1
|
Sakamuri SS, Sakamuri A. Unlocking hypoglycemia-associated brain microvascular dysfunction: critical insights from proteomic analysis. Neural Regen Res 2025; 20:1707-1708. [PMID: 39104104 PMCID: PMC11688553 DOI: 10.4103/nrr.nrr-d-24-00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/27/2024] [Accepted: 05/23/2024] [Indexed: 08/07/2024] Open
Affiliation(s)
| | - Anil Sakamuri
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
2
|
Boettger P, Schwertz H, Platsch H, Mueller-Werdan U, Werdan K, Buerke M. Intramural Administration of Translational Inhibitor Puromycin Upon Balloon Angioplasty Inhibits SMC Proliferation and Protein Synthesis-Vascular Proteome Profiling Analysis. Proteomics Clin Appl 2025; 19:e202400066. [PMID: 39993432 DOI: 10.1002/prca.202400066] [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: 07/04/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Percutaneous transluminal coronary angioplasty (PTCA) is an effective procedure to decrease the severity of stenotic coronary atherosclerotic lesions. However, its long-term success is limited by the formation of restenosis or neointima by increased proliferation of smooth muscle cells (SMCs) and synthesis of extracellular matrix. Polypeptide growth factors are potent SMC mitogens and are involved in SMC proliferation and extracellular matrix (ECM) synthesis. In this line, inhibition of de novo protein synthesis might be beneficial. METHODS We examined the effects of different concentrations of translational inhibitor puromycin on SMC proliferation and apoptosis, in vitro. Further, we examined the effects of local administration of puromycin in a rabbit balloon injury model of the iliac artery. RESULTS Injection of puromycin or its vehicle was performed with an infusion-balloon catheter directly into the vessel wall during angioplasty. PTA in the vehicle group resulted in neointima formation 3 weeks after the vascular intervention. In contrast, puromycin treatment resulted in a significant reduction of intima-media ratio. We observed decreased elastin and collagen III synthesis in puromycin-treated animals. With proteomics, we could demonstrate reduced protein expression of lamin, vimentin, alpha-1 antitrypsin, alpha-actin allowing puromycin treatment. In in vitro experiments, puromycin decreased SMCs proliferation (i.e., BrdU incorporation) following FCS stimulation. PERSPECTIVE Based on the data from our animal experiments, aministration of puromycin directly into the vessel wall during angioplasty may be effective in preventing or reducing restenosis in humans.
Collapse
Affiliation(s)
- Priyanka Boettger
- Department of Medicine II Cardiology, Intensive Care Medicine, Angiology, St. Marien Hospital, Siegen, Germany
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Hansjörg Schwertz
- Molecular Medicine Program The University of Utah, Salt Lake City, Utah, USA
- Division of Occupational Medicine, The University of Utah, Salt Lake City, Utah, USA
- Department of Occupational Medicine, Billings Clinic Bozeman, Bozeman, Montana, USA
| | | | - Ursula Mueller-Werdan
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Karl Werdan
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Michael Buerke
- Department of Medicine II Cardiology, Intensive Care Medicine, Angiology, St. Marien Hospital, Siegen, Germany
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| |
Collapse
|
3
|
Zhao L, Cao H, Yuan Y, Liao C, Huang D, Li X, Zhao Y, Huang Q, Li S, Zhang B. Annexin A5 knockdown inhibits cardiomyocyte apoptosis and alleviates cardiac hypertrophy via activating the PI3K/AKT/Bcl-2 signaling pathway. Sci Rep 2024; 14:31915. [PMID: 39738388 PMCID: PMC11685421 DOI: 10.1038/s41598-024-83244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/12/2024] [Indexed: 01/02/2025] Open
Abstract
Annexin A5 (ANXA5) is a small calcium-dependent protein that binds specifically to negatively charged phosphatidylserine as a marker of apoptosis. Previous studies have shown that ANXA5 expression is elevated in hypertensive patients and is closely related to left ventricular systolic function in hypertensive patients, but its specific mechanism of action has not been clarified. GEO database analysis showed that ANXA5 expression was significantly upregulated in hypertensive myocardial hypertrophy. The expression of ANXA5 protein and mRNA was overexpressed, and knockdown of ANXA5 can effectively attenuate cardiomyocyte apoptosis and inflammatory response, ameliorate myocardial hypertrophy and cardiac dysfunction in ALD-induced hypertrophic cardiomyocytes and in SHR hypertrophic hearts. Mechanistically, ANXA5 has synergistic effect with intracellular calciumion level. In the meantime, ANXA5 knockdown inhibited cell apoptosis along with a decrease of Bax/Bcl-2 ratio, and induction of PI3K/AKT activation. It should be noted that LY290004 (PI3K/Akt signaling pathway inhibitor) can weaken the inhibitory effect of knockdown ANXA5 on cardiomyocyte apoptosis and myocardial protection. Therefore, ANXA5 knockdown improves hypertensive myocardial hypertrophy and cardiac function by inhibiting apoptosis and inflammatory response and activating PI3K/AKT/Bcl-2 pathway, ANXA5 may be a potential therapeutic direction for the treatment of hypertensive myocardial hypertrophy.
Collapse
Affiliation(s)
- Lina Zhao
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Department of Ultrasound Center, The Affiliated Hospital of Guizhou Medical University, NO.28 Guiyi Street, Guiyang, 550004, Guizhou, China
| | - Hongjuan Cao
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yao Yuan
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Chunyan Liao
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Department of Ultrasound Center, The Affiliated Hospital of Guizhou Medical University, NO.28 Guiyi Street, Guiyang, 550004, Guizhou, China
| | - Dan Huang
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xiaoyi Li
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yueyao Zhao
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Quanfeng Huang
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Sha Li
- Department of Ultrasound Center, The Affiliated Hospital of Guizhou Medical University, NO.28 Guiyi Street, Guiyang, 550004, Guizhou, China.
| | - Bei Zhang
- Guizhou Medical University, Guiyang, 550004, Guizhou, China.
- Department of Ultrasound Center, The Affiliated Hospital of Guizhou Medical University, NO.28 Guiyi Street, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
4
|
Ariyasinghe NR, Gupta D, Escopete S, Rai D, Stotland A, Sundararaman N, Ngu B, Dabke K, McCarthy L, Santos RS, McCain ML, Sareen D, Parker SJ. Identification of Disease-Relevant, Sex-Based Proteomic Differences in iPSC-Derived Vascular Smooth Muscle Cells. Int J Mol Sci 2024; 26:187. [PMID: 39796045 PMCID: PMC11719605 DOI: 10.3390/ijms26010187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
The prevalence of cardiovascular disease varies with sex, and the impact of intrinsic sex-based differences on vasculature is not well understood. Animal models can provide important insights into some aspects of human biology; however, not all discoveries in animal systems translate well to humans. To explore the impact of chromosomal sex on proteomic phenotypes, we used iPSC-derived vascular smooth muscle cells from healthy donors of both sexes to identify sex-based proteomic differences and their possible effects on cardiovascular pathophysiology. Our analysis confirmed that differentiated cells have a proteomic profile more similar to healthy primary aortic smooth muscle cells than iPSCs. We also identified sex-based differences in iPSC-derived vascular smooth muscle cells in pathways related to ATP binding, glycogen metabolic process, and cadherin binding as well as multiple proteins relevant to cardiovascular pathophysiology and disease. Additionally, we explored the role of autosomal and sex chromosomes in protein regulation, identifying that proteins on autosomal chromosomes also show sex-based regulation that may affect the protein expression of proteins from autosomal chromosomes. This work supports the biological relevance of iPSC-derived vascular smooth muscle cells as a model for disease, and further exploration of the pathways identified here can lead to the discovery of sex-specific pharmacological targets for cardiovascular disease.
Collapse
Affiliation(s)
- Nethika R. Ariyasinghe
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Divya Gupta
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Sean Escopete
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Deepika Rai
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Benjamin Ngu
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90007, USA; (B.N.); (M.L.M.)
| | - Kruttika Dabke
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Liam McCarthy
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Roberta S. Santos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (R.S.S.); (D.S.)
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, West Hollywood, CA 90069, USA
| | - Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90007, USA; (B.N.); (M.L.M.)
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dhruv Sareen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (R.S.S.); (D.S.)
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, West Hollywood, CA 90069, USA
- iPSC Core, David and Janet Polak Foundation Stem Cell Core Laboratory, Cedars-Sinai Medical Center, West Hollywood, CA 90069, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sarah J. Parker
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Innovation Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
5
|
Zahid R, Wang J, Cai Z, Ishtiaq A, Liu M, Ma D, Liang Y, Xu Y. Single chain fragment variable, a new theranostic approach for cardiovascular diseases. Front Immunol 2024; 15:1443290. [PMID: 39735545 PMCID: PMC11671482 DOI: 10.3389/fimmu.2024.1443290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/26/2024] [Indexed: 12/31/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain a significant global health challenge, leading to substantial morbidity and mortality. Despite recent advancements in CVD management, pharmaceutical treatments often suffer from poor pharmacokinetics and high toxicity. With the rapid progress of modern molecular biology and immunology, however, single-chain fragment variable (scFv) molecule engineering has emerged as a promising theranostic tool to offer specificity and versatility in targeting CVD-related antigens. To represent the latest development on the potential of scFv in the context of CVDs, this review summarized the new mechanism of action and applications as therapeutic, as well as diagnostic agents. Furthermore, the advantages of scFv, including its small size, ease of modification, and ability to be engineered for enhanced affinity and specificity, are also described. Finally, such challenges as immunogenicity, stability, and scalability, alongside strategies to overcome these hurdles, are deeply scrutinized to provide safer and more effective strategies for the diagnosis and treatment of the incurable CVDs.
Collapse
Affiliation(s)
- Rukhshan Zahid
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Juncheng Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Zecheng Cai
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Ayesha Ishtiaq
- College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Meng Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Dan Ma
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Yan Liang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| |
Collapse
|
6
|
Crocco P, Montesanto A, La Grotta R, Paparazzo E, Soraci L, Dato S, Passarino G, Rose G. The Potential Contribution of MyomiRs miR-133a-3p, -133b, and -206 Dysregulation in Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:12772. [PMID: 39684483 DOI: 10.3390/ijms252312772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiovascular disease (CVD) is a major global health concern. The number of people with CVD is expected to rise due to aging populations and increasing risk factors such as obesity and diabetes. Identifying new molecular markers is crucial for early diagnosis and treatment. Among these, plasma levels of some miRNAs, specifically expressed in cardiac and skeletal muscle, known as myomiRs, have gained attention for their roles in cardiovascular health. This study analyzed the plasma levels of miR-133a-3p, -133b, and -206 in the pathogenesis of cardiovascular diseases. Using a case-control study design with patients recruited from several nursing homes from Calabria (southern Italy) characterized by different types of CVD compared with non-CVD controls, we found downregulation of miR-133a-3p in heart failure and miR-133b in stroke, along with the overall decreased expression of miR-133b and miR-206 in CVD patients, although they showed low specificity as biomarkers of CVD (as based on ROC analysis). In silico functional characterization of their targets and signaling pathways revealed their involvement in critical cardiovascular processes. Although further research is necessary to fully elucidate their mechanisms and clinical utility, the findings reported here may provide insight into the potential contribution of myomiRs in the cardiovascular injury framework, also offering indications for new research directions.
Collapse
Affiliation(s)
- Paolina Crocco
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| | - Rossella La Grotta
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| | - Ersilia Paparazzo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
- Unit of Geriatric Medicine, Italian National Research Center on Aging (INRCA-IRCCS), 87100 Cosenza, Italy
| | - Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center on Aging (INRCA-IRCCS), 87100 Cosenza, Italy
| | - Serena Dato
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
7
|
Moxon JV, Pretorius C, Trollope AF, Mittal P, Klingler-Hoffmann M, Hoffmann P, Golledge J. A systematic review and in silico analysis of studies investigating the ischemic penumbra proteome in animal models of experimental stroke. J Cereb Blood Flow Metab 2024; 44:1709-1722. [PMID: 38639008 PMCID: PMC11504113 DOI: 10.1177/0271678x241248502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Ischaemic stroke results in the formation of a cerebral infarction bordered by an ischaemic penumbra. Characterising the proteins within the ischaemic penumbra may identify neuro-protective targets and novel circulating markers to improve patient care. This review assessed data from studies using proteomic platforms to compare ischaemic penumbra tissues to controls following experimental stroke in animal models. Proteins reported to differ significantly between penumbra and control tissues were analysed in silico to identify protein-protein interactions and over-represented pathways. Sixteen studies using rat (n = 12), mouse (n = 2) or primate (n = 2) models were included. Heterogeneity in the design of the studies and definition of the penumbra were observed. Analyses showed high abundance of p53 in the penumbra within 24 hours of permanent ischaemic stroke and was implicated in driving apoptosis, cell cycle progression, and ATM- MAPK- and p53- signalling. Between 1 and 7 days after stroke there were changes in the abundance of proteins involved in the complement and coagulation pathways. Favourable recovery 1 month after stroke was associated with an increase in the abundance of proteins involved in wound healing. Poor recovery was associated with increases in prostaglandin signalling. Findings suggest that p53 may be a target for novel therapeutics for ischaemic stroke.
Collapse
Affiliation(s)
- Joseph V Moxon
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Cornea Pretorius
- Townsville University Hospital, Angus Smith Drive, Douglas, Townsville, Australia
| | - Alexandra F Trollope
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Parul Mittal
- Mass Spectrometry and Proteomics Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Manuela Klingler-Hoffmann
- Mass Spectrometry and Proteomics Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Peter Hoffmann
- Mass Spectrometry and Proteomics Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
- Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Australia
| |
Collapse
|
8
|
Bihaniya H, Rudraprasad D, Joseph J. Pathobiology of Fungal Endophthalmitis: A Major Review. ACS Infect Dis 2024; 10:3126-3137. [PMID: 39267469 DOI: 10.1021/acsinfecdis.4c00442] [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: 09/17/2024]
Abstract
Fungal endophthalmitis is a chronic inflammatory condition of the eye's posterior segment that can lead to irreversible vision loss. While relatively rare in western countries, its incidence is notably higher in Asia, particularly India. The condition's prevalence is exacerbated by factors such as intravenous drug use, antibiotics, and ocular surgeries. Fungal endophthalmitis can be categorized as endogenous, arising from systemic infection, or exogenous, linked to external sources such as trauma or surgery. The fungal agents responsible vary by region, with Candida species common in the West and Aspergillus and Fusarium species more prevalent in India. Management typically involves vitrectomy and intravitreal antifungal drugs such as amphotericin B and voriconazole, though treatment is often complicated by multidrug resistance and culture-negative cases. Recent proteomic and transcriptomic analyses have highlighted the early and sustained activation of the host immune response during infection involving key inflammatory and oxidative stress-related proteins. Given the potential for excessive inflammation to cause retinal damage, targeted immunotherapies are crucial. Immunomodulation, which aims to balance the immune response, shows promise in preserving vision while effectively combating the infection. Key targets for immunomodulation include pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-17), chemokines (CCL2, CXCL8), Toll-like receptors (TLR2, TLR4), and the complement system. Additionally, modulating the activity of macrophages, neutrophils, regulatory T cells, and Th17 cells, as well as targeting inflammasomes, can help control inflammation. Biologic agents and small molecule inhibitors offer further avenues for precise immune response modulation. This review underscores the importance of a comprehensive understanding of host-pathogen interactions in the development of effective therapies for fungal endophthalmitis.
Collapse
Affiliation(s)
- Himanshi Bihaniya
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
| | - Dhanwini Rudraprasad
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
| |
Collapse
|
9
|
Ariyasinghe NR, Gupta D, Escopete S, Stotland AB, Sundararaman N, Ngu B, Dabke K, Rai D, McCarthy L, Santos RS, McCain ML, Sareen D, Parker SJ. Identification of Disease-relevant, Sex-based Proteomic Differences in iPSC-derived Vascular Smooth Muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605659. [PMID: 39211096 PMCID: PMC11361011 DOI: 10.1101/2024.07.30.605659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The prevalence of cardiovascular disease varies with sex, and the impact of intrinsic sex-based differences on vasculature is not well understood. Animal models can provide important insight into some aspects of human biology, however not all discoveries in animal systems translate well to humans. To explore the impact of chromosomal sex on proteomic phenotypes, we used iPSC-derived vascular smooth muscle cells from healthy donors of both sexes to identify sex-based proteomic differences and their possible effects on cardiovascular pathophysiology. Our analysis confirmed that differentiated cells have a proteomic profile more similar to healthy primary aortic smooth muscle than iPSCs. We also identified sex-based differences in iPSC- derived vascular smooth muscle in pathways related to ATP binding, glycogen metabolic process, and cadherin binding as well as multiple proteins relevant to cardiovascular pathophysiology and disease. Additionally, we explored the role of autosomal and sex chromosomes in protein regulation, identifying that proteins on autosomal chromosomes also show sex-based regulation that may affect the protein expression of proteins from autosomal chromosomes. This work supports the biological relevance of iPSC-derived vascular smooth muscle cells as a model for disease, and further exploration of the pathways identified here can lead to the discovery of sex-specific pharmacological targets for cardiovascular disease. Significance In this work, we have differentiated 4 male and 4 female iPSC lines into vascular smooth muscle cells, giving us the ability to identify statistically-significant sex-specific proteomic markers that are relevant to cardiovascular disease risk (such as PCK2, MTOR, IGFBP2, PTGR2, and SULTE1).
Collapse
|
10
|
Hirsinger E, Blavet C, Bonnin MA, Bellenger L, Gharsalli T, Duprez D. Limb connective tissue is organized in a continuum of promiscuous fibroblast identities during development. iScience 2024; 27:110305. [PMID: 39050702 PMCID: PMC11267076 DOI: 10.1016/j.isci.2024.110305] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Connective tissue (CT), which includes tendon and muscle CT, plays critical roles in development, in particular as positional cue provider. Nonetheless, our understanding of fibroblast developmental programs is hampered because fibroblasts are highly heterogeneous and poorly characterized. Combining single-cell RNA-sequencing-based strategies including trajectory inference and in situ hybridization analyses, we address the diversity of fibroblasts and their developmental trajectories during chicken limb fetal development. We show that fibroblasts switch from a positional information to a lineage diversification program at the fetal period onset. Muscle CT and tendon are composed of several fibroblast populations that emerge asynchronously. Once the final muscle pattern is set, transcriptionally close populations are found in neighboring locations in limbs, prefiguring the adult fibroblast layers. We propose that the limb CT is organized in a continuum of promiscuous fibroblast identities, allowing for the robust and efficient connection of muscle to bone and skin.
Collapse
Affiliation(s)
- Estelle Hirsinger
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Cédrine Blavet
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Marie-Ange Bonnin
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Léa Bellenger
- Sorbonne Université, CNRS FR3631, Inserm U1156, Institut de Biologie Paris Seine (IBPS), ARTbio Bioinformatics Analysis Facility, Paris, Institut Français de Bioinformatique (IFB), 75005 Paris, France
| | - Tarek Gharsalli
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
- Inovarion, 75005 Paris, France
| | - Delphine Duprez
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| |
Collapse
|
11
|
Zha D, Wang S, Monaghan-Nichols P, Qian Y, Sampath V, Fu M. Mechanisms of Endothelial Cell Membrane Repair: Progress and Perspectives. Cells 2023; 12:2648. [PMID: 37998383 PMCID: PMC10670313 DOI: 10.3390/cells12222648] [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: 09/08/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Endothelial cells are the crucial inner lining of blood vessels, which are pivotal in vascular homeostasis and integrity. However, these cells are perpetually subjected to a myriad of mechanical, chemical, and biological stresses that can compromise their plasma membranes. A sophisticated repair system involving key molecules, such as calcium, annexins, dysferlin, and MG53, is essential for maintaining endothelial viability. These components orchestrate complex mechanisms, including exocytosis and endocytosis, to repair membrane disruptions. Dysfunctions in this repair machinery, often exacerbated by aging, are linked to endothelial cell death, subsequently contributing to the onset of atherosclerosis and the progression of cardiovascular diseases (CVD) and stroke, major causes of mortality in the United States. Thus, identifying the core machinery for endothelial cell membrane repair is critically important for understanding the pathogenesis of CVD and stroke and developing novel therapeutic strategies for combating CVD and stroke. This review summarizes the recent advances in understanding the mechanisms of endothelial cell membrane repair. The future directions of this research area are also highlighted.
Collapse
Affiliation(s)
- Duoduo Zha
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
- The National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Rd, Honggu District, Nanchang 330031, China;
| | - Shizhen Wang
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA;
| | - Paula Monaghan-Nichols
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
| | - Yisong Qian
- The National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Rd, Honggu District, Nanchang 330031, China;
| | - Venkatesh Sampath
- Department of Pediatric, Children’s Mercy Hospital, Children’s Mercy Research Institute, Kansas City, MO 64108, USA;
| | - Mingui Fu
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
| |
Collapse
|
12
|
Wang C, Paiva TO, Motta C, Speziale P, Pietrocola G, Dufrêne YF. Catch Bond-Mediated Adhesion Drives Staphylococcus aureus Host Cell Invasion. NANO LETTERS 2023. [PMID: 37267288 DOI: 10.1021/acs.nanolett.3c01387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Various viruses and pathogenic bacteria interact with annexin A2 to invade mammalian cells. Here, we show that Staphylococcus aureus engages in extremely strong catch bonds for host cell invasion. By means of single-molecule atomic force microscopy, we find that bacterial surface-located clumping factors bind annexin A2 with extraordinary strength, indicating that these bonds are extremely resilient to mechanical tension. By determining the lifetimes of the complexes under increasing mechanical stress, we demonstrate that the adhesins form catch bonds with their ligand that are capable to sustain forces of 1500-1700 pN. The force-dependent adhesion mechanism identified here provides a molecular framework to explain how S. aureus pathogens tightly attach to host cells during invasion and shows promise for the design of new therapeutics against intracellular S. aureus.
Collapse
Affiliation(s)
- Can Wang
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Telmo O Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Chiara Motta
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Pietro Speziale
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Giampiero Pietrocola
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
13
|
Dawood S, Ali ZJ. Entrectinib for NTRK Fusion-Positive Metastatic Melanoma Progressing on Combined PD-1 and CTLA-4 Inhibition: A Case Report. Case Rep Oncol 2023; 16:1451-1459. [PMID: 38028569 PMCID: PMC10673342 DOI: 10.1159/000534475] [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: 04/30/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The burden of melanoma is increasing globally. Despite the use of immunotherapy and targeted therapy, the prognosis of metastatic melanoma remains relatively poor. The integration of comprehensive molecular profiling can lead to the detection of actionable biomarkers and the expansion of treatment options, thereby prolonging cancer patient survival. Case Presentation We herein present the case of a female 54-year-old patient diagnosed with melanoma of the right knee, for which she underwent surgery. Patient showed progression of disease after 10 cycles of adjuvant nivolumab. Ipilimumab (1 mg/kg every 3 weeks) was added to the treatment regimen but no clinical improvement was observed. Molecular profiling was conducted based on patient tissue, and an ANXA2-NTRK3 fusion was detected in the tumor. This specific fusion has not been previously reported; however, it is in-frame and similar to other known oncogenic NTRK fusions. At the time of entrectinib initiation, the patient had clear disease progression on the right leg on standard of care immunotherapy. She was commenced on entrectinib 200 mg once daily for 2 weeks. Dose escalation was attempted, and treatment intensity was managed based on drug tolerability. Good treatment response was observed on laboratory and radiologic parameters. As of September 2023, i.e., 2.5 years after treatment initiation, patient disease continues to be controlled with entrectinib. Conclusion Profiling of advanced tumors is important to determine the presence of agnostic markers that can be targeted and ultimately improve the prognostic outcome of patients after the failure of standard of care.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Adjunct Clinical Professor, Mohammed Bin Rashid University of Medicine and Health Sciences, Consultant Medical Oncologist, Dubai, UAE
| | | |
Collapse
|