1
|
Bai H, Arnedo AS, Liu Y, Segura T, Muddiman D. Unraveling the molecular dynamics of wound healing: integrating spatially resolved lipidomics and temporally resolved proteomics. Anal Bioanal Chem 2025; 417:3299-3314. [PMID: 40272507 PMCID: PMC12122605 DOI: 10.1007/s00216-025-05865-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/20/2025] [Accepted: 03/12/2025] [Indexed: 04/25/2025]
Abstract
Understanding the spatial-temporal molecular dynamics of wound healing is crucial for devising effective treatments. Three-dimensional mass spectrometry imaging (3D MSI) enables the comprehensive visualization of molecular distribution throughout skin layers, offering valuable insights into the wound healing process. However, traditional 3D MSI often faces challenges in maintaining data integrity and accurate image registration in the third dimension. To address this, we employed infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI), a hybrid ambient ionization technique capable of sequential imaging through consecutive ablation events for precise 3D image reconstruction. Herein, 3D IR-MALDESI MSI was used to compare the lipidome of fresh-frozen wound samples at three stages of wound healing (inflammation, proliferation, and remodeling) with the healthy skin of SKH- 1 mice. Supplementing this data with a refined LC-MS-based proteomics protocol on selected wound biopsies, our integrated approach deepens our understanding of the molecular intricacies inherent in tissue regeneration.
Collapse
Affiliation(s)
- Hongxia Bai
- Biological Imaging Laboratory for Disease and Exposure Research (BILDER), Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | | | - Yining Liu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - David Muddiman
- Biological Imaging Laboratory for Disease and Exposure Research (BILDER), Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
2
|
Lemties J, Scheidt C, Jung JO, Wirsik NM, Lukomski L, Krauss D, Grabenkamp A, Stier AR, Lyu SI, Damanakis AI, Babic B, Quaas A, Schmidt T, Fuchs HF, Bruns CJ, Schröder W, Schiffmann LM. Vascularity of the gastric conduit predicts complications after Ivor-Lewis esophagectomy. Surg Endosc 2025; 39:3839-3847. [PMID: 40342097 PMCID: PMC12116690 DOI: 10.1007/s00464-025-11780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Anastomotic leakage (AL) contributes to postoperative morbidity and mortality after Ivor-Lewis esophagectomy. Vascular high-risk patients show a significantly increased risk of AL. We previously showed that laparoscopic ischemic conditioning (ISCON) of the stomach prior esophagectomy in these high-risk patients is a safe procedure that induces neoangiogenesis at the anastomotic site. Our data also suggested that this directly impacts on anastomotic healing. To further investigate the hypothesis that gastric conduit vascularization directly influences postoperative morbidity, we evaluated gastric conduit vascularity in a cohort of patients undergoing two-stage esophagectomy prior to the ISCON era. MATERIAL AND METHODS Seventy-nine patients who underwent two-stage esophagectomy from 2016 to 2021 at our center were retrospectively analyzed from a prospectively maintained database. Microvessel density (MVD) of the gastric conduit at the anastomotic region was evaluated by CD34 staining of the gastric stapler ring. Analysis of microvessel density (MVD) was performed using ImageJ. Patients were stratified into low- and high-MVD groups, and MVD was correlated with clinical outcomes. RESULTS Patients with a high MVD showed a significantly lower rate of anastomotic leakage (AL) in comparison to patients with low MVD (6.25% vs. 22.58% p=0.043). Furthermore, a high MVD was associated with a lower rate of major complications (Clavien Dindo ≥ IIIb) (12.50% vs. 38.71% p=0.012) and a shorter hospital stay (17.9 days vs. 23.1 days, p=0.045). CONCLUSION Vascularization of the stomach might function as surgical biomarker of AL in patients undergoing two-stage esophagectomy. Prospective trials have to further substantiate this finding.
Collapse
Affiliation(s)
- Julian Lemties
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Carolin Scheidt
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Jin-On Jung
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Naita M Wirsik
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Leandra Lukomski
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Dolores Krauss
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Anders Grabenkamp
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Alexander R Stier
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Su Ir Lyu
- Institute of Pathology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexander I Damanakis
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Benjamin Babic
- Department of General and Visceral Surgery, Sana Klinikum Offenbach, Offenbach am Main, Germany
| | - Alexander Quaas
- Institute of Pathology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Hans F Fuchs
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Christiane J Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Lars M Schiffmann
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
3
|
Bruschi M, Masini S, Biancucci F, Piersanti G, Canonico B, Menotta M, Magnani M, Fraternale A. Redox modulation via a synthetic thiol compound reshapes energy metabolism in endothelial cells and ameliorates angiogenic expression in a co-culture study with activated macrophages. Biochim Biophys Acta Gen Subj 2025; 1869:130803. [PMID: 40187375 DOI: 10.1016/j.bbagen.2025.130803] [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: 12/19/2024] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The vascular endothelium is the first interface exposed to circulating compounds and oxidative as well as pro-inflammatory stimuli. Nowadays, cysteine pro-drugs are emerging as new and potential therapies in cardiovascular and inflammatory diseases due to their cytoprotective effects. In this study, the effects of redox modulation by a synthetic thiol compound, i.e., I-152, a precursor of N-acetylcysteine (NAC) and cysteamine (MEA), were evaluated after 6 h and 24 h treatment on human umbilical cord endothelial cell (HUVECs) energy metabolism. Following I-152 treatment, higher cysteine and glutathione (GSH) content were detected via HPLC, in concomitance with I-152 derivatives, i.e., NAC and MEA. Untargeted metabolomics confirmed GSH upregulation and NAC presence in addition to I-152 itself and other metabolites, such as dithiol compound (NACMEAA) and triacetylated I-152. Mass spectrometry revealed that I-152 boosted ATP production, specifically through the mitochondrial OXPHOS, as determined via Seahorse assay without inducing oxidative stress. Additionally, I-152 treatment of HUVECs before co-culture with LPS-stimulated macrophages provided GSH and cysteine sustainment and attenuated the transcription of adhesion molecules as well as iNOS expression. Identifying the impact of redox regulation in physiological conditions and the possible metabolic targets could aid the application of novel thiol-based therapeutics.
Collapse
Affiliation(s)
- Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Sofia Masini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Federica Biancucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| |
Collapse
|
4
|
Wang Y, Bai W, Wang X. Progress on three-dimensional visualizing skin architecture with multiple immunofluorescence staining and tissue-clearing approaches. Histol Histopathol 2025; 40:645-651. [PMID: 39390892 DOI: 10.14670/hh-18-815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The skin forms the external covering of the body and is its largest organ, comprising many different cell types. Although the diversity of these cells has been widely studied with various histological methods, our understanding of skin architecture is mainly established on thin tissue sections, which restricted the information available to two dimensions. The development of innovative techniques to induce optical transparency ("clearing") in biological tissues has enabled researchers to visualize the three-dimensional reconstruction of intact organs and thick tissue sections at a cellular resolution. With the aid of tissue-clearing treatment, the labeled cutaneous nerve fibers and blood vessels can be followed for a longer distance on the thicker skin section or the whole mount skin under a fluorescence microscopy or a confocal microscopy. It is beneficial for demonstrating the morphological characteristics of nerve fibers and blood vessels themselves, as well as their spatial interconnection. In this review, we provide a brief summary of the literature on the use of tissue optical clearing methods and describe our experience of multiple fluorescent staining and tissue clearing approaches on thicker skin sections and whole-mount skin in our laboratory. Given the existing conventional methods, we expected to provide a more effective approach to comprehensively study skin architecture.
Collapse
Affiliation(s)
- Yuqing Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wanzhu Bai
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoyu Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Kuang X, Liang Z, Xia Y, Shan M, Hao Y, Liu H, Wang Z, He Q, Xia C, Feng C, Chang G, Wang Y. Hydrogen-Rich Saline Combined With Vacuum Sealing Drainage Promotes Wound Healing by Altering Biotin Metabolism. J Cell Mol Med 2025; 29:e70292. [PMID: 39804806 PMCID: PMC11728484 DOI: 10.1111/jcmm.70292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
Impaired wound healing affects the life quality of patients and causes a substantial financial burden. Hydrogen-rich medium is reported to have antioxidant and anti-inflammatory effects. However, the role of hydrogen-rich saline (HRS) in cutaneous wound healing remains largely unexplored, especially by metabolomics. Thus, untargeted metabolomics profiling was analysed to study the effects and mechanism of HRS combined with vacuum sealing drainage (VSD) in a rabbit full-thickness wound model. Our results indicated that the combination treatment of HRS and VSD could accelerate wound healing. In vitro experiments further confirmed its effects on HaCaT keratinocytes. We found that 45 metabolites were significantly changed between the VSD + HRS group and the VSD + saline-treated group. Pathway enrichment analysis indicated that biotin metabolism was the potential target pathway. The biochemical interpretation analysis demonstrated that combining HRS and VSD might enhance mitochondrial function, ATP synthesis, and GSH homeostasis by altering biotin metabolism. The detection of representative indicators of oxidative stress supported the critical metabolic pathway analysis as well. In summary, VSD combined with HRS might provide a new strategy to enhance wound healing.
Collapse
Affiliation(s)
- Xinwen Kuang
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Zhengyun Liang
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
- Department of Dermatology, Shenzhen Center for Chronic Disease ControlShenzhen Institute of DermatologyShenzhenChina
| | - Yijun Xia
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Mengjie Shan
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yan Hao
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Hao Liu
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Zhi Wang
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Qianjun He
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Chao Xia
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Cheng Feng
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Guojing Chang
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
6
|
da Veiga Moreira J, Schwartz L, Jolicoeur M. Singlet Oxygen-Induced Mitochondrial Reset in Cancer: A Novel Approach for Ovarian Cancer Therapy. Metabolites 2024; 14:648. [PMID: 39728429 DOI: 10.3390/metabo14120648] [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: 10/31/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: This study explores the generation of singlet oxygen (SO) through methylene blue (MB) activation as a metabolic intervention for ovarian cancer. We aimed to examine the role of SO in modulating mitochondrial function, cellular metabolism, and proliferation in ovarian cancer cell lines compared to control cells. Methods: The study utilized two ovarian cancer cell lines, OV1369-R2 and TOV1369, along with ARPE-19 control cells. Following MB treatment and light activation, mitochondrial function and ATP synthesis were assessed. Metabolomic analyses were performed to evaluate changes in central carbon metabolism, particularly focusing on markers of the Warburg effect. Results: TOV1369 cells exhibited a pronounced sensitivity to MB treatment, resulting in significant inhibition of ATP synthesis and reduced proliferation. Metabolomic analysis indicated that MB-induced SO production partially reversed the Warburg effect, suggesting a shift from glycolysis to oxidative phosphorylation. These effects were less pronounced in OV1369-R2 and ARPE-19 cells, correlating with their lower MB sensitivity. Conclusions: MB-generated SO selectively modulates mitochondrial energetics in ovarian cancer cells, driving a metabolic reorganization that curtails their proliferative capacity. This approach, leveraging the bacterial-like features of cancer metabolism, offers a promising therapeutic avenue to induce apoptosis and enhance treatment outcomes in ovarian cancer.
Collapse
Affiliation(s)
- Jorgelindo da Veiga Moreira
- Research Laboratory in Applied Metabolic Engineering, Department of Chemical Engineering, Polytechnique Montréal, Centre-Ville Station, P.O. Box 6079, Montréal, QC H3C 3A7, Canada
| | - Laurent Schwartz
- Assistance Publique des Hôpitaux de Paris, Avenue Victoria, 75003 Paris, France
| | - Mario Jolicoeur
- Research Laboratory in Applied Metabolic Engineering, Department of Chemical Engineering, Polytechnique Montréal, Centre-Ville Station, P.O. Box 6079, Montréal, QC H3C 3A7, Canada
| |
Collapse
|
7
|
Kennedy S, Williams C, Tsaturian E, Morgan JT. Dexamethasone Impairs ATP Production and Mitochondrial Performance in Human Trabecular Meshwork Cells. Curr Issues Mol Biol 2024; 46:9867-9880. [PMID: 39329939 PMCID: PMC11430611 DOI: 10.3390/cimb46090587] [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: 08/05/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Mitochondrial damage occurs in human trabecular meshwork (HTM) cells as a result of normal aging and in open angle glaucoma. Using an HTM cell model, we quantified mitochondrial function and ATP generation rates after dexamethasone (Dex) and TGF-β2 treatments, frequently used as in vitro models of glaucoma. Primary HTM cells were assayed for metabolic function using a Seahorse XFp Analyzer. We additionally assessed the mitochondrial copy number and the expression of transcripts associated with mitochondrial biogenesis and oxidative stress regulation. Cells treated with Dex, but not TGF-β2, exhibited a significant decrease in total ATP production and ATP from oxidative phosphorylation relative to that of the control. Dex treatment also resulted in significant decreases in maximal respiration, ATP-linked O2 consumption, and non-mitochondrial O2 consumption. We did not observe significant changes in the level of mitochondrial genomes or mRNA transcripts of genes involved in mitochondrial biogenesis and oxidative stress regulation. Decreased mitochondrial performance and ATP production are consistent with the results of prior studies identifying the effects of Dex on multiple cell types, including HTM cells. Our results are also consistent with in vivo evidence of mitochondrial damage in open-angle glaucoma. Overall, these results demonstrate a decrease in mitochondrial performance in Dex-induced glaucomatous models in vitro, meriting further investigation.
Collapse
Affiliation(s)
- Shane Kennedy
- Department of Molecular, Cell and Systems Biology, University of California-Riverside, Riverside, CA 92521, USA
| | - Clayton Williams
- Department of Bioengineering, University of California-Riverside, Riverside, CA 92521, USA
| | - Emily Tsaturian
- Department of Bioengineering, University of California-Riverside, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California-Riverside, Riverside, CA 92521, USA
| |
Collapse
|
8
|
Zhang Q, Xia Y, Wang L, Wang Y, Bao Y, Zhao GS. Targeted anti-angiogenesis therapy for advanced osteosarcoma. Front Oncol 2024; 14:1413213. [PMID: 39252946 PMCID: PMC11381227 DOI: 10.3389/fonc.2024.1413213] [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: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
To date, despite extensive research, the prognosis of advanced osteosarcoma has not improved significantly. Thus, patients experience a reduced survival rate, suggesting that a reevaluation of current treatment strategies is required. Recently, in addition to routine surgery, chemotherapy and radiotherapy, researchers have explored more effective and safer treatments, including targeted therapy, immunotherapy, anti-angiogenesis therapy, metabolic targets therapy, and nanomedicine therapy. The tumorigenesis and development of osteosarcoma is closely related to angiogenesis. Thus, anti-angiogenesis therapy is crucial to treat osteosarcoma; however, recent clinical trials found that it has insufficient efficacy. To solve this problem, the causes of treatment failure and improve treatment strategies should be investigated. This review focuses on summarizing the pathophysiological mechanisms of angiogenesis in osteosarcoma and recent advances in anti-angiogenesis treatment of osteosarcoma. We also discuss some clinical studies, with the aim of providing new ideas to improve treatment strategies for osteosarcoma and the prognosis of patients.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Pain and Rehabilitation, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuxuan Xia
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - LiYuan Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Sheng Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Zheng Y, Ren X, Qi X, Song R, Zhao C, Ma J, Li X, Deng Q, He Y, Kong L, Qian L, Zhang F, Li M, Sun M, Liu W, Liu H, She G. Bao Yuan decoction alleviates fatigue by restraining inflammation and oxidative stress via the AMPK/CRY2/PER1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118058. [PMID: 38513778 DOI: 10.1016/j.jep.2024.118058] [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: 01/22/2024] [Revised: 02/28/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baoyuan Decoction (BYD) was initially recorded in the classic of "Bo Ai Xin Jian" in the Ming dynasty. It is traditionally used for treating weakness and cowardice, and deficiency of vital energy. In researches related to anti-fatigue effects, the reciprocal regulation of AMPK and circadian clocks likely plays an important role in anti-fatigue mechanism, while it has not yet been revealed. Therefore, we elucidated the anti-fatigue mechanism of BYD through AMPK/CRY2/PER1 pathway. AIM OF THE STUDY To investigate the effect and mechanism of BYD in reducing fatigue, using pharmacodynamics, network pharmacology and transcriptomics through the AMPK/CRY2/PER1 signaling pathway. MATERIALS AND METHODS Firstly, the chemical constituents of BYD were qualitatively identified by UHPLC-Q-Exactive Orbitrap/MS, establishing a comprehensive strategy with an in-house library, Xcalibur software and Pubchem combined. Secondly, a Na2SO3-induced fatigue model and 2,2'-Azobis (2-methylpropionamidine) dihydrochloride (AAPH)-induced oxidative stress model were developed to evaluate the anti-fatigue and anti-oxidant activities of BYD using AB zebrafish. The anti-inflammatory activity of BYD was evaluated using CuSO4-induced and tail cutting-induced Tg (lyz: dsRed) transgenic zebrafish inflammation models. Then, target screening was performed by Swiss ADME, GeneCards, OMIM and DrugBank databases, the network was constructed using Cytoscape 3.9.0. Transcriptome and network pharmacology technology were used to investigate the related signaling pathways and potential mechanisms after treatment with BYD, which were verified by real-time quantitative PCR (RT-qPCR). RESULTS In total, 114 compounds from the water extract of BYD were identified as major compounds. Na₂SO₃-induced fatigue model and AAPH-induced oxidative stress model indicated that BYD has significant anti-fatigue and antioxidant effects. Meanwhile, BYD showed significant anti-inflammatory effects on CuSO4-induced and tail cutting-induced zebrafish inflammation models. The KEGG result of network pharmacology showed that the anti-fatigue function of BYD was mainly effected through AMPK signaling pathway. Besides, transcriptome analysis indicated that the circadian rhythm, AMPK and IL-17 signaling pathways were recommended as the main pathways related to the anti-fatigue effect of BYD. The RT-qPCR results showed that compared with a model control group, the treatment of BYD significantly elevated the expression mRNA of AMPK, CRY2 and PER1. CONCLUSION Herein, we identified 114 chemical constituents of BYD, performed zebrafish activity validation, while demonstrated that BYD can relieve fatigue by AMPK/CRY2/PER1 signaling pathway through network pharmacology and transcriptome.
Collapse
Affiliation(s)
- Yuan Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xueyang Ren
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiaodan Qi
- Dong'e Ejiao Co., Ltd., Liaocheng, 252200, China; Shandong Key Laboratory of Gelatine TCM Research and Development, Liaocheng, 252200, China; Shandong Technology Innovation Center of Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China; National Engineering Technology Research Center for Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China
| | - Ruolan Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Chongjun Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jiamu Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xianxian Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qingyue Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yingyu He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Lingmei Kong
- Dong'e Ejiao Co., Ltd., Liaocheng, 252200, China; Shandong Key Laboratory of Gelatine TCM Research and Development, Liaocheng, 252200, China; Shandong Technology Innovation Center of Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China; National Engineering Technology Research Center for Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China
| | - Liyan Qian
- Dong'e Ejiao Co., Ltd., Liaocheng, 252200, China; Shandong Key Laboratory of Gelatine TCM Research and Development, Liaocheng, 252200, China; Shandong Technology Innovation Center of Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China; National Engineering Technology Research Center for Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China
| | - Feng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mingxia Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mengyu Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wei Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haibin Liu
- Dong'e Ejiao Co., Ltd., Liaocheng, 252200, China; Shandong Key Laboratory of Gelatine TCM Research and Development, Liaocheng, 252200, China; Shandong Technology Innovation Center of Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China; National Engineering Technology Research Center for Gelatin-based Traditional Chinese Medicine, Liaocheng, 252200, China.
| | - Gaimei She
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
10
|
Lin W, Ye C, Sun L, Chen Z, Qu C, Zhu M, Li J, Kong R, Xu Z. A novel mitochondrial metabolism-related gene signature for predicting the prognosis of oesophageal squamous cell carcinoma. Aging (Albany NY) 2024; 16:9649-9679. [PMID: 38843392 PMCID: PMC11210263 DOI: 10.18632/aging.205892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/03/2024] [Indexed: 06/11/2024]
Abstract
Oesophageal squamous cell carcinoma (ESCC) is one of the most lethal cancers worldwide. Due to the important role of mitochondrial metabolism in cancer progression, a clinical prognostic model based on mitochondrial metabolism and clinical features was constructed in this study to predict the prognosis of ESCC. Firstly, the mitochondrial metabolism scores (MMs) were calculated based on 152 mitochondrial metabolism-related genes (MMRGs) by single sample gene set enrichment analysis (ssGSEA). Subsequently, univariate Cox regression and LASSO algorithm were used to identify prognosis-associated MMRG and risk-stratify patients. Functional enrichment, interaction network and immune-related analyses were performed to explore the features differences in patients at different risks. Finally, a prognostic nomogram incorporating clinical factors was constructed to assess the prognosis of ESCC. Our results found there were differences in clinical features between the MMs-high group and the MMs-low group in the TCGA-ESCC dataset (P<0.05). Afterwards, we identified 6 MMRGs (COX10, ACADVL, IDH3B, AKR1A1, LIAS, and NDUFB8) signature that could accurately distinguish high-risk and low-risk ESCC patients. A predictive nomogram that combined the 6 MMRGs with sex and N stage to predict the prognosis of ESCC was constructed, and the areas under the receiver operating characteristic (ROC) curve at 1, 2 and 3 years were 0.948, 0.927 and 0.848, respectively. Finally, we found that COX10, one of 6 MMRGs, could inhibit the malignant progression of ESCC in vitro. In summary, we constructed a clinical prognosis model based on 6 MMRGs and clinical features which can accurately predict the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Wenhao Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, Shaanxi, China
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Changchun Ye
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Liangzhang Sun
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, Shaanxi, China
| | - Zilu Chen
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Chao Qu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Minxia Zhu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, Shaanxi, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jianzhong Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, Shaanxi, China
| | - Ranran Kong
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, Shaanxi, China
| | - Zhengshui Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, Shaanxi, China
- Key Laboratory of Surgery Critical Care and Life Support (Xi’an Jiaotong University), Ministry of Education, Xi’an 710061, Shaanxi, China
| |
Collapse
|
11
|
Raz D, Ben-Yaakov K, Levi M, Bertolin M, Ferrari S, Ponzin D, Busin M, Leiba H, Marcovich AL, Eisenberg-Lerner A, Rotfogel Z. Mitochondria Transplantation Promotes Corneal Epithelial Wound Healing. Invest Ophthalmol Vis Sci 2024; 65:14. [PMID: 38848077 PMCID: PMC11166225 DOI: 10.1167/iovs.65.6.14] [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: 07/23/2023] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
Purpose The integrity of the corneal epithelium is essential in maintaining normal corneal function. Conditions disrupting the corneal epithelial layer range from chemical burns to dry eye disease and may result in impairment of both corneal transparency and sensation. Identifying factors that regulate corneal wound healing is key for the development of new treatment strategies. Here, we investigated a direct role of mitochondria in corneal wound healing via mitochondria transplantation. Methods Human corneal epithelial cells (hCECs) were isolated from human corneas and incubated with mitochondria which were isolated from human ARPE-19 cells. We determined the effect of mitochondria transplantation on wound healing and proliferation of hCECs. In vivo, we used a mouse model of corneal chemical injury. Mitochondria were isolated from mouse livers and topically applied to the ocular surface following injury. We evaluated the time of wound repair, corneal re-epithelization, and stromal abnormalities. Results Mitochondria transplantation induced the proliferation and wound healing of primary hCECs. Further, mitochondria transplantation promoted wound healing in vivo. Specifically, mice receiving mitochondria recovered twice as fast as control mice following corneal injury, presenting both enhanced and improved repair. Corneas treated with mitochondria demonstrated the re-epithelization of the wound area to a multi-layer appearance, compared to thinning and complete loss of the epithelium in control mice. Mitochondria transplantation also prevented the thickening and disorganization of the corneal stromal lamella, restoring normal corneal dehydration. Conclusions Mitochondria promote corneal re-epithelization and wound healing. Augmentation of mitochondria levels via mitochondria transplantation may serve as an effective treatment for inducing the rapid repair of corneal epithelial defects.
Collapse
Affiliation(s)
- Daniel Raz
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Keren Ben-Yaakov
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
| | - Michal Levi
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
- Department of Ophthalmology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Diego Ponzin
- Fondazione Banca degli Occhi del Veneto, Venice, Italy
| | - Massimo Busin
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Department of Ophthalmology, Ospedali Privati Forlì “Villa Igea,” Forlì, Italy
- Istituto Internazionale per la Ricerca e Formazione in Oftalmologia, Forlì, Italy
| | - Hana Leiba
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
- Department of Ophthalmology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Arie L. Marcovich
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
- Department of Ophthalmology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Avital Eisenberg-Lerner
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
| | - Ziv Rotfogel
- Ophthalmology Research Laboratory, Department of Ophthalmology, Kaplan Medical Center, Israel
- Department of Ophthalmology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| |
Collapse
|
12
|
Ichegiri A, Kodolikar K, Bagade V, Selukar M, Dey T. Mitochondria: A source of potential biomarkers for non-communicable diseases. Adv Clin Chem 2024; 121:334-365. [PMID: 38797544 DOI: 10.1016/bs.acc.2024.04.007] [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: 05/29/2024]
Abstract
Mitochondria, as an endosymbiont of eukaryotic cells, controls multiple cellular activities, including respiration, reactive oxygen species production, fatty acid synthesis, and death. Though the majority of functional mitochondrial proteins are translated through a nucleus-controlled process, very few of them (∼10%) are translated within mitochondria through their own machinery. Germline and somatic mutations in mitochondrial and nuclear DNA significantly impact mitochondrial homeostasis and function. Such modifications disturbing mitochondrial biogenesis, metabolism, or mitophagy eventually resulted in cellular pathophysiology. In this chapter, we discussed the impact of mitochondria and its dysfunction on several non-communicable diseases like cancer, diabetes, neurodegenerative, and cardiovascular problems. Mitochondrial dysfunction and its outcome could be screened by currently available omics-based techniques, flow cytometry, and high-resolution imaging. Such characterization could be evaluated as potential biomarkers to assess the disease burden and prognosis.
Collapse
Affiliation(s)
- Amulya Ichegiri
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Kshitij Kodolikar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Vaibhavi Bagade
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Mrunal Selukar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Tuli Dey
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India.
| |
Collapse
|
13
|
Bansal R, Torres M, Hunt M, Wang N, Chatzopoulou M, Manchanda M, Taddeo EP, Shu C, Shirihai OS, Bachar-Wikstrom E, Wikstrom JD. Role of the mitochondrial protein cyclophilin D in skin wound healing and collagen secretion. JCI Insight 2024; 9:e169213. [PMID: 38564292 PMCID: PMC11141914 DOI: 10.1172/jci.insight.169213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Central for wound healing is the formation of granulation tissue, which largely consists of collagen and whose importance stretches past wound healing, including being implicated in both fibrosis and skin aging. Cyclophilin D (CyD) is a mitochondrial protein that regulates the permeability transition pore, known for its role in apoptosis and ischemia-reperfusion. To date, the role of CyD in human wound healing and collagen generation has been largely unexplored. Here, we show that CyD was upregulated in normal wounds and venous ulcers, likely adaptive as CyD inhibition impaired reepithelialization, granulation tissue formation, and wound closure in both human and pig models. Overexpression of CyD increased keratinocyte migration and fibroblast proliferation, while its inhibition reduced migration. Independent of wound healing, CyD inhibition in fibroblasts reduced collagen secretion and caused endoplasmic reticulum collagen accumulation, while its overexpression increased collagen secretion. This was confirmed in a Ppif-KO mouse model, which showed a reduction in skin collagen. Overall, this study revealed previously unreported roles of CyD in skin, with implications for wound healing and beyond.
Collapse
Affiliation(s)
- Ritu Bansal
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Nuoqi Wang
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Margarita Chatzopoulou
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Mansi Manchanda
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Evan P. Taddeo
- Metabolism Theme
- Department of Molecular and Medical Pharmacology, and
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Cynthia Shu
- Metabolism Theme
- Department of Molecular and Medical Pharmacology, and
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Orian S. Shirihai
- Metabolism Theme
- Department of Molecular and Medical Pharmacology, and
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D. Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Dunaway LS, Luse MA, Nyshadham S, Bulut G, Alencar GF, Chavkin NW, Cortese-Krott M, Hirschi KK, Isakson BE. Obesogenic diet disrupts tissue-specific mitochondrial gene signatures in the artery and capillary endothelium. Physiol Genomics 2024; 56:113-127. [PMID: 37982169 PMCID: PMC11281809 DOI: 10.1152/physiolgenomics.00109.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Endothelial cells (ECs) adapt to the unique needs of their resident tissue and metabolic perturbations, such as obesity. We sought to understand how obesity affects EC metabolic phenotypes, specifically mitochondrial gene expression. We investigated the mesenteric and adipose endothelium because these vascular beds have distinct roles in lipid homeostasis. Initially, we performed bulk RNA sequencing on ECs from mouse adipose and mesenteric vasculatures after a normal chow (NC) diet or high-fat diet (HFD) and found higher mitochondrial gene expression in adipose ECs compared with mesenteric ECs in both NC and HFD mice. Next, we performed single-cell RNA sequencing and categorized ECs as arterial, capillary, venous, or lymphatic. We found mitochondrial genes to be enriched in adipose compared with mesentery under NC conditions in artery and capillary ECs. After HFD, these genes were decreased in adipose ECs, becoming like mesenteric ECs. Transcription factor analysis revealed that peroxisome proliferator-activated receptor-γ (PPAR-γ) had high specificity in NC adipose artery and capillary ECs. These findings were recapitulated in single-nuclei RNA-sequencing data from human visceral adipose. The sum of these findings suggests that mesenteric and adipose arterial ECs metabolize lipids differently, and the transcriptional phenotype of the vascular beds converges in obesity due to downregulation of PPAR-γ in adipose artery and capillary ECs.NEW & NOTEWORTHY Using bulk and single-cell RNA sequencing on endothelial cells from adipose and mesentery, we found that an obesogenic diet induces a reduction in adipose endothelial oxidative phosphorylation gene expression, resulting in a phenotypic convergence of mesenteric and adipose endothelial cells. Furthermore, we found evidence that PPAR-γ drives this phenotypic shift. Mining of human data sets segregated based on body mass index supported these findings. These data point to novel mechanisms by which obesity induces endothelial dysfunction.
Collapse
Affiliation(s)
- Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Melissa A Luse
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Shruthi Nyshadham
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Gamze Bulut
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Gabriel F Alencar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Nicholas W Chavkin
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Miriam Cortese-Krott
- Department of Cardiology, Pneumology and Angiology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Karen K Hirschi
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| |
Collapse
|
15
|
Guo H, Li P, Zhao J, Xin Q, Miao Y, Li L, Li X, Wang S, Mo H, Zeng L, Ju Z, Liu Z, Shen X, Cong W. Sheng Mai Yin shows anti-fatigue, anti-hypoxia and cardioprotective potential in an experimental joint model of fatigue and acute myocardial infarction. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117338. [PMID: 37890804 DOI: 10.1016/j.jep.2023.117338] [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: 09/11/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular disease (CVD) and fatigue are two common diseases endangering human life and health that may interact and reinforce one another. Myocardial infarction survivors frequently experience fatigue, and acute myocardial infarction (AMI) is one of the most common cardiovascular diseases that cause fatigue-induced sudden death. Sheng Mai Yin (SMY), a Chinese medicine prescription, is traditionally used for the treatment of diabetes and cardiovascular disease, and has been demonstrated to reduce fatigue and safeguard cardiac function. AIM OF THE STUDY This study aims to investigate the effects and underlying mechanisms of SMY in treating fatigue and AMI. MATERIALS AND METHODS The pharmacological mechanisms of SMY in treating fatigue and AMI were predicted by bioinformatics and network pharmacology methods. After administering SMY at high, medium and low doses, the swimming time to exhaustion, hemoglobin level, serological parameters and hypoxia tolerance time were detected in C57BL/6N mice, and the left ventricular ejection fractions (LVEF), left ventricular fractional shortening (LVFS), grasp strength, cardiac histopathology, serological parameters and the expression of PINK1 and Parkin proteins were examined in Wistar rats. RESULTS 371 core targets for SMY and 282 disease targets for fatigue and AMI were obtained using bioinformatics and network pharmacology methods. Enrichment analysis of target genes revealed that SMY might interfere with fatigue and AMI through biological processes such as mitochondrial autophagy, apoptosis, and oxidative stress. For in vivo experiments, SMY showed significant anti-fatigue and hypoxia tolerance effects in mice; It also improved the cardiac function and grasp strength, decreased their cardiac index, myocardial injury and fibrosis degree, and induced serological parameters levels and the expression of PTEN-induced putative kinase 1 (PINK1) and Parkin proteins in myocardium, suggesting that SMY may exert cardioprotective effects in a joint rat model of fatigue and AMI by inhibiting excessive mitochondrial autophagy. CONCLUSION This study revealed the anti-fatigue, anti-hypoxia and cardioprotective effects of SMY in a joint model of fatigue-AMI, and the pharmacological mechanism may be related to the inhibition of mitochondrial autophagy in cardiomyocytes through the PINK1/Parkin pathway. The discoveries may provide new ideas for the mechanism study of traditional Chinese medicine, especially complex prescriptions, in treating fatigue and AMI.
Collapse
Affiliation(s)
- Hao Guo
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China
| | - Jun Zhao
- Traditional Chinese Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China
| | - Li Li
- Chenland Research Institute, Irvine, CA, 92614, USA
| | - Xin Li
- Chenland Research Institute, Irvine, CA, 92614, USA
| | | | - Hui Mo
- Macao Health Bureau, Macao, 999078, China
| | - Li Zeng
- Macau University of Science and Technology, Macao, 999078, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Zimin Liu
- Chenland Research Institute, Irvine, CA, 92614, USA.
| | - Xiaoxu Shen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100091, China.
| | - Weihong Cong
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China.
| |
Collapse
|
16
|
da Veiga Moreira J, Nleme N, Schwartz L, Leclerc-Desaulniers K, Carmona E, Mes-Masson AM, Jolicoeur M. Methylene Blue Metabolic Therapy Restrains In Vivo Ovarian Tumor Growth. Cancers (Basel) 2024; 16:355. [PMID: 38254843 PMCID: PMC10814748 DOI: 10.3390/cancers16020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Ovarian cancer remains a significant challenge, especially in platinum-resistant cases where treatment options are limited. In this study, we investigated the potential of methylene blue (MB) as a metabolic therapy and complementary treatment approach for ovarian cancer. Our findings demonstrated a significant in vivo reduction in the proliferation of TOV112D-based ovarian-cell-line xenografts. In this preclinical study, which used a carboplatin-resistant ovarian cancer tumor model implanted into mice, MB-mediated metabolic therapy exhibited superior tumor slowdown compared to carboplatin treatment alone. This indicates, for the first time, MB's potential as an alternative or adjuvant treatment, especially for resistant cases. Our in vitro study on TOV112D and ARPE-19 sheds light on the impact of such an MB-based metabolic therapy on mitochondrial energetics (respiration and membrane potential). MB showed a modulatory role in the oxygen consumption rate and the mitochondrial membrane potential. These results revealed, for the first time, that MB specifically targets TOV112D mitochondria and probably induces cell apoptosis. The differential response of normal (ARPE-19) and cancer (TOV112D) cells to the MB treatment suggests potential alterations in cancer cell mitochondria, opening avenues for therapeutic approaches that target the mitochondria. Overall, our findings suggest the efficacy of MB as a possible treatment for ovarian cancer and provide valuable insights into the mechanisms underlying the efficacy of methylene blue metabolic therapy in ovarian cancer treatment.
Collapse
Affiliation(s)
- Jorgelindo da Veiga Moreira
- Research Laboratory in Applied Metabolic Engineering, Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| | - Nancy Nleme
- Research Laboratory in Applied Metabolic Engineering, Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| | | | - Kim Leclerc-Desaulniers
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada (A.-M.M.-M.)
| | - Euridice Carmona
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada (A.-M.M.-M.)
| | - Anne-Marie Mes-Masson
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada (A.-M.M.-M.)
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Mario Jolicoeur
- Research Laboratory in Applied Metabolic Engineering, Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
17
|
Schorn F, Werthenbach JP, Hoffmann M, Daoud M, Stachelscheid J, Schiffmann LM, Hildebrandt X, Lyu SI, Peltzer N, Quaas A, Vucic D, Silke J, Pasparakis M, Kashkar H. cIAPs control RIPK1 kinase activity-dependent and -independent cell death and tissue inflammation. EMBO J 2023; 42:e113614. [PMID: 37789765 PMCID: PMC10646551 DOI: 10.15252/embj.2023113614] [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: 01/27/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Cellular inhibitor of apoptosis proteins (cIAPs) are RING-containing E3 ubiquitin ligases that ubiquitylate receptor-interacting protein kinase 1 (RIPK1) to regulate TNF signalling. Here, we established mice simultaneously expressing enzymatically inactive cIAP1/2 variants, bearing mutations in the RING domains of cIAP1/2 (cIAP1/2 mutant RING, cIAP1/2MutR ). cIap1/2MutR/MutR mice died during embryonic development due to RIPK1-mediated apoptosis. While expression of kinase-inactive RIPK1D138N rescued embryonic development, Ripk1D138N/D138N /cIap1/2MutR/MutR mice developed systemic inflammation and died postweaning. Cells expressing cIAP1/2MutR and RIPK1D138N were still susceptible to TNF-induced apoptosis and necroptosis, implying additional kinase-independent RIPK1 activities in regulating TNF signalling. Although further ablation of Ripk3 did not lead to any phenotypic improvement, Tnfr1 gene knock-out prevented early onset of systemic inflammation and premature mortality, indicating that cIAPs control TNFR1-mediated toxicity independent of RIPK1 and RIPK3. Beyond providing novel molecular insights into TNF-signalling, the mouse model established in this study can serve as a useful tool to further evaluate ongoing therapeutic protocols using inhibitors of TNF, cIAPs and RIPK1.
Collapse
Affiliation(s)
- Fabian Schorn
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - J Paul Werthenbach
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Mattes Hoffmann
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Mila Daoud
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Johanna Stachelscheid
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Lars M Schiffmann
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral, Cancer and Transplantation SurgeryUniversity of CologneCologneGermany
| | - Ximena Hildebrandt
- Faculty of Medicine and University Hospital of Cologne, Department of Translational GenomicsUniversity of CologneCologneGermany
| | - Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology and Center for Integrated Oncology (CIO) Cologne BonnUniversity of CologneCologneGermany
| | - Nieves Peltzer
- Faculty of Medicine and University Hospital of Cologne, Department of Translational GenomicsUniversity of CologneCologneGermany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology and Center for Integrated Oncology (CIO) Cologne BonnUniversity of CologneCologneGermany
| | - Domagoj Vucic
- Department of Immunology DiscoveryGenentechSouth San FranciscoCAUSA
| | - John Silke
- The Walter and Eliza Hall Institute for Medical ResearchMelbourneVic.Australia
| | - Manolis Pasparakis
- Institute for GeneticsUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| | - Hamid Kashkar
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| |
Collapse
|
18
|
Do MH, Shi W, Ji L, Ladewig E, Zhang X, Srivastava RM, Capistrano KJ, Edwards C, Malik I, Nixon BG, Stamatiades EG, Liu M, Li S, Li P, Chou C, Xu K, Hsu TW, Wang X, Chan TA, Leslie CS, Li MO. Reprogramming tumor-associated macrophages to outcompete endovascular endothelial progenitor cells and suppress tumor neoangiogenesis. Immunity 2023; 56:2555-2569.e5. [PMID: 37967531 PMCID: PMC11284818 DOI: 10.1016/j.immuni.2023.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/03/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023]
Abstract
Tumors develop by invoking a supportive environment characterized by aberrant angiogenesis and infiltration of tumor-associated macrophages (TAMs). In a transgenic model of breast cancer, we found that TAMs localized to the tumor parenchyma and were smaller than mammary tissue macrophages. TAMs had low activity of the metabolic regulator mammalian/mechanistic target of rapamycin complex 1 (mTORC1), and depletion of negative regulator of mTORC1 signaling, tuberous sclerosis complex 1 (TSC1), in TAMs inhibited tumor growth in a manner independent of adaptive lymphocytes. Whereas wild-type TAMs exhibited inflammatory and angiogenic gene expression profiles, TSC1-deficient TAMs had a pro-resolving phenotype. TSC1-deficient TAMs relocated to a perivascular niche, depleted protein C receptor (PROCR)-expressing endovascular endothelial progenitor cells, and rectified the hyperpermeable blood vasculature, causing tumor tissue hypoxia and cancer cell death. TSC1-deficient TAMs were metabolically active and effectively eliminated PROCR-expressing endothelial cells in cell competition experiments. Thus, TAMs exhibit a TSC1-dependent mTORC1-low state, and increasing mTORC1 signaling promotes a pro-resolving state that suppresses tumor growth, defining an innate immune tumor suppression pathway that may be exploited for cancer immunotherapy.
Collapse
Affiliation(s)
- Mytrang H Do
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Wei Shi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Liangliang Ji
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Erik Ladewig
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xian Zhang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raghvendra M Srivastava
- Immunogenomics & Precision Oncology Platform (IPOP), Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kristelle J Capistrano
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chaucie Edwards
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isha Malik
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Briana G Nixon
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Efstathios G Stamatiades
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming Liu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shun Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Peng Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chun Chou
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ke Xu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Ting-Wei Hsu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Xinxin Wang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Timothy A Chan
- Immunogenomics & Precision Oncology Platform (IPOP), Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming O Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
19
|
Wu M, Zhao Y, Tao M, Fu M, Wang Y, Liu Q, Lu Z, Guo J. Malate-Based Biodegradable Scaffolds Activate Cellular Energetic Metabolism for Accelerated Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50836-50853. [PMID: 37903387 DOI: 10.1021/acsami.3c09394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
The latest advancements in cellular bioenergetics have revealed the potential of transferring chemical energy to biological energy for therapeutic applications. Despite efforts, a three-dimensional (3D) scaffold that can induce long-term bioenergetic effects and facilitate tissue regeneration remains a big challenge. Herein, the cellular energetic metabolism promotion ability of l-malate, an important intermediate of the tricarboxylic acid (TCA) cycle, was proved, and a series of bioenergetic porous scaffolds were fabricated by synthesizing poly(diol l-malate) (PDoM) prepolymers via a facial one-pot polycondensation of l-malic acid and aliphatic diols, followed by scaffold fabrication and thermal-cross-linking. The degradation products of the developed PDoM scaffolds can regulate the metabolic microenvironment by entering mitochondria and participating in the TCA cycle to elevate intracellular adenosine triphosphate (ATP) levels, thus promoting the cellular biosynthesis, including the production of collagen type I (Col1a1), fibronectin 1 (Fn1), and actin alpha 2 (Acta2/α-Sma). The porous PDoM scaffold was demonstrated to support the growth of the cocultured mesenchymal stem cells (MSCs) and promote their secretion of bioactive molecules [such as vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1), and basic fibroblast growth factor (bFGF)], and this stem cells-laden scaffold architecture was proved to accelerate wound healing in a critical full-thickness skin defect model on rats.
Collapse
Affiliation(s)
- Min Wu
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yitao Zhao
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Meihan Tao
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Meimei Fu
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yue Wang
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Qi Liu
- Regenerative Medicine and Tissue Repair Research Center, Huangpu Institute of Materials, Guangzhou 511363, P. R. China
| | - Zhihui Lu
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Regenerative Medicine and Tissue Repair Research Center, Huangpu Institute of Materials, Guangzhou 511363, P. R. China
| | - Jinshan Guo
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
20
|
Guo F, Mao S, Long Y, Zhou B, Gao L, Huang H. The Influences of Perinatal Androgenic Exposure on Cardiovascular and Metabolic Disease of Offspring of PCOS. Reprod Sci 2023; 30:3179-3189. [PMID: 37380913 DOI: 10.1007/s43032-023-01286-w] [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: 01/11/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
Hyperandrogenism is an endocrine disorder affecting a large population of reproductive-aged women, thus proportionally high number of fetuses are subjected to prenatal androgenic exposure (PNA). The short-term stimulations at critical ontogenetic stages can wield lasting influences on the health. The most commonly diagnosed conditions in reproductive age women is polycystic ovary syndrome (PCOS). PNA may affect the growth and development of many systems in the whole body and disrupts the normal metabolic trajectory in the offspring of PCOS, contributing to the prevalence of cardiovascular and metabolic diseases (CVMD), including myocardial hypertrophy, hypertension, hyperinsulinemia, insulin resistance, hyperglycemia, obesity, and dyslipidemia, which are the leading causes of hospitalizations in young PCOS offspring. In this review, we focus on the effects of prenatal androgenic exposure on the cardiovascular and metabolic diseases in offspring, discuss the possible pathogenesis respectively, and summarize potential management strategies to improve metabolic health of PCOS offspring. It is expected that the incidence of CVMD and the medical burden will be reduced in the future.
Collapse
Affiliation(s)
- Fei Guo
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Suqing Mao
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Yuhang Long
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Bokang Zhou
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ling Gao
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hefeng Huang
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
21
|
Schiffmann LM, de Groot E, Albert MC, Quaas A, Pinto Dos Santos D, Babic B, Fuchs HF, Walczak H, Chon SH, Ruurda JP, Kashkar H, Bruns CJ, Schröder W, van Hillegersberg R. Laparoscopic ischemic conditioning of the stomach prior to esophagectomy induces gastric neo-angiogenesis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:107096. [PMID: 37801834 DOI: 10.1016/j.ejso.2023.107096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND The risk of an anastomotic leakage (AL) following Ivor-Lewis esophagectomy is increased in patients with calcifications of the aorta or a stenosis of the celiac trunc. Ischemic conditioning (ISCON) of the gastric conduit prior to esophagectomy is supposed to improve gastric vascularization at the anastomotic site. The prospective ISCON trial was conducted to proof the safety and feasibility of this strategy with partial gastric devascularization 14 days before esophagectomy in esophageal cancer patients with a compromised vascular status. This work reports the results from a translational project of the ISCON trial aimed to investigate variables of neo-angiogenesis. METHODS Twenty esophageal cancer patients scheduled for esophagectomy were included in the ISCON trial. Serum samples (n = 11) were collected for measurement of biomarkers and biopsies (n = 12) of the gastric fundus were taken before and after ISCON of the gastric conduit. Serum samples were analyzed including 62 different cytokines. Vascularization of the gastric mucosa was assessed on paraffin-embedded sections stained against CD34 to detect the degree of microvascular density and vessel size. RESULTS Between November 2019 and January 2022 patients were included in the ISCON Trial. While serum samples showed no differences regarding cytokine levels before and after ISCON biopsies of the gastric mucosa demonstrated a significant increase in microvascular density after ISCON as compared to the corresponding gastric sample before the intervention. CONCLUSION The data prove that ISCON of the gastric conduit as esophageal substitute induces significant neo-angiogenesis in the gastric fundus which is considered as surrogate of an improved vascularization at the anastomotic site.
Collapse
Affiliation(s)
- L M Schiffmann
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - E de Groot
- Department of Surgery, University Medical Center Utrecht, POBOX 85500, 3508 GA, Utrecht, the Netherlands
| | - M C Albert
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - A Quaas
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - D Pinto Dos Santos
- Institute of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - B Babic
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - H F Fuchs
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - H Walczak
- Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - S-H Chon
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - J P Ruurda
- Department of Surgery, University Medical Center Utrecht, POBOX 85500, 3508 GA, Utrecht, the Netherlands
| | - H Kashkar
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Molecular Immunology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - C J Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - W Schröder
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - R van Hillegersberg
- Department of Surgery, University Medical Center Utrecht, POBOX 85500, 3508 GA, Utrecht, the Netherlands.
| |
Collapse
|
22
|
Hunt M, Torres M, Bachar-Wikström E, Wikström JD. Multifaceted roles of mitochondria in wound healing and chronic wound pathogenesis. Front Cell Dev Biol 2023; 11:1252318. [PMID: 37771375 PMCID: PMC10523588 DOI: 10.3389/fcell.2023.1252318] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Mitochondria are intracellular organelles that play a critical role in numerous cellular processes including the regulation of metabolism, cellular stress response, and cell fate. Mitochondria themselves are subject to well-orchestrated regulation in order to maintain organelle and cellular homeostasis. Wound healing is a multifactorial process that involves the stringent regulation of several cell types and cellular processes. In the event of dysregulated wound healing, hard-to-heal chronic wounds form and can place a significant burden on healthcare systems. Importantly, treatment options remain limited owing to the multifactorial nature of chronic wound pathogenesis. One area that has received more attention in recent years is the role of mitochondria in wound healing. With regards to this, current literature has demonstrated an important role for mitochondria in several areas of wound healing and chronic wound pathogenesis including metabolism, apoptosis, and redox signalling. Additionally, the influence of mitochondrial dynamics and mitophagy has also been investigated. However, few studies have utilised patient tissue when studying mitochondria in wound healing, instead using various animal models. In this review we dissect the current knowledge of the role of mitochondria in wound healing and discuss how future research can potentially aid in the progression of wound healing research.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D. Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Clahsen T, Hadrian K, Notara M, Schlereth SL, Howaldt A, Prokosch V, Volatier T, Hos D, Schroedl F, Kaser-Eichberger A, Heindl LM, Steven P, Bosch JJ, Steinkasserer A, Rokohl AC, Liu H, Mestanoglu M, Kashkar H, Schumacher B, Kiefer F, Schulte-Merker S, Matthaei M, Hou Y, Fassbender S, Jantsch J, Zhang W, Enders P, Bachmann B, Bock F, Cursiefen C. The novel role of lymphatic vessels in the pathogenesis of ocular diseases. Prog Retin Eye Res 2023; 96:101157. [PMID: 36759312 DOI: 10.1016/j.preteyeres.2022.101157] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 02/10/2023]
Abstract
Historically, the eye has been considered as an organ free of lymphatic vessels. In recent years, however, it became evident, that lymphatic vessels or lymphatic-like vessels contribute to several ocular pathologies at various peri- and intraocular locations. The aim of this review is to outline the pathogenetic role of ocular lymphatics, the respective molecular mechanisms and to discuss current and future therapeutic options based thereon. We will give an overview on the vascular anatomy of the healthy ocular surface and the molecular mechanisms contributing to corneal (lymph)angiogenic privilege. In addition, we present (i) current insights into the cellular and molecular mechanisms occurring during pathological neovascularization of the cornea triggered e.g. by inflammation or trauma, (ii) the role of lymphatic vessels in different ocular surface pathologies such as dry eye disease, corneal graft rejection, ocular graft versus host disease, allergy, and pterygium, (iii) the involvement of lymphatic vessels in ocular tumors and metastasis, and (iv) the novel role of the lymphatic-like structure of Schlemm's canal in glaucoma. Identification of the underlying molecular mechanisms and of novel modulators of lymphangiogenesis will contribute to the development of new therapeutic targets for the treatment of ocular diseases associated with pathological lymphangiogenesis in the future. The preclinical data presented here outline novel therapeutic concepts for promoting transplant survival, inhibiting metastasis of ocular tumors, reducing inflammation of the ocular surface, and treating glaucoma. Initial data from clinical trials suggest first success of novel treatment strategies to promote transplant survival based on pretransplant corneal lymphangioregression.
Collapse
Affiliation(s)
- Thomas Clahsen
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Maria Notara
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Simona L Schlereth
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Antonia Howaldt
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Verena Prokosch
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Volatier
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Falk Schroedl
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig M Heindl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Steven
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany
| | - Jacobus J Bosch
- Centre for Human Drug Research and Leiden University Medical Center, Leiden, the Netherlands
| | | | - Alexander C Rokohl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Mert Mestanoglu
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Institute for Molecular Immunology, Center for Molecular Medicine Cologne (CMMC), CECAD Research Center, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Björn Schumacher
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany
| | - Friedemann Kiefer
- European Institute for Molecular Imaging (EIMI), University of Münster, 48149, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster, Germany
| | - Mario Matthaei
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Yanhong Hou
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, China
| | - Sonja Fassbender
- IUF‒Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Immunology and Environment, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wei Zhang
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philip Enders
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Björn Bachmann
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Felix Bock
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany.
| |
Collapse
|
24
|
Li S, Yang C, Li J, Zhang C, Zhu L, Song Y, Guo Y, Wang R, Gan D, Shi J, Ma P, Gao F, Su H. Progress in Pluronic F127 Derivatives for Application in Wound Healing and Repair. Int J Nanomedicine 2023; 18:4485-4505. [PMID: 37576462 PMCID: PMC10416793 DOI: 10.2147/ijn.s418534] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Pluronic F127 hydrogel biomaterial has garnered considerable attention in wound healing and repair due to its remarkable properties including temperature sensitivity, injectability, biodegradability, and maintain a moist wound environment. This comprehensive review provides an in-depth exploration of the recent advancements in Pluronic F127-derived hydrogels, such as F127-CHO, F127-NH2, and F127-DA, focusing on their applications in the treatment of various types of wounds, ranging from burns and acute wounds to infected wounds, diabetic wounds, cutaneous tumor wounds, and uterine scars. Furthermore, the review meticulously examines the intricate interaction mechanisms employed by these hydrogels within the wound microenvironment. By elucidating the underlying mechanisms, discussing the strengths and weaknesses of Pluronic F127, analyzing the current state of wound healing development, and expanding on the trend of targeting mitochondria and cells with F127 as a nanomaterial. The review enhances our understanding of the therapeutic effects of these hydrogels aims to foster the development of effective and safe wound-healing modalities. The valuable insights provided this review have the potential to inspire novel ideas for clinical treatment and facilitate the advancement of innovative wound management approaches.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Cheng Yang
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Junqiang Li
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Chao Zhang
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Liaoliao Zhu
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Yang Song
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Yongdong Guo
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Ronglin Wang
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Dongxue Gan
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Jingjie Shi
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Peixiang Ma
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| | - Fei Gao
- Center for Peptide Functional Materials and Innovative Drugs, Institute of Translational Medicine, Shanghai University, ShangHai City, People’s Republic of China
| | - Haichuan Su
- Department of Oncology, The Second Affiliated Hospital, Air Force Medical University, Xi’an City, People’s Republic of China
| |
Collapse
|
25
|
Platelets Facilitate Wound Healing by Mitochondrial Transfer and Reducing Oxidative Stress in Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2345279. [PMID: 36860732 PMCID: PMC9970712 DOI: 10.1155/2023/2345279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/22/2023]
Abstract
As a critical member in wound healing, vascular endothelial cells (ECs) impaired under high levels of reactive oxygen species (ROS) would hamper neovascularization. Mitochondria transfer can reduce intracellular ROS damage under pathological condition. Meanwhile, platelets can release mitochondria and alleviate oxidative stress. However, the mechanism by which platelets promote cell survival and reduce oxidative stress damage has not been clarified. Here, first, we selected ultrasound as the best method for subsequent experiments by detecting the growth factors and mitochondria released from manipulation platelet concentrates (PCs), as well as the effect of manipulation PCs on the proliferation and migration of HUVECs. Then, we found that sonicate platelet concentrates (SPC) decreased the level of ROS in HUVECs treated with hydrogen peroxide in advance, increased mitochondrial membrane potential, and reduced apoptosis. By transmission electron microscope, we saw that two kinds of mitochondria, free or wrapped in vesicles, were released by activated platelets. In addition, we explored that platelet-derived mitochondria were transferred to HUVECs partly by means of dynamin-dependent clathrin-mediated endocytosis. Consistently, we determined that platelet-derived mitochondria reduced apoptosis of HUVECs caused by oxidative stress. What is more, we screened survivin as the target of platelet-derived mitochondria via high-throughput sequencing. Finally, we demonstrated that platelet-derived mitochondria promoted wound healing in vivo. Overall, these findings revealed that platelets are important donors of mitochondria, and platelet-derived mitochondria can promote wound healing by reducing apoptosis caused by oxidative stress in vascular endothelial cells. And survivin is a potential target. These results further expand the knowledge of the platelet function and provide new insights into the role of platelet-derived mitochondria in wound healing.
Collapse
|
26
|
Man HSJ, Subramaniam N, Downs T, Sukumar AN, Saha AD, Nair R, Chen L, Teitelbaum D, Turgeon PJ, Ku KH, Tran E, de Perrot M, Marsden PA. Long noncoding RNA GATA2-AS1 augments endothelial Hypoxia Inducible Factor 1-α induction and regulates hypoxic signaling. J Biol Chem 2023; 299:103029. [PMID: 36806681 PMCID: PMC10148162 DOI: 10.1016/j.jbc.2023.103029] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/18/2023] Open
Abstract
Vascular endothelial cells form the inner cellular lining of blood vessels and have myriad physiologic functions including angiogenesis and response to hypoxia. We recently identified a set of endothelial cell (EC)-enriched long noncoding RNAs (lncRNAs) in differentiated human primary cell types and described the role of the STEEL lncRNA in angiogenic patterning. We sought to further understand the role of EC-enriched lncRNAs in physiologic adaptation of the vascular endothelium. In this work, we describe an abundant, cytoplasmic, and EC-enriched lncRNA, GATA2-AS1, that is divergently transcribed from the EC-enriched developmental regulator, GATA2. While GATA2-AS1 is largely co-expressed with GATA2 in ECs, GATA2-AS1 and GATA2 appear to be complementary rather than synergistic as they have mostly distinct target genes. Common single nucleotide variants (SNVs) in GATA2-AS1 exons are associated with early onset coronary artery disease (CAD) and decreased expression of GATA2-AS1 in endothelial cell lines. In most cells, HIF1-α is central to the transcriptional response to hypoxia, while in ECs, both HIF1-α and HIF2-α are required to coordinate an acute and chronic response respectively. In this setting, GATA2-AS1 contributes to the "HIF switch" and augments HIF1-α induction in acute hypoxia to regulate HIF1-α/ HIF2-α balance. In hypoxia, GATA2-AS1 orchestrates HIF1-α-dependent induction of the glycolytic pathway, and HIF1-α-independent maintenance of mitochondrial biogenesis. Similarly, GATA2-AS1 coordinates both metabolism and "tip/stalk" cell signaling to regulate angiogenesis in hypoxic ECs. Furthermore, we find that GATA2-AS1 expression patterns are perturbed in atherosclerotic disease. Together, these results define a role for GATA2-AS1 in the EC-specific response to hypoxia.
Collapse
Affiliation(s)
- H S Jeffrey Man
- Institute of Medical Science, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Department of Respirology, University Health Network, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Noeline Subramaniam
- Institute of Medical Science, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Tiana Downs
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Aravin N Sukumar
- Institute of Medical Science, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Aninda D Saha
- Institute of Medical Science, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ranju Nair
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lucy Chen
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Teitelbaum
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Paul J Turgeon
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kyung Ha Ku
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eileen Tran
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Division of Thoracic Surgery, Toronto General Hospital, Toronto, Ontario, Canada
| | - Philip A Marsden
- Institute of Medical Science, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
27
|
Cyclic stretch promotes vascular homing of endothelial progenitor cells via Acsl1 regulation of mitochondrial fatty acid oxidation. Proc Natl Acad Sci U S A 2023; 120:e2219630120. [PMID: 36716379 PMCID: PMC9963562 DOI: 10.1073/pnas.2219630120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Endothelial progenitor cells (EPCs) play an important role in vascular repair and re-endothelialization after vessel injury. EPCs in blood vessels are subjected to cyclic stretch (CS) due to the pulsatile pressure, but the role of CS in metabolic reprogramming of EPC, particularly its vascular homing and repair, is largely unknown. In the current study, physiological CS applied to EPCs at a magnitude of 10% and a frequency of 1 Hz significantly promoted their vascular adhesion and endothelial differentiation. CS enhanced mitochondrial elongation and oxidative phosphorylation (OXPHOS), as well as adenosine triphosphate production. Metabolomic study and Ultra-high performance liquid chromatography-mass spectrometry assay revealed that CS significantly decreased the content of long-chain fatty acids (LCFAs) and markedly induced long-chain fatty acyl-CoA synthetase 1 (Acsl1), which in turn facilitated the catabolism of LCFAs in mitochondria via fatty acid β-oxidation and OXPHOS. In a rat carotid artery injury model, transplantation of EPCs overexpressing Acsl1 enhanced the adhesion and re-endothelialization of EPCs in vivo. MRI and vascular morphology staining showed that Acsl1 overexpression in EPCs improved vascular repair and inhibited vascular stenosis. This study reveals a mechanotransduction mechanism by which physiological CS enhances endothelial repair via EPC patency.
Collapse
|
28
|
Hyroššová P, Milošević M, Škoda J, Vachtenheim Jr J, Rohlena J, Rohlenová K. Effects of metabolic cancer therapy on tumor microenvironment. Front Oncol 2022; 12:1046630. [PMID: 36582801 PMCID: PMC9793001 DOI: 10.3389/fonc.2022.1046630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Targeting tumor metabolism for cancer therapy is an old strategy. In fact, historically the first effective cancer therapeutics were directed at nucleotide metabolism. The spectrum of metabolic drugs considered in cancer increases rapidly - clinical trials are in progress for agents directed at glycolysis, oxidative phosphorylation, glutaminolysis and several others. These pathways are essential for cancer cell proliferation and redox homeostasis, but are also required, to various degrees, in other cell types present in the tumor microenvironment, including immune cells, endothelial cells and fibroblasts. How metabolism-targeted treatments impact these tumor-associated cell types is not fully understood, even though their response may co-determine the overall effectivity of therapy. Indeed, the metabolic dependencies of stromal cells have been overlooked for a long time. Therefore, it is important that metabolic therapy is considered in the context of tumor microenvironment, as understanding the metabolic vulnerabilities of both cancer and stromal cells can guide new treatment concepts and help better understand treatment resistance. In this review we discuss recent findings covering the impact of metabolic interventions on cellular components of the tumor microenvironment and their implications for metabolic cancer therapy.
Collapse
Affiliation(s)
- Petra Hyroššová
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Mirko Milošević
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Josef Škoda
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Jiří Vachtenheim Jr
- 3rd Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jakub Rohlena
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Kateřina Rohlenová
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
29
|
Mitochondrial dysfunction induces ALK5-SMAD2-mediated hypovascularization and arteriovenous malformations in mouse retinas. Nat Commun 2022; 13:7637. [PMID: 36496409 PMCID: PMC9741628 DOI: 10.1038/s41467-022-35262-w] [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: 02/16/2021] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Although mitochondrial activity is critical for angiogenesis, its mechanism is not entirely clear. Here we show that mice with endothelial deficiency of any one of the three nuclear genes encoding for mitochondrial proteins, transcriptional factor (TFAM), respiratory complex IV component (COX10), or redox protein thioredoxin 2 (TRX2), exhibit retarded retinal vessel growth and arteriovenous malformations (AVM). Single-cell RNA-seq analyses indicate that retinal ECs from the three mutant mice have increased TGFβ signaling and altered gene expressions associated with vascular maturation and extracellular matrix, correlating with vascular malformation and increased basement membrane thickening in microvesels of mutant retinas. Mechanistic studies suggest that mitochondrial dysfunction from Tfam, Cox10, or Trx2 depletion induces a mitochondrial localization and MAPKs-mediated phosphorylation of SMAD2, leading to enhanced ALK5-SMAD2 signaling. Importantly, pharmacological blockade of ALK5 signaling or genetic deficiency of SMAD2 prevented retinal vessel growth retardation and AVM in all three mutant mice. Our studies uncover a novel mechanism whereby mitochondrial dysfunction via the ALK5-SMAD2 signaling induces retinal vascular malformations, and have therapeutic values for the alleviation of angiogenesis-associated human retinal diseases.
Collapse
|
30
|
Wilson C, Lee MD, Buckley C, Zhang X, McCarron JG. Mitochondrial ATP Production is Required for Endothelial Cell Control of Vascular Tone. FUNCTION 2022; 4:zqac063. [PMID: 36778749 PMCID: PMC9909368 DOI: 10.1093/function/zqac063] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Arteries and veins are lined by nonproliferating endothelial cells that play a critical role in regulating blood flow. Endothelial cells also regulate tissue perfusion, metabolite exchange, and thrombosis. It is thought that endothelial cells rely on ATP generated via glycolysis, rather than mitochondrial oxidative phosphorylation, to fuel each of these energy-demanding processes. However, endothelial metabolism has mainly been studied in the context of proliferative cells, and little is known about energy production in endothelial cells within the fully formed vascular wall. Using intact arteries isolated from rats and mice, we show that inhibiting mitochondrial respiration disrupts endothelial control of vascular tone. Basal, mechanically activated, and agonist-evoked calcium activity in intact artery endothelial cells are each prevented by inhibiting mitochondrial ATP synthesis. Agonist-evoked calcium activity was also inhibited by blocking the transport of pyruvate, the master fuel for mitochondrial energy production, through the mitochondrial pyruvate carrier. The role for mitochondria in endothelial cell energy production is independent of species, sex, or vascular bed. These data show that a mitochondrial ATP supply is necessary for calcium-dependent, nitric oxide-mediated endothelial control of vascular tone, and identifies the critical role of endothelial mitochondrial energy production in fueling perfused blood vessel function.
Collapse
Affiliation(s)
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | | |
Collapse
|
31
|
Xu S, Li S, Bjorklund M, Xu S. Mitochondrial fragmentation and ROS signaling in wound response and repair. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:38. [PMID: 36451031 PMCID: PMC9712903 DOI: 10.1186/s13619-022-00141-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/14/2022] [Indexed: 12/03/2022]
Abstract
Mitochondria are organelles that serve numerous critical cellular functions, including energy production, Ca2+ homeostasis, redox signaling, and metabolism. These functions are intimately linked to mitochondrial morphology, which is highly dynamic and capable of rapid and transient changes to alter cellular functions in response to environmental cues and cellular demands. Mitochondrial morphology and activity are critical for various physiological processes, including wound healing. In mammals, wound healing is a complex process that requires coordinated function of multiple cell types and progresses in partially overlapping but distinct stages: hemostasis and inflammation, cell proliferation and migration, and tissue remodeling. The repair process at the single-cell level forms the basis for wound healing and regeneration in tissues. Recent findings reveal that mitochondria fulfill the intensive energy demand for wound repair and aid wound closure by cytoskeleton remodeling via morphological changes and mitochondrial reactive oxygen species (mtROS) signaling. In this review, we will mainly elucidate how wounding induces changes in mitochondrial morphology and activity and how these changes, in turn, contribute to cellular wound response and repair.
Collapse
Affiliation(s)
- Shiqi Xu
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China
| | - Shiyao Li
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China
| | - Mikael Bjorklund
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China
| | - Suhong Xu
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China.
- Center for Stem Cell and Regenerative Medicine and Department of Burn and wound repair of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
32
|
Zhu W, Wang J, Liu X, Xu Y, Zhai R, Zhang J, Wang M, Wang M, Liu L. lncRNA CYTOR promotes aberrant glycolysis and mitochondrial respiration via HNRNPC-mediated ZEB1 stabilization in oral squamous cell carcinoma. Cell Death Dis 2022; 13:703. [PMID: 35963855 PMCID: PMC9376070 DOI: 10.1038/s41419-022-05157-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 01/21/2023]
Abstract
Oral squamous cell carcinoma (OSCC), the most common malignancy of the oral and maxillofacial region, severely affects human health. However, current treatments for OSCC commonly show only a ~60% 5-year survival rate of patients with distant metastases, indicating an urgent need for targeted treatments for patients with advanced metastases. Here, we report a survival-related long non-coding RNA, CYTOR, which is highly expressed in the lesions of oral cancer patients. We found that CYTOR can promote both migration and invasion in oral cancer cells as well as the epithelial-mesenchymal transition (EMT). RNA-sequencing of CYTOR-knockdown oral cancer cells revealed that CYTOR can regulate mitochondrial respiration and RNA splicing. Mechanistically, we found that nuclear-localized CYTOR interacts with HNRNPC, resulting in stabilization of ZEB1 mRNAs by inhibiting the nondegradative ubiquitination of HNRNPC. By synthesizing CYTOR-targeting small interfering RNAs (siRNAs) encapsulated in Nanoscale Metal Organic Frameworks (NMOFs), we demonstrate the targeted suppression of CYTOR to inhibit invasion and metastasis of oral cancer cells in a nude mouse model. Cumulatively, this study reveals the potential role of the CYTOR-HNRNPC-ZEB1 axis in regulating mitochondrial metabolism and glycolysis of oral cancer cells, and illustrates the effective use of lncRNA targeting in anti-metastatic cancer therapies.
Collapse
Affiliation(s)
- Weiwen Zhu
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Jie Wang
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Xiang Liu
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Yanbin Xu
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Rundong Zhai
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Jiayi Zhang
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Mengqi Wang
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Mengyao Wang
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Laikui Liu
- grid.89957.3a0000 0000 9255 8984Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China ,grid.89957.3a0000 0000 9255 8984Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China ,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| |
Collapse
|
33
|
Ho WT, Chang JS, Chen TC, Wang JK, Chang SW, Yang MH, Jou TS, Wang IJ. Inhibition of Rho-associated protein kinase activity enhances oxidative phosphorylation to support corneal endothelial cell migration. FASEB J 2022; 36:e22397. [PMID: 35661268 DOI: 10.1096/fj.202101442rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
Corneal endothelial cell (CEC) dysfunction causes corneal edema and severe visual impairment that require transplantation to restore vision. To address the unmet need of organ shortage, descemetorhexis without endothelial keratoplasty has been specifically employed to treat early stage Fuchs endothelial corneal dystrophy, which is pathophysiologically related to oxidative stress and exhibits centrally located corneal guttae. After stripping off central Descemet's membrane, rho-associated protein kinase (ROCK) inhibitor has been found to facilitate CEC migration, an energy-demanding task, thereby achieving wound closure. However, the correlation between ROCK inhibition and the change in bioenergetic status of CECs remained to be elucidated. Through transcriptomic profiling, we found that the inhibition of ROCK activity by the selective inhibitor, ripasudil or Y27632, promoted enrichment of oxidative phosphorylation (OXPHOS) gene set in bovine CECs (BCECs). Functional analysis revealed that ripasudil, a clinically approved anti-glaucoma agent, enhanced mitochondrial respiration, increased spare respiratory capacity, and induced overexpression of electron transport chain components through upregulation of AMP-activated protein kinase (AMPK) pathway. Accelerated BCEC migration and in vitro wound healing by ripasudil were diminished by OXPHOS and AMPK inhibition, but not by glycolysis inhibition. Correspondingly, lamellipodial protrusion and actin assembly that were augmented by ripasudil became reduced with additional OXPHOS or AMPK inhibition. These results indicate that ROCK inhibition induces metabolic reprogramming toward OXPHOS to support migration of CECs.
Collapse
Affiliation(s)
- Wei-Ting Ho
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jung-Shen Chang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsan-Chi Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Jia-Kang Wang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Electrical Engineering, Yuan Ze University, Taoyuan, Taiwan
| | - Shu-Wen Chang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzuu-Shuh Jou
- College of Medicine, National Taiwan University, Taipei, Taiwan.,Center of Precision Medicine, College of Medicine, National Taiwan University, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Daoud M, Broxtermann PN, Schorn F, Werthenbach JP, Seeger JM, Schiffmann LM, Brinkmann K, Vucic D, Tüting T, Mauch C, Kulms D, Zigrino P, Kashkar H. XIAP promotes melanoma growth by inducing tumour neutrophil infiltration. EMBO Rep 2022; 23:e53608. [PMID: 35437868 PMCID: PMC9171690 DOI: 10.15252/embr.202153608] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/22/2022] Open
Abstract
Elevated expression of the X‐linked inhibitor of apoptosis protein (XIAP) has been frequently reported in malignant melanoma suggesting that XIAP renders apoptosis resistance and thereby supports melanoma progression. Independent of its anti‐apoptotic function, XIAP mediates cellular inflammatory signalling and promotes immunity against bacterial infection. The pro‐inflammatory function of XIAP has not yet been considered in cancer. By providing detailed in vitro analyses, utilising two independent mouse melanoma models and including human melanoma samples, we show here that XIAP is an important mediator of melanoma neutrophil infiltration. Neutrophils represent a major driver of melanoma progression and are increasingly considered as a valuable therapeutic target in solid cancer. Our data reveal that XIAP ubiquitylates RIPK2, involve TAB1/RIPK2 complex and induce the transcriptional up‐regulation and secretion of chemokines such as IL8, that are responsible for intra‐tumour neutrophil accumulation. Alteration of the XIAP‐RIPK2‐TAB1 inflammatory axis or the depletion of neutrophils in mice reduced melanoma growth. Our data shed new light on how XIAP contributes to tumour growth and provides important insights for novel XIAP targeting strategies in cancer.
Collapse
Affiliation(s)
- Mila Daoud
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Pia Nora Broxtermann
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Fabian Schorn
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - J Paul Werthenbach
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jens Michael Seeger
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lars M Schiffmann
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral, Cancer and Transplant Surgery, University of Cologne, Cologne, Germany
| | - Kerstin Brinkmann
- The Walter & Eliza Hall Institute of Medical Research (WEHI) and Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Thomas Tüting
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital Magdeburg, Magdeburg, Germany
| | - Cornelia Mauch
- Faculty of Medicine and University Hospital of Cologne, Department of Dermatology and Venereology, University of Cologne, Cologne, Germany
| | - Dagmar Kulms
- Department of Dermatology, Experimental Dermatology, TU-Dresden, Dresden, Germany.,National Center for Tumor Diseases Dresden, TU-Dresden, Dresden, Germany
| | - Paola Zigrino
- Faculty of Medicine and University Hospital of Cologne, Department of Dermatology and Venereology, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
35
|
Mammalian Sterile 20-Like Kinase 1 Mediates Neuropathic Pain Associated with Its Effects on Regulating Mitophagy in Schwann Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3458283. [PMID: 35656021 PMCID: PMC9155917 DOI: 10.1155/2022/3458283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/20/2022]
Abstract
Myelin degradation initiated by Schwann cells (SCs) after nerve injury is connected to the induction and chronicity of neuropathic pain (NP). Mitophagy, a selective clearance of damaged mitochondria via autophagy, contributes to the maintenance of normal function in SCs. Mitochondrial function and mitophagy activity are highly modulated by mammalian ste20-like kinase1 (Mst1). However, whether Mst1 can regulate mitophagy in SCs to play a role in NP remains poorly understood. In the present study, Sprague-Dawley rats were subjected to chronic constriction injury (CCI) on the sciatic nerve to induce NP. Small interfering RNA of Mst1 was applied to the injured sciatic nerve to knockdown Mst1. Behavioral tests were performed to evaluate NP, and myelin degeneration was assessed by transmission electron microscope and immunofluorescence. Autophagy and mitophagy were detected in the injured sciatic nerve and cultured SCs (RSC96 cells) by Western blot. ROS level, mitochondria membrane potential, and apoptosis were assessed in vitro via flow cytometry and Western blot. Mst1 knockdown alleviated mechanical allodynia and thermal hyperalgesia in the CCI-induced NP model and rescued myelin degeneration of the injured nerve. Meanwhile, CCI-increased levels of Parkin and p62 were reversed by Mst1 knockdown. In vitro RSC96 cells were subjected to starvation to induce mitophagy. Protein levels of mitochondrial Parkin and mitochondrial p62 significantly increased after Mst1 knockdown, while those in the cytosol diminished indicate that the translocation of Parkin and p62 from the cytosol to the mitochondria was promoted by the knockdown of Mst1. In addition, Mst1 knockdown reduced ROS level and apoptosis activity, while enhancing mitochondria membrane potential in RSC96 cells. The study showed that Mst1 knockdown alleviated CCI-induced NP, associated with enhanced Parkin recruitment to mitochondria and subsequent mitophagy degradation, thus preserving mitochondrial function and myelin integrity.
Collapse
|
36
|
Schiffmann LM, Bruns CJ, Schmidt T. Resistance Mechanisms of the Metastatic Tumor Microenvironment to Anti-Angiogenic Therapy. Front Oncol 2022; 12:897927. [PMID: 35664794 PMCID: PMC9162757 DOI: 10.3389/fonc.2022.897927] [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: 03/16/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Angiogenesis describes the formation of blood vessels from an existing vascular network. Anti-angiogenic drugs that target tumor blood vessels have become standard of care in many cancer entities. Though very promising results in preclinical evaluation, anti-angiogenic treatments fell short of expectations in clinical trials. Patients develop resistance over time or are primarily refractory to anti-angiogenic therapies similar to conventional chemotherapy. To further improve efficacy and outcome to these therapies, a deeper understanding of mechanisms that mediate resistance to anti-angiogenic therapies is needed. The field has done tremendous efforts to gain knowledge about how tumors engage tumor cell and microenvironmental mechanisms to do so. This review highlights the current state of knowledge with special focus on the metastatic tumor site and potential therapeutic relevance of this understanding from a translational and clinical perspective.
Collapse
Affiliation(s)
- Lars M. Schiffmann
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Thomas Schmidt
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
37
|
Madan S, Uttekar B, Chowdhary S, Rikhy R. Mitochondria Lead the Way: Mitochondrial Dynamics and Function in Cellular Movements in Development and Disease. Front Cell Dev Biol 2022; 9:781933. [PMID: 35186947 PMCID: PMC8848284 DOI: 10.3389/fcell.2021.781933] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023] Open
Abstract
The dynamics, distribution and activity of subcellular organelles are integral to regulating cell shape changes during various physiological processes such as epithelial cell formation, cell migration and morphogenesis. Mitochondria are famously known as the powerhouse of the cell and play an important role in buffering calcium, releasing reactive oxygen species and key metabolites for various activities in a eukaryotic cell. Mitochondrial dynamics and morphology changes regulate these functions and their regulation is, in turn, crucial for various morphogenetic processes. In this review, we evaluate recent literature which highlights the role of mitochondrial morphology and activity during cell shape changes in epithelial cell formation, cell division, cell migration and tissue morphogenesis during organism development and in disease. In general, we find that mitochondrial shape is regulated for their distribution or translocation to the sites of active cell shape dynamics or morphogenesis. Often, key metabolites released locally and molecules buffered by mitochondria play crucial roles in regulating signaling pathways that motivate changes in cell shape, mitochondrial shape and mitochondrial activity. We conclude that mechanistic analysis of interactions between mitochondrial morphology, activity, signaling pathways and cell shape changes across the various cell and animal-based model systems holds the key to deciphering the common principles for this interaction.
Collapse
|
38
|
Mendez DL, Lowder EP, Tillman DE, Sutherland MC, Collier AL, Rau MJ, Fitzpatrick JA, Kranz RG. Cryo-EM of CcsBA reveals the basis for cytochrome c biogenesis and heme transport. Nat Chem Biol 2022; 18:101-108. [PMID: 34931065 PMCID: PMC8712405 DOI: 10.1038/s41589-021-00935-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/22/2021] [Indexed: 01/09/2023]
Abstract
Although the individual structures and respiratory functions of cytochromes are well studied, the structural basis for their assembly, including transport of heme for attachment, are unknown. We describe cryo-electron microscopy (cryo-EM) structures of CcsBA, a bifunctional heme transporter and cytochrome c (cyt c) synthase. Models built from the cryo-EM densities show that CcsBA is trapped with heme in two conformations, herein termed the closed and open states. The closed state has heme located solely at a transmembrane (TM) site, with a large periplasmic domain oriented such that access of heme to the cytochrome acceptor is denied. The open conformation contains two heme moieties, one in the TM-heme site and another in an external site (P-heme site). The presence of heme in the periplasmic site at the base of a chamber induces a large conformational shift that exposes the heme for reaction with apocytochrome c (apocyt c). Consistent with these structures, in vivo and in vitro cyt c synthase studies suggest a mechanism for transfer of the periplasmic heme to cytochrome.
Collapse
Affiliation(s)
- Deanna L. Mendez
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Ethan P. Lowder
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Dustin E. Tillman
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Molly C. Sutherland
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrea L. Collier
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael J. Rau
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James A.J. Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA.,Departments of Cell Biology & Physiology and Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Robert G. Kranz
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA,Corresponding author is Robert G. Kranz:
| |
Collapse
|
39
|
Fabri M, Villa M, Stanczak MA, Edwards-Hicks J, Corrado M, Pearce EL. Research Techniques Made Simple: Profiling Cellular Energy Metabolism. J Invest Dermatol 2021; 141:2767-2774.e2. [PMID: 34802549 DOI: 10.1016/j.jid.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 10/19/2022]
Abstract
The analysis of cellular metabolism is attracting increasing interest. Glycolysis and oxidative phosphorylation are intertwined with one another and dozens of other pathways to ultimately produce energy and maintain cellular fitness. However, cellular metabolism is much more than this. Metabolism underlies the proliferation, differentiation, and function of cells as well as the coordination of intercellular communication. Investigating metabolism allows the interpretation of cellular behavior in health and disease. In this article, we aim to demystify the complexity of cellular metabolism and explain the common approaches to study it. Whereas the analysis of cellular metabolism by western blot or flow cytometry might be accessible to most investigators, the functional and comprehensive analyses obtained with a Seahorse Analyzer or mass spectrometer come with monetary and logistical hurdles. We believe that the application of these techniques, together with collaborative efforts between scientists and clinicians, will uncover disease mechanisms and open novel therapeutic avenues for unmet clinical needs in the field of dermatology.
Collapse
Affiliation(s)
- Mario Fabri
- Department of Dermatology and Venereology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany.
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Michal A Stanczak
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany; The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Mauro Corrado
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Erika L Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany; The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Theobald SJ, Gräb J, Fritsch M, Suárez I, Eisfeld HS, Winter S, Koch M, Hölscher C, Pasparakis M, Kashkar H, Rybniker J. Gasdermin D mediates host cell death but not interleukin-1β secretion in Mycobacterium tuberculosis-infected macrophages. Cell Death Discov 2021; 7:327. [PMID: 34718331 PMCID: PMC8557205 DOI: 10.1038/s41420-021-00716-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 11/09/2022] Open
Abstract
Necrotic cell death represents a major pathogenic mechanism of Mycobacterium tuberculosis (Mtb) infection. It is increasingly evident that Mtb induces several types of regulated necrosis but how these are interconnected and linked to the release of pro-inflammatory cytokines remains unknown. Exploiting a clinical cohort of tuberculosis patients, we show here that the number and size of necrotic lesions correlates with IL-1β plasma levels as a strong indicator of inflammasome activation. Our mechanistic studies reveal that Mtb triggers mitochondrial permeability transition (mPT) and subsequently extensive macrophage necrosis, which requires activation of the NLRP3 inflammasome. NLRP3-driven mitochondrial damage is dependent on proteolytic activation of the pore-forming effector protein gasdermin D (GSDMD), which links two distinct cell death machineries. Intriguingly, GSDMD, but not the membranolytic mycobacterial ESX-1 secretion system, is dispensable for IL-1β secretion from Mtb-infected macrophages. Thus, our study dissects a novel mechanism of pathogen-induced regulated necrosis by identifying mitochondria as central regulatory hubs capable of delineating cytokine secretion and lytic cell death.
Collapse
Affiliation(s)
- Sebastian J Theobald
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Jessica Gräb
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Melanie Fritsch
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.,Institute for Medical Microbiology, Immunology and Hygiene (IMMIH), University of Cologne, 50935, Cologne, Germany
| | - Isabelle Suárez
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Hannah S Eisfeld
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Sandra Winter
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Maximilian Koch
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Christoph Hölscher
- Division of Infection Immunology, Research Center Borstel, 23845, Borstel, Germany.,German Center for Infection Research (DZIF), Partner Site Borstel, 23845, Borstel, Germany
| | - Manolis Pasparakis
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.,Institute for Genetics, University of Cologne, 50674, Cologne, Germany
| | - Hamid Kashkar
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.,Institute for Medical Microbiology, Immunology and Hygiene (IMMIH), University of Cologne, 50935, Cologne, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, University of Cologne, 50937, Cologne, Germany. .,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany. .,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
| |
Collapse
|
41
|
Tumor Microenvironment of Esophageal Cancer. Cancers (Basel) 2021; 13:cancers13184678. [PMID: 34572905 PMCID: PMC8472305 DOI: 10.3390/cancers13184678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Esophageal cancer is one of the top ten most deadly cancers. Even when diagnosed in a curable stage, patients prognosis poor. One of the parameters that is very relevant for long-term survival is response to radio(chemo)therapy prior surgery. Complete response rates are between 24 and 50 percent. This puts more than a half of every esophageal cancer patient that is diagnosed in a non-metastasized stage at high risk of recurrence. To improve response rates of treatment regimens prior curative surgery is, therefore, a major challenge in treating esophageal cancer. Not only the response of the cancer cell itself to cancer therapy is determining patients’ fate. Cells around the tumor cells called the tumor microenvironment that together with the cancer cell constitute a malignant tumor are also involved in tumor progression and therapy response. This review depicts the most important parts of the esophageal cancer microenvironment, evaluates chances and challenges of current already established therapeutic concepts that target this microenvironment. It furthermore elucidates specific pathways that are potential valuable targets in the future. Abstract Esophageal cancer is among the top ten most deadly cancers worldwide with adenocarcinomas of the esophagus showing increasing incidences over the last years. The prognosis is determined by tumor stage at diagnosis and in locally advanced stages by response to (radio-)chemotherapy followed by radical surgery. Less than a third of patients with esophageal adenocarcinomas completely respond to neoadjuvant therapies which urgently asks for further strategies to improve these rates. Aiming at the tumor microenvironment with novel targeted therapies can be one strategy to achieve this goal. This review connects experimental, translational, and clinical findings on each component of the esophageal cancer tumor microenvironment involving tumor angiogenesis, tumor-infiltrating immune cells, such as macrophages, T-cells, myeloid-derived suppressor cells, and cancer-associated fibroblasts. The review evaluates the current state of already approved concepts and depicts novel potentially targetable pathways related to esophageal cancer tumor microenvironment.
Collapse
|
42
|
Endothelial Heterogeneity in Development and Wound Healing. Cells 2021; 10:cells10092338. [PMID: 34571987 PMCID: PMC8469713 DOI: 10.3390/cells10092338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature is comprised of endothelial cells that are heterogeneous in nature. From tissue resident progenitors to mature differentiated endothelial cells, the diversity of these populations allows for the formation, maintenance, and regeneration of the vascular system in development and disease, particularly during situations of wound healing. Additionally, the de-differentiation and plasticity of different endothelial cells, especially their capacity to undergo endothelial to mesenchymal transition, has also garnered significant interest due to its implication in disease progression, with emphasis on scarring and fibrosis. In this review, we will pinpoint the seminal discoveries defining the phenotype and mechanisms of endothelial heterogeneity in development and disease, with a specific focus only on wound healing.
Collapse
|
43
|
Metabolic orchestration of the wound healing response. Cell Metab 2021; 33:1726-1743. [PMID: 34384520 DOI: 10.1016/j.cmet.2021.07.017] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/16/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Wound healing requires cooperation between different cell types, among which macrophages play a central role. In particular, inflammatory macrophages are engaged in the initial response to wounding, and alternatively activated macrophages are essential for wound closure and the resolution of tissue repair. The links between temporal activation-induced changes in the metabolism of such macrophages and the influence this has on their functional states, along with the realization that metabolites play both intrinsic and extrinsic roles in the cells that produce them, has focused attention on the metabolism of wound healing. Here, we discuss macrophage metabolism during distinct stages of normal healing and its related pathologic processes, such as during cancer and fibrosis. Further, we frame these insights in a broader context of the current understanding of macrophage metabolic reprogramming linked to cellular activation and function. Finally, we discuss parallels between the metabolism of macrophages and fibroblasts, the latter being a key stromal cell type in wound healing, and consider the importance of the metabolic interplay between different cell types in the wound microenvironment.
Collapse
|
44
|
Laschke MW, Menger MD. The simpler, the better: tissue vascularization using the body's own resources. Trends Biotechnol 2021; 40:281-290. [PMID: 34404555 DOI: 10.1016/j.tibtech.2021.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
Tissue regeneration is crucially dependent on sufficient vascularization. In regenerative medicine, this can be effectively achieved by autologous vascularization strategies using the body's own resources. These strategies include the administration of blood-derived factor preparations, adipose tissue-based vascularization, and the in situ engineering of vascularized tissue. Due to their simplicity, the translation of these strategies into clinical practice is easier in terms of feasibility, safety requirements, and regulatory hurdles compared with complex and time-consuming procedures involving intensive cell manipulation. Hence, they are close to clinical application or are already being used to successfully treat patients by distinct personalized medicine concepts.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany.
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|
45
|
Evans P, Wojta J, Hoefer IE, Waltenberger J, Guzik T, Badimon L, Weber C. The year in basic vascular biology research: from mechanoreceptors and neutrophil extracellular traps to smartphone data and omics. Cardiovasc Res 2021; 117:1814-1822. [PMID: 33744925 PMCID: PMC8083796 DOI: 10.1093/cvr/cvab105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022] Open
Abstract
2020 has been an extraordinary year. The emergence of COVID-19 has driven urgent research in pulmonary and cardiovascular science and other fields. It has also shaped the way that we work with many experimental laboratories shutting down for several months, while bioinformatics approaches and other large data projects have gained prominence. Despite these setbacks, vascular biology research is stronger than ever. On behalf of the European Society of Cardiology Council for Basic Cardiovascular Science (ESC CBCS), here we review some of the vascular biology research highlights for 2020. This review is not exhaustive and there are many outstanding vascular biology publications that we were unable to cite due to page limits. Notwithstanding this, we have provided a snapshot of vascular biology research excellence in 2020 and identify topics that are in the ascendency and likely to gain prominence in coming years.
Collapse
Affiliation(s)
- Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.,Insigneo Institute, Sheffield, UK
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Imo E Hoefer
- Central Diagnostic Laboratory, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.,Department of Medicine, Jagiellonian University Collegium Medicum, Cracow, Poland
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian-Universität (LMU) München, München, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; and.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
46
|
Liang X, Lin F, Ding Y, Zhang Y, Li M, Zhou X, Meng Q, Ma X, Wei L, Fan H, Liu Z. Conditioned medium from induced pluripotent stem cell-derived mesenchymal stem cells accelerates cutaneous wound healing through enhanced angiogenesis. Stem Cell Res Ther 2021; 12:295. [PMID: 34016178 PMCID: PMC8139053 DOI: 10.1186/s13287-021-02366-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/02/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) can improve cutaneous wound healing via the secretion of growth factors. However, the therapeutic efficacy of MSCs varies depending upon their source. Induced pluripotent stem cells are emerging as a promising source of MSCs with the potential to overcome several limitations of adult MSCs. This study compared the effectiveness of conditioned medium of MSCs derived from induced pluripotent stem cells (iMSC-CdM) with that derived from umbilical cord MSCs (uMSC-CdM) in a mouse cutaneous wound healing model. We also investigated the mechanisms of protection. METHODS The iMSC-CdM or uMSC-CdM were topically applied to mice cutaneous wound model. The recovery rate, scar formation, inflammation and angiogenesis were measured. We compared angiogenesis cytokine expression between iMSC-CdM and uMSC-CdM and their protective effects on human umbilical vein endothelial cells (HUVECs) under H2O2-induced injury. The effects of iMSC-CdM on energy metabolism, mitochondria fragmentation and apoptosis were measured. RESULTS Topical application of iMSC-CdM was superior to the uMSC-CdM in accelerating wound closure and enhancing angiogenesis. Expression levels of angiogenetic cytokines were higher in iMSC-CdM than they were in uMSC-CdM. The iMSC-CdM protected HUVECs from H2O2 induced injury more effectively than uMSC-CdM did. Administration of iMSC-CdM stimulated HUVEC proliferation, tube formation and energy metabolism via the ERK pathway. Mechanistically, iMSC-CdM inhibited H2O2-induced mitochondrial fragmentation and apoptosis of HUVECs. CONCLUSION Collectively, these findings indicate that iMSC-CdM is more effective than uMSC-CdM in treating cutaneous wounds, and in this way, iMSC-CdM may serve as a more constant and sustainable source for cell-free therapeutic approach.
Collapse
Affiliation(s)
- Xiaoting Liang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Fang Lin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
- Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University, Shanghai, People's Republic of China
| | - Yue Ding
- Department of Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yuelin Zhang
- Department of Emergency, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, People's Republic of China
| | - Mimi Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xiaoxue Ma
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Lu Wei
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Huimin Fan
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China.
| | - Zhongmin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China.
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China.
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
47
|
Pasut A, Becker LM, Cuypers A, Carmeliet P. Endothelial cell plasticity at the single-cell level. Angiogenesis 2021; 24:311-326. [PMID: 34061284 PMCID: PMC8169404 DOI: 10.1007/s10456-021-09797-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 02/08/2023]
Abstract
The vascular endothelium is characterized by a remarkable level of plasticity, which is the driving force not only of physiological repair/remodeling of adult tissues but also of pathological angiogenesis. The resulting heterogeneity of endothelial cells (ECs) makes targeting the endothelium challenging, no less because many EC phenotypes are yet to be identified and functionally inventorized. Efforts to map the vasculature at the single-cell level have been instrumental to capture the diversity of EC types and states at a remarkable depth in both normal and pathological states. Here, we discuss new EC subtypes and functions emerging from recent single-cell studies in health and disease. Interestingly, such studies revealed distinct metabolic gene signatures in different EC phenotypes, which deserve further consideration for therapy. We highlight how this metabolic targeting strategy could potentially be used to promote (for tissue repair) or block (in tumor) angiogenesis in a tissue or even vascular bed-specific manner.
Collapse
Affiliation(s)
- Alessandra Pasut
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, B-3000, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Lisa M Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, B-3000, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Anne Cuypers
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, B-3000, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, B-3000, Leuven, Belgium.
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium.
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark.
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|