively. ADSC differentiation and paracrine secretion were assessed. Nude mice were implanted with human adipocytes and ADSCs. The adipose tissue was subsequently evaluated by histological analysis. RESULTS CM from hypoxic-stimulated human adipocytes significantly facilitated the differentiation ability and paracrine levels of ADSCs. NPTX1 was significantly up-regulated in human adipocytes exposed to hypoxic conditions. In vitro, CM derived from hypoxia-stimulated human adipocytes or NPTX1-overexpressing human adipocytes exposed to normoxia promoted ADSC differentiation and paracrine; after silencing NPTX1, the facilitating effects of hypoxia-treated human adipocytes on ADSC activation were eliminated. Similarly, in vivo, the NPTX1 OE + normoxia-CM group saw improved histological morphology and fat integrity, less fibrosis and inflammation, and increased vessel numbers compared with the OE NC + normoxia-CM group; the adipocyte grafts of the si-NC + hypoxia-CM group yielded the most improved histological morphology, fat integrity, and the most vessel numbers. However, these enhancements of ADSC activation and adipose graft survival were partially abolished by NPTX1 knockdown in human adipocytes. CONCLUSION NPTX1 might mediate the facilitating effects of hypoxia-stimulated human adipocytes on ADSC activation, thereby improving adipose tissue survival rate after autologous fat transplantation and the effectiveness of autologous fat transplantation through promoting ADSC activation. LEVEL OF EVIDENCE III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Yi Tian
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiancheng Wang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Yang Sun
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiang Xiong
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Weiliang Zeng
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Kai Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Hongli Zhao
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yiwen Deng
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Dandan Song
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| |
Collapse
|
10
|
Jiahong C, Junfeng D, Shuxian L, Tao W, Liyun W, Hongfu W. The role of immune cell death in spermatogenesis and male fertility. J Reprod Immunol 2024; 165:104291. [PMID: 38986230 DOI: 10.1016/j.jri.2024.104291] [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: 05/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
The male reproductive system provides a distinctive shield to the immune system, safeguarding germ cells (GCs) from autoimmune harm. The testis in mammals creates a unique immunological setting due to its exceptional immune privilege and potent local innate immunity. which can result from a number of different circumstances, including disorders of the pituitary gland, GC aplasia, and immunological elements. Apoptosis, or programmed cell death (PCD), is essential for mammalian spermatogenesis to maintain and ensure an appropriate number of GCs that correspond with the supporting capability of the Sertoli cells. Apoptosis is substantial in controlling the number of GCs in the testis throughout spermatogenesis, and any dysregulation of this process has been linked to male infertility. There is a number of evidence about the potential of PCD in designing novel therapeutic approaches in the treatment of infertility. A detailed understanding of PCD and the processes that underlie immunological infertility can contribute to the progress in designing strategies to prevent and treat male infertility. This review will provide a summary of the role of immune cell death in male reproduction and infertility and describe the therapeutic strategies and agents for treatment based on immune cell death.
Collapse
Affiliation(s)
- Chen Jiahong
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Department of Venereal Diseases and Integrated Chinese and Western Medicine and Bone Paralysis, Longjiang Hospital of Shunde District, Foshan, China
| | - Dong Junfeng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Liu Shuxian
- Guangzhou Huadu District Maternal and Child Health Care Hospital (Huzhong Hospital of Huadu District), Guangzhou, China
| | - Wang Tao
- Department of Venereal Diseases and Integrated Chinese and Western Medicine and Bone Paralysis, Longjiang Hospital of Shunde District, Foshan, China.
| | - Wang Liyun
- Guangzhou Huadu District Maternal and Child Health Care Hospital (Huzhong Hospital of Huadu District), Guangzhou, China.
| | - Wu Hongfu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
11
|
Guo Z, Zhang J, Li M, Xing Z, Li X, Qing J, Zhang Y, Zhu L, Qi M, Zou X. Mechanism of action of quercetin in regulating cellular autophagy in multiple organs of Goto-Kakizaki rats through the PI3K/Akt/mTOR pathway. Front Med (Lausanne) 2024; 11:1442071. [PMID: 39211336 PMCID: PMC11357923 DOI: 10.3389/fmed.2024.1442071] [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: 06/01/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Objective This experimental study investigated the protective function of quercetin on the liver, spleen, and kidneys of Goto-Kakizaki (GK) rats and explores its mechanism of action on autophagy-related factors and pathways. Materials and methods GK rats were randomly divided into three groups: DM, DM + L-Que, and DM + H-Que, with age-matched Wistar rats serving as the control group. The control and DM groups were gavaged with saline, and the quercetin-treated group was gavaged with quercetin for 8 weeks each. Weekly blood glucose levels were monitored. Upon conclusion of the experiment, blood samples were gathered for lipid and hepatic and renal function analyses. The histopathologic morphology and lipid deposition in rats were examined. Disease-related targets were identified using molecular docking methods and network pharmacology analysis. Subsequently, immunohistochemical analysis was performed, followed by Western blotting to evaluate the levels of autophagy-related proteins and proteins in the AKT/PI3K/mTOR pathway, as well as their phosphorylation levels. Results The results showed that, compared with the control group, the DM group exhibited significant increases in blood glucose, serum liver and kidney markers, liver fat vacuoles, and inflammatory cell infiltration. Immunohistochemistry (IHC) results indicated that quercetin reduced the extensive expression of AKT, P62, and mTOR in the liver and spleen of diabetic rats. The expression of autophagy and pathway-related proteins, such as P62, PI3K, P-PI3K, Akt, P-AKT, mTOR, and P-mTOR, was upregulated, while the expression of LC3A/LC3B, Beclin-1, Pink-1, and Parkin was downregulated. Conversely, the quercetin group showed a reduction in liver and kidney injury serum markers by decreasing lipid deposition and cell necrosis, indicating that quercetin has protective effects on the liver, spleen, and kidneys of GK rats. Additionally, in the quercetin group, the expression of autophagy and pathway-related proteins such as LC3A/LC3B, Beclin-1, Pink-1, and Parkin was upregulated, while the expression of P62, PI3K, P-PI3K, Akt, P-AKT, mTOR, and P-mTOR was downregulated, with statistically significant correlations. Conclusion Quercetin markedly ameliorates liver, spleen, and kidney damage in GK rats, potentially through the inhibition of the PI3K/Akt/mTOR pathway, promoting autophagy. This research offers a rationale to the therapeutic potential of quercetin in mitigating organ damage associated with diabetes.
Collapse
Affiliation(s)
- Zhiqun Guo
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Jingyu Zhang
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Mianxin Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Zengwei Xing
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Xi Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Jiaqi Qing
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Yuan Zhang
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Lemei Zhu
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| | - Mingxu Qi
- Department of Cardiovascular Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Xuemin Zou
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha, China
- School of Public Health, Changsha Medical University, Changsha, China
| |
Collapse
|
12
|
Patiño-Martinez E, Nakabo S, Jiang K, Carmona-Rivera C, Tsai WL, Claybaugh D, Yu ZX, Romero A, Bohrnsen E, Schwarz B, Solís-Barbosa MA, Blanco LP, Naqi M, Temesgen-Oyelakin Y, Davis M, Manna Z, Gupta S, Mehta N, Naz F, dell'Orso S, Hasni S, Kaplan MJ. The Aconitate Decarboxylase 1/Itaconate Pathway Modulates Immune Dysregulation and Associates with Cardiovascular Disease Markers and Disease Activity in Systemic Lupus Erythematosus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:419-434. [PMID: 38949522 DOI: 10.4049/jimmunol.2400241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
The Krebs cycle enzyme aconitate decarboxylase 1 (ACOD1) mediates itaconate synthesis in monocytes and macrophages. Previously, we reported that administration of 4-octyl itaconate to lupus-prone mice abrogated immune dysregulation and clinical features. In this study, we explore the role of the endogenous ACOD1/itaconate pathway in the development of TLR7-induced lupus (imiquimod [IMQ] model). We found that, in vitro, ACOD1 was induced in mouse bone marrow-derived macrophages and human monocyte-derived macrophages following TLR7 stimulation. This induction was partially dependent on type I IFN receptor signaling and on specific intracellular pathways. In the IMQ-induced mouse model of lupus, ACOD1 knockout (Acod1-/-) displayed disruptions of the splenic architecture, increased serum levels of anti-dsDNA and proinflammatory cytokines, and enhanced kidney immune complex deposition and proteinuria, when compared with the IMQ-treated wild-type mice. Consistent with these results, Acod1-/- bone marrow-derived macrophages treated in vitro with IMQ showed higher proinflammatory features. Furthermore, itaconate serum levels in systemic lupus erythematosus patients were decreased compared with healthy individuals, in association with disease activity and specific perturbed cardiometabolic parameters. These findings suggest that the ACOD1/itaconate pathway plays important immunomodulatory and vasculoprotective roles in systemic lupus erythematosus, supporting the potential therapeutic role of itaconate analogs in autoimmune diseases.
Collapse
Affiliation(s)
- Eduardo Patiño-Martinez
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Shuichiro Nakabo
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Kan Jiang
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Wanxia Li Tsai
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Dillon Claybaugh
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Aracely Romero
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Eric Bohrnsen
- Protein & Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Benjamin Schwarz
- Protein & Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Miguel A Solís-Barbosa
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luz P Blanco
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Mohammad Naqi
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Yenealem Temesgen-Oyelakin
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Michael Davis
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Zerai Manna
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Sarthak Gupta
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Nehal Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Faiza Naz
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Stefania dell'Orso
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Sarfaraz Hasni
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
13
|
Ren Q, Dong Y, Huang Y, Xiao J, Ma Y, Liu Y, Sun H, Dai Y, Shi C, Wang S. Nrf2 induces angiogenesis in spinal cystic echinococcosis by activating autophagy via regulating oxidative stress. Biochem Pharmacol 2024; 226:116337. [PMID: 38844265 DOI: 10.1016/j.bcp.2024.116337] [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: 01/31/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Spinal cystic echinococcosis (CE) is a rare but malignant zoonosis that can cause disability or even death in more than half of patients. Due to the complex pathological features, it is not curable by conventional drugs and surgery, so new therapeutic targets urgently need to be discovered. In this study, we clarify the occurrence of the phenomenon of spinal encapsulation angiogenesis and explore its underlying molecular mechanisms. A co-culture system was established by protoscoleces (PSCs) with human umbilical vein endothelial cells (HUVECs) which showed a high expression level of Nrf2. A short hairpin RNA (shRNA) and Sulforaphane (SFN) affecting the expression of Nrf2 were used to treat HUVECs. The results showed that Nrf2 could promote the tube formation of HUVECs. Nrf2 also exerts a protective effect against HUVECs, which is achieved by promoting NQO1 expression to stabilize ROS levels. Furthermore, autophagy activation significantly promotes angiogenesis in the spinal echinococcosis model (SEM) as a result of Nrf2 regulation of oxidative stress. These results suggest that the ROS/Nrf2/autophagy axis can induce angiogenesis and may be a potential target for the treatment of spinal cystic echinococcosis.
Collapse
Affiliation(s)
- Qian Ren
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yimin Dong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430101, China
| | - Yiping Huang
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Jun Xiao
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yibo Ma
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yaqing Liu
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Haohao Sun
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yi Dai
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Chenhui Shi
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China.
| | - Sibo Wang
- Department of Spine Surgery, Xi'an Jiao Tong University Affiliated HongHui Hospital, Xi'an 710054, China.
| |
Collapse
|
14
|
Ayaz M, Alam A, Zainab, Elhenawy AA, Ur Rehman N, Ur Rahman S, Ali M, Latif A, Al-Harrasi A, Ahmad M. Designing and Synthesis of Novel Fexofenadine-Derived Hydrazone-Schiff Bases as Potential Urease Inhibitors: In-Vitro, Molecular Docking and DFT Investigations. Chem Biodivers 2024; 21:e202400704. [PMID: 38781003 DOI: 10.1002/cbdv.202400704] [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: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Thirteen novel hydrazone-Schiff bases (3-15) of fexofenadine were succesfully synthesized, structurally deduced and finally assessed their capability to inhibit urease enzyme (in vitro). In the series, six compounds 12 (IC50=10.19±0.16 μM), 11 (IC50=15.05±1.11 μM), 10 (IC50=17.01±1.23 μM), 9 (IC50=17.22±0.81 μM), 13 (IC50=19.31±0.18 μM), and 14 (IC50=19.62±0.21 μM) displayed strong inhibitory action better than the standard thiourea (IC50=21.14±0.24 μM), while the remaining compounds displayed significant to less inhibition. LUMO and HOMO showed the transferring of charges from molecules to biological transfer and MEP map showed the chemically reactive zone appropriate for drug action are calculated using DFT. AIM charges, non-bonding orbitals, and ELF are also computed. The urease protein binding analysis benefited from the docking studies.
Collapse
Affiliation(s)
- Muhammad Ayaz
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Zainab
- College of Chemistry and Materials Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Najeeb Ur Rehman
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Sajjad Ur Rahman
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Mumtaz Ali
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Abdul Latif
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| |
Collapse
|
15
|
Kong X, Xu L, Mou Z, Lyu W, Shan K, Wang L, Liu F, Rong F, Li J, Wei P. The anti-inflammatory effects of itaconate and its derivatives in neurological disorders. Cytokine Growth Factor Rev 2024; 78:37-49. [PMID: 38981775 DOI: 10.1016/j.cytogfr.2024.07.001] [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: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Almost 16 % of the global population is affected by neurological disorders, including neurodegenerative and cerebral neuroimmune diseases, triggered by acute or chronic inflammation. Neuroinflammation is recognized as a common pathogenic mechanism in a wide array of neurological conditions including Alzheimer's disease, Parkinson's disease, postoperative cognitive dysfunction, stroke, traumatic brain injury, and multiple sclerosis. Inflammatory process in the central nervous system (CNS) can lead to neuronal damage and neuronal apoptosis, consequently exacerbating these diseases. Itaconate, an immunomodulatory metabolite from the tricarboxylic acid cycle, suppresses neuroinflammation and modulates the CNS immune response. Emerging human studies suggest that itaconate levels in plasma and cerebrospinal fluid may serve as biomarkers associated with inflammatory responses in neurological disorders. Preclinical studies have shown that itaconate and its highly cell-permeable derivatives are promising candidates for preventing and treating neuroinflammation-related neurological disorders. The underlying mechanism may involve the regulation of immune cells in the CNS and neuroinflammation-related signaling pathways and molecules including Nrf2/KEAP1 signaling pathway, reactive oxygen species, and NLRP3 inflammasome. Here, we introduce the metabolism and function of itaconate and the synthesis and development of its derivatives. We summarize the potential impact and therapeutic potential of itaconate and its derivatives on brain immune cells and the associated signaling pathways and molecules, based on preclinical evidence via various neurological disorder models. We also discuss the challenges and potential solutions for clinical translation to promote further research on itaconate and its derivatives for neuroinflammation-related neurological disorders.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Lin Xu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Zheng Mou
- Department of Pharmacy, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Wenyuan Lyu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Kaiyue Shan
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Longfei Wang
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Fanghao Liu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Fei Rong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Jianjun Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Penghui Wei
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
| |
Collapse
|
16
|
Zheng M, Zhao F. The IL-12 family of heterodimeric cytokines in polycystic ovarian syndrome: biological role in induction, regulation, and treatment. Immunol Res 2024; 72:583-591. [PMID: 38771486 DOI: 10.1007/s12026-024-09487-4] [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: 03/26/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a diverse endocrine disorder widely recognized as the prevailing metabolic condition among women in their reproductive years. The precise pathophysiological mechanisms underlying PCOS remain incompletely understood. However, existing evidence suggests that the development of PCOS may be linked to factors such as abdominal obesity, hyperandrogenism, and insulin resistance (IR). Excessive central adiposity in women with PCOS may lead to the development of a chronic, low-grade inflammation characterized by the activation of proinflammatory cytokines. The cytokines that belong to the IL-12 family are a collection of distinct heterodimeric cytokines that include IL-12, IL-23, IL-27, and IL-35. Recent research has provided further evidence regarding the significance of IL-12 cytokines in influencing both innate and adaptive immune responses in different diseases. Additionally, these studies have discovered diverse roles for certain members of the IL-12 family, encompassing multiple immunological functions that can either act as effectors or regulators. In this discourse, we examine the distinctive and atypical structural and functional attributes of this particular cytokine family. This study aims to offer a comprehensive overview of the pathophysiological significance of the IL-12 family cytokines in PCOS patients. Additionally, the therapeutic potential of the cytokines as novel approaches for PCOS treatment will be proposed.
Collapse
Affiliation(s)
- Mingyan Zheng
- Gynaecology and Obstetrics, Weifang People's Hospital, Weifang, 261000, Shandong, China
| | - Feng Zhao
- Gynaecology and Obstetrics, Weifang People's Hospital, Weifang, 261000, Shandong, China.
| |
Collapse
|
17
|
Yin X, Feng L, Hua Q, Ye J, Cai L. Progress in the study of mechanisms and pathways related to the survival of random skin flaps. Updates Surg 2024; 76:1195-1202. [PMID: 38308185 DOI: 10.1007/s13304-023-01746-7] [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: 11/21/2023] [Accepted: 12/29/2023] [Indexed: 02/04/2024]
Abstract
The clinical application of random flaps in wound repair has been a topic of discussion. Random flaps are prone to necrosis due to the lack of well-defined vascular blood supply during transfer surgery. Their clinical utility is restricted, financial and psychological burdens is imposed on patients due to this limitation. The survival of random skin flaps depends on factors such as ischemia-reperfusion injury, oxidative stress, local inflammatory response, and neovascularization. This review aims to provide an overview of the evidence supporting the use of random flaps in clinical practice. In addition, this review explores the impact of different medications on signaling pathways within the flap's local microcirculation and investigates the interconnections between these pathways.
Collapse
Affiliation(s)
- Xinghao Yin
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Liang Feng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Qianqian Hua
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiangtian Ye
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Leyi Cai
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, XueYuan West Road, Luheng District, Wenzhou, Zhejiang, 325000, People's Republic of China.
| |
Collapse
|
18
|
Saleem M, Hussain A, Hanif M, Ahmad H, Khan SU, Haider S, Rafiq M, Paracha RN, Park SH. Synthesis, Invitro Cytotoxic Activity and Optical Analysis of Substituted Schiff Base Derivatives. J Fluoresc 2024:10.1007/s10895-024-03803-9. [PMID: 38913090 DOI: 10.1007/s10895-024-03803-9] [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: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 06/25/2024]
Abstract
Fluorescent cytotoxic compounds with readout delivery are crucial in chemotherapy. The growing demands of these treatment strategies require the novel heterocyclic molecules with better selectivity alongside fluorescence marker potential. In this context, a series of nine isatin Schiff base derivatives 4a-i were synthesized, characterized and evaluated for UV-visible, fluorescence, thermal and bioanalysis in order to explore the effect of structure on their bioprofiles. The analogue 4d exhibited maximum cytotoxic activity on Hella cells with percentage inhibition of 83% at 50 µM and 100% at 150 µM concentrations while 4c showed minimum cytotoxic activity with the value of 19% at 50 µM and 22% at 150 µM concentrations. Meanwhile, 4g was found to exhibit maximum inhibition potential towards Vero Cells with the percentage inhibition values of 83 at 50 µM concentration. The overall SAR study showed that the para-fluoro-substituted isatin moieties exhibited the appreciable percentage inhibition while the least activity was delivered by the isatin derivatives with para-bromo substitution.
Collapse
Affiliation(s)
- Muhammad Saleem
- Department of Chemistry, Thal University Bhakkar, Bhakkar, Pakistan.
- Department of Chemistry, University of Sargodha, Sargodha, Pakistan.
| | - Abrar Hussain
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute, Jeongeup, 56212, Republic of Korea
- Radiation Science, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Muhammad Hanif
- Department of Chemistry, GC University Faisalabad, Sub Campus Layyah, -31200, Pakistan
| | - Hufsa Ahmad
- Department of Chemistry, The University of Lahore, Sargodha Campus, Sargodha, Pakistan
| | - Salah Uddin Khan
- College of Engineering, King Saud University, P.O.Box 800, 11421, Riyadh, Saudi Arabia
| | - Sajjad Haider
- Chemical Engineering Department, College of Engineering, King Saud University, P.O.Box 800, 11421, Riyadh, Saudi Arabia
| | - Muhammad Rafiq
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, -6300, Pakistan
| | | | - Sang Hyun Park
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute, Jeongeup, 56212, Republic of Korea.
- Radiation Science, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
19
|
Su H, Wang Z, Zhou L, Liu D, Zhang N. Regulation of the Nrf2/HO-1 axis by mesenchymal stem cells-derived extracellular vesicles: implications for disease treatment. Front Cell Dev Biol 2024; 12:1397954. [PMID: 38915448 PMCID: PMC11194436 DOI: 10.3389/fcell.2024.1397954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/06/2024] [Indexed: 06/26/2024] Open
Abstract
This comprehensive review inspects the therapeutic potential of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) across multiple organ systems. Examining their impact on the integumentary, respiratory, cardiovascular, urinary, and skeletal systems, the study highlights the versatility of MSC-EVs in addressing diverse medical conditions. Key pathways, such as Nrf2/HO-1, consistently emerge as central mediators of their antioxidative and anti-inflammatory effects. From expediting diabetic wound healing to mitigating oxidative stress-induced skin injuries, alleviating acute lung injuries, and even offering solutions for conditions like myocardial infarction and renal ischemia-reperfusion injury, MSC-EVs demonstrate promising therapeutic efficacy. Their adaptability to different administration routes and identifying specific factors opens avenues for innovative regenerative strategies. This review positions MSC-EVs as promising candidates for future clinical applications, providing a comprehensive overview of their potential impact on regenerative medicine.
Collapse
Affiliation(s)
- Hua Su
- Xingyi People’s Hospital, Xingyi, China
| | | | - Lidan Zhou
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dezhi Liu
- Xingyi People’s Hospital, Xingyi, China
| | | |
Collapse
|
20
|
Bordbar S, Li Z, Lotfibakhshaiesh N, Ai J, Tavassoli A, Beheshtizadeh N, Vainieri L, Khanmohammadi M, Sayahpour FA, Baghaban Eslaminejad M, Azami M, Grad S, Alini M. Cartilage tissue engineering using decellularized biomatrix hydrogel containing TGF-β-loaded alginate microspheres in mechanically loaded bioreactor. Sci Rep 2024; 14:11991. [PMID: 38796487 PMCID: PMC11127927 DOI: 10.1038/s41598-024-62474-5] [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: 07/29/2023] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Physiochemical tissue inducers and mechanical stimulation are both efficient variables in cartilage tissue fabrication and regeneration. In the presence of biomolecules, decellularized extracellular matrix (ECM) may trigger and enhance stem cell proliferation and differentiation. Here, we investigated the controlled release of transforming growth factor beta (TGF-β1) as an active mediator of mesenchymal stromal cells (MSCs) in a biocompatible scaffold and mechanical stimulation for cartilage tissue engineering. ECM-derived hydrogel with TGF-β1-loaded alginate-based microspheres (MSs) was created to promote human MSC chondrogenic development. Ex vivo explants and a complicated multiaxial loading bioreactor replicated the physiological conditions. Hydrogels with/without MSs and TGF-β1 were highly cytocompatible. MSCs in ECM-derived hydrogel containing TGF-β1/MSs showed comparable chondrogenic gene expression levels as those hydrogels with TGF-β1 added in culture media or those without TGF-β1. However, constructs with TGF-β1 directly added within the hydrogel had inferior properties under unloaded conditions. The ECM-derived hydrogel group including TGF-β1/MSs under loading circumstances formed better cartilage matrix in an ex vivo osteochondral defect than control settings. This study demonstrates that controlled local delivery of TGF-β1 using MSs and mechanical loading is essential for neocartilage formation by MSCs and that further optimization is needed to prevent MSC differentiation towards hypertrophy.
Collapse
Affiliation(s)
- Sima Bordbar
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- AO Research Institute Davos, Davos, Switzerland
| | - Zhen Li
- AO Research Institute Davos, Davos, Switzerland
| | - Nasrin Lotfibakhshaiesh
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Jafar Ai
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Tavassoli
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mehdi Khanmohammadi
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Wołoska 141, 02-507, Warsaw, Poland
| | | | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mahmoud Azami
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland.
| |
Collapse
|
21
|
Alam A, Gul S, Zainab, Khan M, Elhenawy AA, Islam MS, Ali M, Ali Shah SA, Latif A, Ahmad M. Synthesis of 2,4-dihydroxyacetophenone derivatives as potent PDE-1 and -3 inhibitors: in vitro and in silico insights. Future Med Chem 2024; 16:1185-1203. [PMID: 38989989 PMCID: PMC11382721 DOI: 10.1080/17568919.2024.2342707] [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: 10/29/2023] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Aim: Synthesis of novel bis-Schiff bases having potent inhibitory activity against phosphodiesterase (PDE-1 and -3) enzymes, potentially offering therapeutic implications for various conditions. Methods: Bis-Schiff bases were synthesized by refluxing 2,4-dihydroxyacetophenone with hydrazine hydrate, followed by treatment of substituted aldehydes with the resulting hydrazone to obtain the product compounds. After structural confirmation, the compounds were screened for their in vitro PDE-1 and -3 inhibitory activities. Results: The prepared compounds exhibited noteworthy inhibitory efficacy against PDE-1 and -3 enzymes by comparing with suramin standard. To clarify the binding interactions between the drugs, PDE-1 and -3 active sites, molecular docking studies were carried out. Conclusion: The potent compounds discovered in this study may be good candidates for drug development.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Sana Gul
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Zainab
- College of Chemistry & Materials Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Majid Khan
- H.E.J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mumtaz Ali
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Puncak Alam Campus, 42300 Bandar Puncak Alam,Selangor D. E., Malaysia
| | - Abdul Latif
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| |
Collapse
|
22
|
Bellahcene F, Benarous K, Mermer A, Boulebd H, Serseg T, Linani A, Kaouka A, Yousfi M, Syed A, Elgorban AM, Ozeki Y, Kawsar SM. Unveiling potent Schiff base derivatives with selective xanthine oxidase inhibition: In silico and in vitro approach. Saudi Pharm J 2024; 32:102062. [PMID: 38601975 PMCID: PMC11004395 DOI: 10.1016/j.jsps.2024.102062] [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: 02/24/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
This research describes the synthesis by an environmentally-friendly method, microwave irradiation, development and analysis of three novel and one previously identified Schiff base derivative as a potential inhibitor of bovine xanthine oxidase (BXO), a key enzyme implicated in the progression of gout. Meticulous experimentation revealed that these compounds (10, 9, 4, and 7) have noteworthy inhibitory effects on BXO, with IC50 values ranging from 149.56 µM to 263.60 µM, indicating their good efficacy compared to that of the standard control. The validation of these results was further enhanced through comprehensive in silico studies, which revealed the pivotal interactions between the inhibitors and the catalytic sites of BXO, with a particular emphasis on the imine group (-C = N-) functionalities. Intriguingly, the compounds exhibiting the highest inhibition rates also showcase advantageous ADMET profiles, alongside encouraging initial assessments via PASS, hinting at their broad-spectrum potential. The implications of these findings are profound, suggesting that these Schiff base derivatives not only offer a new vantage point for the inhibition of BXO but also hold considerable promise as innovative therapeutic agents in the management and treatment of gout, marking a significant leap forward in the quest for more effective gout interventions.
Collapse
Affiliation(s)
- Fatna Bellahcene
- Laboratory of Fundamental Sciences, Faculty of Sciences, University of Amar Telidji, Laghouat, Algeria
| | - Khedidja Benarous
- Laboratory of Fundamental Sciences, Faculty of Sciences, University of Amar Telidji, Laghouat, Algeria
| | - Arif Mermer
- Department of Biotechnology, University of Health Sciences, İstanbul, Turkey
- Experimental Medicine Application and Research Center, Validebag Research Park, University of Health Sciences, İstanbul, Turkey
- Department of Pharmacy, University of Health Sciences, İstanbul, Turkey
| | - Houssem Boulebd
- Department of Chemistry, Faculty of Exact Sciences, University of Constantine 1, Constantine, Algeria
| | - Talia Serseg
- Laboratory of Fundamental Sciences, Faculty of Sciences, University of Amar Telidji, Laghouat, Algeria
- Laboratoire des Sciences Appliquées et Didactiques, Ecole Normale Supérieure de Laghouat, Algeria
| | - Abderahmane Linani
- Laboratory of Fundamental Sciences, Faculty of Sciences, University of Amar Telidji, Laghouat, Algeria
| | - Alaeddine Kaouka
- Laboratoire des Sciences Appliquées et Didactiques, Ecole Normale Supérieure de Laghouat, Algeria
| | - Mohamed Yousfi
- Laboratory of Fundamental Sciences, Faculty of Sciences, University of Amar Telidji, Laghouat, Algeria
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Abdallah M. Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Yasuhiro Ozeki
- Graduate School of NanoBio Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Sarkar M.A. Kawsar
- Laboratory of Carbohydrate and Nucleoside Chemistry, Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong 4331, Bangladesh
| |
Collapse
|
23
|
Zhang YW, Pang X, Yang Y. Hydrogels containing KYNA promote angiogenesis and inhibit inflammation to improve the survival rate of multi-territory perforator flaps. Biomed Pharmacother 2024; 174:116454. [PMID: 38640710 DOI: 10.1016/j.biopha.2024.116454] [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: 11/07/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND A new spray adhesive (KYNA-PF127) was established through the combination of thermosensitive hydrogel (Pluronic F127) and KYNA, aimed to investigate the effect of KYNA-PF127 on multi-territory perforator flaps and its possible molecular mechanism. MATERIALS AND METHODS 36 SD male rats with 250-300 g were randomly divided into 3 groups (n = 12): control group, blank glue group and KYNA-PF127 group. KYNA-PF127 hydrogel was prepared and characterized for its morphology and properties using scanning electron microscopy. CCK-8 assay, scratch wound assay, transwell assay, tube formation assay and Ki67 staining were used to study the effect of KYNA-PF127 on the proliferation, migration, and tube formation of HUVECs. VEGF and FGF2 were measured by qPCR to evaluate the angiogenesis capacity of HUVECs in vitro. In vivo, the effect of each group on the survival area of the cross-zone perforator flap was evaluated, and angiogenesis was evaluated by HE and immunofluorescence (CD31 and MMP-9). The effect of inflammation on skin collagen fibers was assessed by Masson. Immunohistochemistry (SOD1, IL-1β, TNF-α) was used to evaluate the effects of oxidative stress and inflammatory factors on multi-territory flaps. RESULTS KYNA-PF127 has good sustained release and biocompatibility at 25% concentration. KYNA-PF127 promoted the proliferation, migration, and angiogenesis of HUVECs in vitro. In vivo, the survival area of multi-territory perforator flaps and angiogenic capability have increased after KYNA-PF127 intervention. KYNA-PF127 could effectively reduce the oxidative stress and inflammation of multi-territory perforator flaps. CONCLUSION KYNA-PF127 promotes angiogenesis through its antioxidant stress and anti-inflammatory effects, and shows potential clinical value in promoting the survival viability and drug delivery of multi-territory perforator flaps.
Collapse
Affiliation(s)
- Ya-Wei Zhang
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiaoyang Pang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yan Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.
| |
Collapse
|
24
|
Huo H, Chang Y. Hemodynamic study of the ICA aneurysm evolution to attain the cerebral aneurysm rupture risk. Sci Rep 2024; 14:8984. [PMID: 38637544 PMCID: PMC11026371 DOI: 10.1038/s41598-024-59242-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
The influence of the aneurysm evolution on the hemodynamic characteristic of the blood flow inside the sac region is comprehensively investigated. By using the computational method, the blood flow through the vessel and aneurysm of the sac region is examined to find the role of aneurysm evolution on the wall shear stress, pressure, and risk of aneurysm rupture. Three different models of ICA aneurysms are chosen for the investigation of the aneurysm evolution at risk of rupture. Obtained data shows that the evolution of the aneurysm decreases the wall shear stress and pressure on the sac surface while an oscillatory index of blood increases on the aneurysm wall.
Collapse
Affiliation(s)
- Huaying Huo
- Shanxi Provincial People's Hospital, TaiYuan, Shanxi, 030012, China
| | - Yigang Chang
- Shanxi Provincial People's Hospital, TaiYuan, Shanxi, 030012, China.
| |
Collapse
|
25
|
Saleem M, Hussain A, Khan SU, Haider S, Lee KH, Park SH. Symmetrical Ligand's Fabricated Porous Silicon Surface Based Photoluminescence Sensor for Metal Detection and Entrapment. J Fluoresc 2024:10.1007/s10895-024-03697-7. [PMID: 38625572 DOI: 10.1007/s10895-024-03697-7] [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: 03/01/2024] [Accepted: 03/26/2024] [Indexed: 04/17/2024]
Abstract
This study was based on the development of surface-based photoluminescence sensor for metal detection, quantification, and sample purification employing the solid sensory chip having the capability of metal entrapment. The Co(II), Cu(II) and Hg(II) sensitive fluorescence sensor (TP) was first synthesized and characterized its sensing abilities towards tested metal ions by using fluorescence spectral investigation while the synthesis and complexation of the receptor was confirmed by the chromogenic, optical, spectroscopic and spectrometric analysis. Under optical investigation, the ligand solution exhibited substantial chromogenic changes as well as spectral variations upon reacting with copper, cobalt, and mercuric ions, while these behaviors were not seen for the rest of tested metallic ions i.e., Na+, Ag+, Ni2+, Mn2+, Pd2+, Pb2+, Cd2+, Zn2+, Sn2+, Fe2+, Fe3+, Cr3+, and Al3+. These colorimetric alterations and spectral shifting could potentially be employed to detect and quantify these specific metal ions. After the establishment of the ligand's selective complexation ability towards selected metals, it was fabricated over the substituted porous silicon surface (FPS) keeping in view of the development of surface-based photoluminescence sensor (TP-FPS) for the selected metal sensation and entrapment to purify the sample just be putting off the metal entrapped sensory solid chip. Surface characterization and ligand fabrication was inspected by plan and cross sectional electron microscopic investigations, vibrational and electronic spectral analysis. The sensitivity of the ligand (TP) in the solution phase metal discrimination was determined by employing the fluorescence titration analysis of the ligand solution after progressive induction of Co2+, Cu2+, and Hg2+, which afford the detection limit values of 2.14 × 10- 8, 3.47 × 10- 8 and 3.13 × 10- 3, respectively. Concurrently, photoluminescence titration of the surface fabricated sensor (TP-FPS) revealed detection limit values of 3.14 × 10- 9, 7.43 × 10- 9, and 8.21 × 10- 4, respectively, for the selected metal ions.
Collapse
Affiliation(s)
- Muhammad Saleem
- Department of Chemistry, Thal University Bhakkar, Bhakkar, Pakistan.
- Department of Chemistry, University of Sargodha, Sargodha, Pakistan.
| | - Abrar Hussain
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute, Jeongeup, 56212, Republic of Korea
- Radiation Science, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Salah Uddin Khan
- College of Engineering, King Saud University, P.O.Box 800, Riyadh, 11421, Saudi Arabia
| | - Sajjad Haider
- Chemical Engineering Department, College of Engineering, King Saud University, P.O.Box 800, Riyadh, 11421, Saudi Arabia
| | - Ki Hwan Lee
- Kongju National University, Gongju, Chungnam, 314-701, Republic of Korea
| | - Sang Hyun Park
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute, Jeongeup, 56212, Republic of Korea.
- Radiation Science, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
26
|
Lagzian A, Askari M, Haeri MS, Sheikhi N, Banihashemi S, Nabi-Afjadi M, Malekzadegan Y. Increased V-ATPase activity can lead to chemo-resistance in oral squamous cell carcinoma via autophagy induction: new insights. Med Oncol 2024; 41:108. [PMID: 38592406 DOI: 10.1007/s12032-024-02313-9] [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: 11/27/2023] [Accepted: 01/23/2024] [Indexed: 04/10/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a cancer type with a high rate of recurrence and a poor prognosis. Tumor chemo-resistance remains an issue for OSCC patients despite the availability of multimodal therapy options, which causes an increase in tumor invasiveness. Vacuolar ATPase (V-ATPase), appears to be one of the most significant molecules implicated in MDR in tumors like OSCC. It is primarily responsible for controlling the acidity in the solid tumors' microenvironment, which interferes with the absorption of chemotherapeutic medications. However, the exact cellular and molecular mechanisms V-ATPase plays in OSCC chemo-resistance have not been understood. Uncovering these mechanisms can contribute to combating OSCC chemo-resistance and poor prognosis. Hence, in this review, we suggest that one of these underlying mechanisms is autophagy induced by V-ATPase which can potentially contribute to OSCC chemo-resistance. Finally, specialized autophagy and V-ATPase inhibitors may be beneficial as an approach to reduce drug resistance to anticancer therapies in addition to serving as coadjuvants in antitumor treatments. Also, V-ATPase could be a prognostic factor for OSCC patients. However, in the future, more investigations are required to demonstrate these suggestions and hypotheses.
Collapse
Affiliation(s)
- Ahmadreza Lagzian
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Marziye Askari
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Melika Sadat Haeri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nastaran Sheikhi
- Biotechnology Department, Biological Sciences Faculty, Alzahra University, Tehran, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trend University, Nottingham, UK
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Yalda Malekzadegan
- Department of Microbiology, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
27
|
Xu M, Zhao M, Zhu M, Yuan H, Li Z, Yan P, Ma C, Zhao H, Wang S, Wan R, Wang L, Yu G. Hibiscus manihot L. flower extract induces anticancer activity through modulation of apoptosis and autophagy in A549 cells. Sci Rep 2024; 14:8102. [PMID: 38582921 PMCID: PMC10998869 DOI: 10.1038/s41598-024-58439-3] [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: 01/21/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024] Open
Abstract
Lung cancer is a major public health issue and heavy burden in China and worldwide due to its high incidence and mortality without effective treatment. It's imperative to develop new treatments to overcome drug resistance. Natural products from food source, given their wide-ranging and long-term benefits, have been increasingly used in tumor prevention and treatment. This study revealed that Hibiscus manihot L. flower extract (HML) suppressed the proliferation and migration of A549 cells in a dose and time dependent manner and disrupting cell cycle progression. HML markedly enhanced the accumulation of ROS, stimulated the dissipation of mitochondrial membrane potential (MMP) and that facilitated mitophagy through the loss of mitochondrial function. In addition, HML induced apoptosis by activation of the PTEN-P53 pathway and inhibition of ATG5/7-dependent autophagy induced by PINK1-mediated mitophagy in A549 cells. Moreover, HML exert anticancer effects together with 5-FU through synergistic effect. Taken together, HML may serve as a potential tumor prevention and adjuvant treatment for its functional attributes.
Collapse
Affiliation(s)
- Minglu Xu
- School of Chemistry and Chemical Engineering, Henan Institute of Science and Technology, Xinxiang, 453-003, Henan, China
| | - Mengxia Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Miaomiao Zhu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Hongmei Yuan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Peishuo Yan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Chi Ma
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Huabin Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Shenghui Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Ruyan Wan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China.
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal university, 46 Jianshe Road, Xinxiang, 453007, Henan, China.
| |
Collapse
|
28
|
Lei X, Ye W, Safdarin F, Baghaei S. Microfluidics devices for sports: A review on technology for biomedical application used in fields such as biomedicine, drug encapsulation, preparation of nanoparticles, cell targeting, analysis, diagnosis, and cell culture. Tissue Cell 2024; 87:102339. [PMID: 38432127 DOI: 10.1016/j.tice.2024.102339] [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: 11/28/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
Microfluidics is an interdisciplinary field that combines knowledge from various disciplines, including biology, chemistry, sports medicine, fluid dynamics, kinetic biomechanics, and microelectronics, to manipulate and control fluids and particles in micron-scale channels and chambers. These channels and chambers can be fabricated using different materials and methods to achieve various geometries and shapes. Microfluidics has numerous biomedical applications, such as drug encapsulation, nanoparticle preparation, cell targeting, analysis, diagnosis, and treatment of sports injuries in both professional and non-professional athletes. It can also be used in other fields, such as biological analysis, chemical synthesis, optics, and acceleration in the treatment of critical sports injuries. The objective of this review is to provide a comprehensive overview of microfluidic technology, including its fabrication methods, current platform materials, and its applications in sports medicine. Biocompatible, biodegradable, and semi-crystalline polymers with unique mechanical and thermal properties are one of the promising materials in microfluidic technology. Despite the numerous advantages of microfluidic technology, further research and development are necessary. Although the technology offers benefits such as ease of operation and cost efficiency, it is still in its early stages. In conclusion, this review emphasizes the potential of microfluidic technology and highlights the need for continued research to fully exploit its potential in the biomedical field and sport applications.
Collapse
Affiliation(s)
- Xuehui Lei
- Graduate School of Wuhan Institute of Physical Education, Wuhan 430079, China
| | - Weiwu Ye
- National Traditional Sports College of Harbin Sports University, Harbin 150008, China.
| | - F Safdarin
- Mechanical Engineering Department, lslamic Azad University, Esfahan, Iran
| | - Sh Baghaei
- Mechanical Engineering Department, lslamic Azad University, Esfahan, Iran
| |
Collapse
|
29
|
Patiño-Martinez E, Nakabo S, Jiang K, Carmona- Rivera C, Tsai WL, Claybaugh D, Yu ZX, Romero A, Bohrnsen E, Schwarz B, Solís-Barbosa MA, Blanco LP, Naqi M, Temesgen-Oyelakim Y, Davis M, Manna Z, Mehta N, Naz F, Brooks S, dell’Orso S, Hasni S, Kaplan MJ. The aconitate decarboxylase 1/itaconate pathway modulates immune dysregulation and associates with cardiovascular disease markers in SLE. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.20.24303097. [PMID: 38605883 PMCID: PMC11007756 DOI: 10.1101/2024.02.20.24303097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Objective The Krebs cycle enzyme Aconitate Decarboxylase 1 (ACOD1) mediates itaconate synthesis in myeloid cells.. Previously, we reported that administration of 4-octyl itaconate abrogated lupus phenotype in mice. Here, we explore the role of the endogenous ACOD1/itaconate pathway in the development of murine lupus as well as their relevance in premature cardiovascular damage in SLE. Methods We characterized Acod1 protein expression in bone marrow-derived macrophages and human monocyte-derived macrophages, following a TLR7 agonist (imiquimod, IMQ). Wild type and Acod1-/- mice were exposed to topical IMQ for 5 weeks to induce an SLE phenotype and immune dysregulation was quantified. Itaconate serum levels were quantified in SLE patients and associated to cardiometabolic parameters and disease activity. Results ACOD1 was induced in mouse bone marrow-derived macrophages (BMDM) and human monocyte-derived macrophages following in vitro TLR7 stimulation. This induction was partially dependent on type I Interferon receptor signaling and specific intracellular pathways. In the IMQ-induced mouse model of lupus, ACOD1 knockout (Acod1-/-) displayed disruptions of the splenic architecture, increased serum anti-dsDNA and proinflammatory cytokine levels, enhanced kidney immune complex deposition and proteinuria, when compared to the IMQ-treated WT mice. Consistent with these results, Acod1-/- BMDM exposed to IMQ showed higher proinflammatory features in vitro. Itaconate levels were decreased in SLE serum compared to healthy control sera, in association with specific perturbed cardiometabolic parameters and subclinical vascular disease. Conclusion These findings suggest that the ACOD1/itaconate pathway plays important immunomodulatory and vasculoprotective roles in SLE, supporting the potential therapeutic role of itaconate analogs in autoimmune diseases.
Collapse
Affiliation(s)
- Eduardo Patiño-Martinez
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Shuichiro Nakabo
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Kan Jiang
- Biodata Mining and Discovery Section, NIAMS/NIH
| | - Carmelo Carmona- Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | | - Dillon Claybaugh
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute (NHLBI), NIH
| | - Aracely Romero
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Eric Bohrnsen
- Protein & Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Hamilton, MT, USA
| | - Benjamin Schwarz
- Protein & Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Hamilton, MT, USA
| | - Miguel A. Solís-Barbosa
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N, 07360 Mexico City, Mexico
| | - Luz P. Blanco
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | | | | | | | | - Nehal Mehta
- National Heart, Lung, and Blood Institute (NHLBI), NIH
| | - Faiza Naz
- Office of Science and Technology, NIAMS/NIH
| | | | | | | | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
30
|
Luo G, Zhou Z, Cao Z, Huang C, Li C, Li X, Deng C, Wu P, Yang Z, Tang J, Qing L. M2 macrophage-derived exosomes induce angiogenesis and increase skin flap survival through HIF1AN/HIF-1α/VEGFA control. Arch Biochem Biophys 2024; 751:109822. [PMID: 38030054 DOI: 10.1016/j.abb.2023.109822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/24/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Skin flap transplantation is a routine strategy in plastic and reconstructive surgery for skin-soft tissue defects. Recent research has shown that M2 macrophages have the potential for pro-angiogenesis during tissue healing. METHODS In our research, we extracted the exosomes from M2 macrophages(M2-exo) and applied the exosomes in the model of skin flap transplantation. The flap survival area was measured, and the choke vessels were assessed by morphological observation. Hematoxylin and eosin (H&E) staining and Immunohistochemistry were applied to assess the neovascularization. The effect of M2-exo on the function of Human umbilical vein endothelial cells (HUVECs) was also investigated. We also administrated 2-methoxyestradiol (2-ME2, an inhibitor of HIF-1α) to explore the underlying mechanism. We tested the effects of M2-Exo on the proliferation of HUVECs through CCK8 assay and EdU staining assay. RESULTS The survival area and number of micro-vessels in the skin flaps were increased in the M2-exo group. Besides, the dilation rate of choke vessels was also enhanced in the M2-exo group. Additionally, compared with the control group, M2-exo could accelerate the proliferation, migration and tube formation of HUVECs in vitro. Furthermore, the expression of the pro-angiogenesis factors, HIF-1α and VEGFA, were overexpressed with the treatment of the M2-exo. The expression of HIF1AN protein level was decreased in the M2-exo group. Finally, treatment with HIF-1α inhibitor reverses the pro-survival effect of M2-exo on skin flaps by interfering with the HIF1AN/HIF-1α/VEGFA signaling pathway. CONCLUSION This study showed that M2-exosomes promote skin flap survival by enhancing angiogenesis, with HIF1AN/HIF-1α/VEGFA playing a crucial role in this process.
Collapse
Affiliation(s)
- Gaojie Luo
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zekun Zhou
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zheming Cao
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chengxiong Huang
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Li
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Li
- Department of Pathology, Changsha Medical University, Changsha, China
| | - Chao Deng
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Panfeng Wu
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenni Yang
- Hunan University of Medicine, Huaihua, China
| | - Juyu Tang
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.
| | - Liming Qing
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|