1
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Zhao Y, Zhang Y, Liu X, Zhang J, Gao Y, Li S, Chang C, Liu X, Yang G. Comparative proteomic analysis of plasma exosomes reveals the functional contribution of N-acetyl-alpha-glucosaminidase to Parkinson's disease. Neural Regen Res 2025; 20:2998-3012. [PMID: 38993127 PMCID: PMC11826475 DOI: 10.4103/nrr.nrr-d-23-01500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/12/2024] [Accepted: 04/08/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00029/figure1/v/2024-11-26T163120Z/r/image-tiff Parkinson's disease is the second most common progressive neurodegenerative disorder, and few reliable biomarkers are available to track disease progression. The proteins, DNA, mRNA, and lipids carried by exosomes reflect intracellular changes, and thus can serve as biomarkers for a variety of conditions. In this study, we investigated alterations in the protein content of plasma exosomes derived from patients with Parkinson's disease and the potential therapeutic roles of these proteins in Parkinson's disease. Using a tandem mass tag-based quantitative proteomics approach, we characterized the proteomes of plasma exosomes derived from individual patients, identified exosomal protein signatures specific to patients with Parkinson's disease, and identified N-acetyl-alpha-glucosaminidase as a differentially expressed protein. N-acetyl-alpha-glucosaminidase expression levels in exosomes from the plasma of patients and healthy controls were validated by enzyme-linked immunosorbent assay and western blot. The results demonstrated that the exosomal N-acetyl-alpha-glucosaminidase concentration was not only lower in Parkinson's disease, but also decreased with increasing Hoehn-Yahr stage, suggesting that N-acetyl-alpha-glucosaminidase could be used to rapidly evaluate Parkinson's disease severity. Furthermore, western blot and immunohistochemistry analysis showed that N-acetyl-alpha-glucosaminidase levels were markedly reduced both in cells treated with 1-methyl-4-phenylpyridinium and cells overexpressing α-synuclein compared with control cells. Additionally, N-acetyl-alpha-glucosaminidase overexpression significantly increased cell viability and inhibited α-synuclein expression in 1-methyl-4-phenylpyridinium-treated cells. Taken together, our findings demonstrate for the first time that exosomal N-acetyl-alpha-glucosaminidase may serve as a biomarker for Parkinson's disease diagnosis, and that N-acetyl-alpha-glucosaminidase may reduce α-synuclein expression and 1-methyl-4-phenylpyridinium-induced neurotoxicity, thus providing a new therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yidan Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xin Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jian Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ya Gao
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Shuyue Li
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Cui Chang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiang Liu
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Guofeng Yang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
3
|
Wan C, Xu Y, Zhu Y, Cao X, Wang P, Gu Y. NLRP3 inflammasome expression affects immune cell infiltration and clinical prognosis in Helicobacter pylori infection‑associated gastric cancer. Mol Med Rep 2025; 32:185. [PMID: 40314099 PMCID: PMC12059518 DOI: 10.3892/mmr.2025.13550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/26/2025] [Indexed: 05/03/2025] Open
Abstract
High Helicobacter pylori infection rates contribute to high gastric cancer (GC) incidence. While H. pylori infection is associated with GC development its mechanisms are still being studied. The aim of the present study was to examine the differences between H. pylori infection‑induced GC and non‑infected tissues, and to investigate the correlation between nucleotide‑binding oligomerization domain, leucine rich repeat and pyrin domain containing 3 (NLRP3) inflammasome expression and immune cell infiltration in GC, thus providing a theoretical basis for clinical prognosis and immunotherapy. High‑throughput RNA‑sequencing expression data from The Cancer Genome Atlas (TCGA) were analyzed. Additionally, TIMER2.0 and Kaplan‑Meier Plotter were used to analyze the differential expression of NLRP3 mRNA in various tumors, the effect of H. pylori infection on gene expression, and the association between NLRP3 and clinical prognosis among patients with GC. Immunohistochemistry (IHC) was used to assess the effects of NLRP3 protein expression on immune cell infiltration in clinical tissues with or without H. pylori infection. R software was used for data visualization and statistical analysis. TCGA data revealed that the expression levels of NLRP3 in GC tissues were increased compared with those in normal tissues (P<0.05), which was further validated in clinical samples. Furthermore, NLRP3 mRNA expression was significantly elevated in clinical GC tissues infected with H. pylori. Notably higher relative levels of NLRP3 mRNA were observed in tumor tissues with a tumor size ≥5 cm, lymph node metastasis, Tumor‑Node‑Metastasis stage III + IV or poor differentiation compared with the respective controls (P<0.05). IHC confirmed a significant increase in NLRP3 expression within H. pylori‑infected GC tissues compared with that in non‑infected tissues. In GC immune infiltration, NLRP3 expression was revealed to be associated with natural killer cell, whereas it was negatively correlated with regulatory T cells and CD8+ T cells. These findings indicated that NLRP3 may promote the polarization of tumor‑associated macrophages towards the M2 phenotype. High NLRP3 expression also promoted the infiltration of CD3+ and CD206+ cells, which significantly affected the survival rate of patients with GC. The immune infiltration of regulatory T lymphocytes was associated with better survival benefits for patients with GC; however, M2 macrophage infiltration was not conducive to the survival of patients with GC. Furthermore, survival analysis showed that high expression of NLRP3 was associated with a poorer 5‑year overall survival, progression‑free survival and post‑progression survival rates. In conclusion, elevated NLRP3 expression, which may be induced by H. pylori infection, could promote immune cell infiltration potentially by regulating cancer cell proliferation and migration, ultimately leading to an unfavorable prognosis and a notable reduction in the 5‑year survival rate.
Collapse
Affiliation(s)
- Chuandan Wan
- Central Laboratory, Changshu Medical Examination Institute, Changshu, Jiangsu 215500, P.R. China
| | - Yeqiong Xu
- Central Laboratory, Changshu Medical Examination Institute, Changshu, Jiangsu 215500, P.R. China
| | - Yanping Zhu
- Central Laboratory, Changshu Medical Examination Institute, Changshu, Jiangsu 215500, P.R. China
| | - Xuexian Cao
- Department of Oncology and Radiotherapy, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| | - Ping Wang
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yulan Gu
- Department of Oncology and Radiotherapy, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| |
Collapse
|
4
|
Fan F, Guo R, Pan K, Xu H, Chu X. Mucus and mucin: changes in the mucus barrier in disease states. Tissue Barriers 2025:2499752. [PMID: 40338015 DOI: 10.1080/21688370.2025.2499752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
In this review we discuss mucus, the viscoelastic secretion from goblet or mucous producing cells that covers and protects all non-keratinized wet epithelial surfaces. In addition to the surface of organs directly contacting with the external environment such as the eyes, this layer provides protection to the underlying gastrointestinal, respiratory and female reproductive tracts by trapping pathogens, irritants, environmental fine particles and potentially harmful foreign substances. Mucins, the primary structural components of mucus, form structurally different mucus layers at different sites in a process regulated by a variety of factors. Currently, more and more studies have shown that the mucus barrier is not only closely related to various intestinal mucus diseases, but also involved in the occurrence and development of various airway diseases and mucus-related diseases, thus it may become a new target for the treatment of various related diseases in the future. Since the dysfunction of the mucous layer is closely related to various pathological processes, in-depth understanding of its molecular mechanism and physiological role is of great theoretical and practical significance for disease prevention and treatment. Here, we discuss different aspects of the mucus layer by focusing on its chemical composition, synthetic pathways, and some of the characteristics of the mucus layer in physiological and pathological situations.
Collapse
Affiliation(s)
- Fangfang Fan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ruihan Guo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Kun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hongye Xu
- Quality Assurance department, Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui Province, China
| |
Collapse
|
5
|
Phi LTH, Cheng Y, Funakoshi Y, Bertucci F, Finetti P, Van Laere SJ, Zou F, Long JP, Ogata S, Krishnamurthy S, Reuben JM, Foulks JM, Warner SL, Rosenbluth JM, Sood AK, Tripathy D, Ueno NT, Wang X. AXL promotes inflammatory breast cancer progression by regulating immunosuppressive macrophage polarization. Breast Cancer Res 2025; 27:70. [PMID: 40329335 PMCID: PMC12057249 DOI: 10.1186/s13058-025-02015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are key promoters of inflammatory breast cancer (IBC), the most aggressive form of breast cancer. The receptor tyrosine kinase AXL is highly expressed in various cancer types, including IBC, but its role in TAMs remains unexplored. METHODS We examined the effects of AXL inhibitor TP-0903 on tumor growth and tumor microenvironment (TME) component M2 macrophages (CD206+) in IBC and triple-negative breast cancer mouse models using flow cytometry and immunohistochemical staining. Additionally, we knocked out AXL expression in human THP-1 monocytes and evaluated the effect of AXL signaling on immunosuppressive M2 macrophage polarization and IBC cell growth and migration. We then investigated the underlying mechanisms through RNA sequencing analysis. Last, we performed CIBERSORT deconvolution to analyze the association between AXL expression and tumor-infiltrating immune cell types in tumor samples from the Inflammatory Breast Cancer International Consortium. RESULTS We found that inhibiting the AXL pathway significantly reduced IBC tumor growth and decreased CD206+ macrophage populations within tumors. Mechanistically, our in vitro data showed that AXL promoted M2 macrophage polarization and enhanced the secretion of immunosuppressive chemokines, including CCL20, CCL26, and epiregulin, via the transcription factor STAT6 and thereby accelerated IBC cell growth and migration. RNA sequencing analysis further indicated that AXL signaling in immunosuppressive M2 macrophages regulated the expression of molecules and cytokines, contributing to an immunosuppressive TME in IBC. Moreover, high AXL expression was correlated with larger populations of immunosuppressive immune cells but smaller populations of immunoactive immune cells in tissues from patients with IBC. CONCLUSIONS AXL signaling promotes IBC growth by inducing M2 macrophage polarization and driving the secretion of immunosuppressive molecules and cytokines via STAT6 signaling, thereby contributing to an immunosuppressive TME. Collectively, these findings highlight the potential of targeting AXL signaling as a novel therapeutic approach for IBC that warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Lan T H Phi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yating Cheng
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yohei Funakoshi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francois Bertucci
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, Marseille, France
| | - Pascal Finetti
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Steven J Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Fang Zou
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suguru Ogata
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
- University of Hawai'i Inflammatory Breast Cancer Clinic and Research Program, Honolulu, HI, USA
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Jennifer M Rosenbluth
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA.
- University of Hawai'i Inflammatory Breast Cancer Clinic and Research Program, Honolulu, HI, USA.
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA.
- University of Hawai'i Inflammatory Breast Cancer Clinic and Research Program, Honolulu, HI, USA.
| |
Collapse
|
6
|
Adamis KS, Georgoulakis M, Angelonidis I, Korovesis D, Papadopoulos C, Kapsalis M, Tavernarakis N, Eleftheriadis N, Neochoritis CG. The Evolution of Fluorescein into A Potential Theranostic Tool. Chemistry 2025:e202501513. [PMID: 40317604 DOI: 10.1002/chem.202501513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Recent advances in drug discovery and development have been marked by the emergence of new modalities, including small molecule theranostic agents. While initial results from clinical trials have been promising, modern detectable inhibitors are still in an early stage of development. In this study, we present a strategy for chemically evolving a fluorescent imaging agent into a potent therapeutic entity, which not only retains its properties but also enhances its inhibition and detection applicability. By utilizing 15-LOX-1 as a model system, we leverage prior knowledge of its inhibitors to rationally functionalize fluorescein, enabling the targeted and highly efficient synthesis of over 20 derivatives across four different scaffolds. This approach ultimately led to the development of a potent, cell-permeable inhibitor that effectively engages its target in live cells and enables real-time visualization. These findings validate our new strategy for the development of small molecule diagnostic modulators, paving the way for application in other targets as well.
Collapse
Affiliation(s)
| | | | | | - Dimitris Korovesis
- Foundation for Research and Technology Hellas, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| | | | - Michael Kapsalis
- Department of Chemistry, University of Crete, Voutes, Heraklion, Greece
| | - Nektarios Tavernarakis
- Foundation for Research and Technology Hellas, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
- Division of Basic Sciences, School of Medicine, University of Crete, Voutes, Heraklion, Greece
| | | | | |
Collapse
|
7
|
Marrone MT, Reuss JE, Crawford A, Neelon B, Liu JO, Brahmer JR, Platz EA. Statin Use With Immune Checkpoint Inhibitors and Survival in Nonsmall Cell Lung Cancer. Clin Lung Cancer 2025; 26:201-209. [PMID: 39818516 PMCID: PMC12037305 DOI: 10.1016/j.cllc.2024.12.008] [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: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
OBJECTIVE To determine the association between concurrent statin use with immune checkpoint inhibitors (ICIs) and lung cancer-specific and overall mortality in patients with nonsmall cell lung cancer (NSCLC). MATERIALS AND METHODS SEER-Medicare was used to conduct a retrospective study of Medicare beneficiaries ≥65 years of age diagnosed with NSCLC between 2007 and 2017 treated with an ICI. Patients were followed from date of first ICI claim until death, 1 month from last ICI claim, or 12/31/2018, whichever came first. Associations for time-updated statin use and lung cancer-specific mortality, and overall mortality were estimated using Cox models adjusted for demographic, pathological, treatment-related factors, and a propensity score for statin use. RESULTS Among 1,401 patients, concurrent statin use with any ICI was associated with 41% lower risk of lung-cancer specific mortality compared to patients receiving ICI not using a statin (HR = 0.59; 95% CI = 0.35-0.99). Statin use was associated with a similarly lower risk of overall mortality (HR = 0.62; 95% CI = 0.41-0.94). Consistent inverse associations were observed when restricting to PD-1 inhibitors and by statin type. Limited anti-PD-L1 treatment prevented analysis in this subgroup. CONCLUSION Concurrent statin use with ICIs was associated with lower risk of lung cancer-specific and overall mortality in a population-based sample of older patients with NSCLC. Future work is needed to confirm these findings in prospective studies and randomized trials, including evaluating concurrent statin use with frontline ICIs, deciphering the underlying mechanism, and determining the optimal statin-ICI combination that maximize clinical benefit.
Collapse
Affiliation(s)
- Michael T Marrone
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC.
| | - Joshua E Reuss
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Anna Crawford
- Hollings Cancer Center Biostatistics Shared Resource, Medical University of South Carolina, Charleston, SC
| | - Brian Neelon
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC; Hollings Cancer Center Biostatistics Shared Resource, Medical University of South Carolina, Charleston, SC
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Julie R Brahmer
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
8
|
Zhang Z, She L, Bai M. Efficacy of exosomes in acute kidney injury treatment and the associated mechanism (Review). Mol Med Rep 2025; 31:137. [PMID: 40145555 PMCID: PMC11963750 DOI: 10.3892/mmr.2025.13503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/31/2025] [Indexed: 03/28/2025] Open
Abstract
Acute kidney injury (AKI) is a syndrome characterized by rapid loss of renal function with a high morbidity and mortality. However, due to the complex pathophysiologic mechanisms of AKI, no specific treatment for this disease is currently available. Animal models have demonstrated the protective effects of exosomes on AKI; however, the underlying mechanisms require further investigation. The present review focuses on the efficacy of exosomes derived from different cell sources, including mesenchymal stem cells, endothelial progenitor cells and tubular epithelial cells, in the treatment of AKI and the associated mechanism. Furthermore, the effects of exosomal contents, including microRNAs, circular RNAs, long non‑coding RNAs, messenger RNAs and proteins, on the repair of renal tubules, protection against renal tubular epithelial cell injury, protection against fibrosis, inhibition of early endoplasmic reticulum stress and mediation of inflammation during AKI are also summarized in the present review.
Collapse
Affiliation(s)
- Zehao Zhang
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lecheng She
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ming Bai
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
9
|
Fei X, Li N, Xu X, Zhu Y. Macrophage biology in the pathogenesis of Helicobacter pylori infection. Crit Rev Microbiol 2025; 51:399-416. [PMID: 39086061 DOI: 10.1080/1040841x.2024.2366944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Infection with H. pylori induces chronic gastric inflammation, progressing to peptic ulcer and stomach adenocarcinoma. Macrophages function as innate immune cells and play a vital role in host immune defense against bacterial infection. However, the distinctive mechanism by which H. pylori evades phagocytosis allows it to colonize the stomach and further aggravate gastric preneoplastic pathology. H. pylori exacerbates gastric inflammation by promoting oxidative stress, resisting macrophage phagocytosis, and inducing M1 macrophage polarization. M2 macrophages facilitate the proliferation, invasion, and migration of gastric cancer cells. Various molecular mechanisms governing macrophage function in the pathogenesis of H. pylori infection have been identified. In this review, we summarize recent findings of macrophage interactions with H. pylori infection, with an emphasis on the regulatory mechanisms that determine the clinical outcome of bacterial infection.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Yang Y, He Y, Xing H, Zhao Z, Wang J, Li S, Xu X, Ma D, Hu Y. Hyaluronic acid-liposomes hybridized with HucMSC exosomes for enhanced exosomes transdermal delivery and acute skin photodamage repair. Int J Biol Macromol 2025; 306:141606. [PMID: 40024399 DOI: 10.1016/j.ijbiomac.2025.141606] [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: 10/09/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Long-term exposure to ultraviolet (UV) radiation can damage human skin, resulting in photodamage. Repairing photodamaged skin has been a major focus of research in recent years. Extensive research has shown that human umbilical cord mesenchymal stem cell-derived exosomes (Exo) possess anti-inflammatory, pro-angiogenic, and wound healing properties, holding great potential for treating skin damage. However, due to the limitations of exosomes alone, such as poor transdermal penetration, instability, and low utilization, there is an urgent need for new delivery strategies. We designed a hybrid nanovesicle (HL@Exo) by combining ultrasonic incubation with membrane extrusion to fuse Exo with HL. HL@Exo capitalizes on the advantages of liposomal carriers and the permeation-enhancing properties of hyaluronic acid to effectively facilitate transdermal delivery of Exo. The successful fusion of HL@Exo and its skin penetration were verified by methods such as fluorescent labeling, western blotting, Transwell assays, and in vivo imaging. In vitro studies on photodamaged keratinocytes and endothelial cells demonstrated HL@Exo ability to promote cell proliferation, repair, angiogenesis, and reduce inflammation. In a laser-induced skin photodamage model, HL@Exo enhanced collagen regeneration, accelerated wound healing, and demonstrated significant anti-inflammatory effects, indicating its potential as a non-invasive treatment and offering a novel strategy for the clinical application of Exo.
Collapse
Affiliation(s)
- Yuhui Yang
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510235, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yong He
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Hui Xing
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Ziyi Zhao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Jianjin Wang
- Honest Medical China Co., Ltd, Zhuhai 519000, China
| | - Shanying Li
- Honest Medical China Co., Ltd, Zhuhai 519000, China
| | - Xiaosong Xu
- Hengqin Perfect-Medical Laboratory Co. Ltd, Zhuhai 519000, China
| | - Dong Ma
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510235, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Biomedical Engineering, Jinan University, Guangzhou 510632, China; MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China.
| | - Yunfeng Hu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
11
|
Keshtkar S, Asvar Z, Najafi H, Heidari M, Kaviani M, Sarvestani FS, Tamaddon AM, Sadati MS, Hamidizadeh N, Azarpira N. Exosomes as natural vectors for therapeutic delivery of bioactive compounds in skin diseases. Front Pharmacol 2025; 16:1485769. [PMID: 40356952 PMCID: PMC12066514 DOI: 10.3389/fphar.2025.1485769] [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: 08/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Skin diseases are a broad category of diseases and each has complex conditions, which makes it challenging for dermatologists to provide targeted treatment. Exosomes are natural vesicles secreted by cells and play a key role in cell communication. Due to their unique characteristics, including inherent stability, minimal immunogenicity, high biocompatibility, and exceptional ability to penetrate cells, exosomes are being explored as potential delivery vehicles for therapeutics across various diseases including skin problems. Utilizing exosomes for drug delivery in skin diseases can improve treatment outcomes and reduce the side effects of traditional drug delivery methods. Indeed, exosomes can be engineered or utilized as an innovative approach to deliver therapeutic agents such as small molecule drugs, genes, or proteins specifically to affected skin cells. In addition to targeting specific skin cells or tissues, these engineered exosome-based nanocarriers can reduce off-target effects and improve drug efficacy. Hence, this article highlights the transformative potential of this technology in revolutionizing drug delivery in dermatology and improving patient outcomes.
Collapse
Affiliation(s)
- Somayeh Keshtkar
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Asvar
- Nanotechnology School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haniyeh Najafi
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Sadat Sadati
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrin Hamidizadeh
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Lozano-López DA, Hernández-Ortega LD, González-Mariscal L, Díaz-Coránguez M, Pinto-Dueñas DC, Castañeda-Arellano R. Preserving Blood-Brain Barrier Integrity in Ischemic Stroke: a Review on MSCs-sEVs Content and Potential Molecular Targets. Mol Neurobiol 2025:10.1007/s12035-025-04956-9. [PMID: 40259172 DOI: 10.1007/s12035-025-04956-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/23/2025]
Abstract
Ischemic stroke (IS) is a life-threatening condition that constitutes the second leading cause of death globally. Despite its high impact on public health, there is a shortage of treatments due to the complexity of the cellular and molecular mechanisms implicated. One main limiting factor for successful IS therapeutic intervention is stroke-induced blood-brain barrier (BBB) damage, particularly over tight junction proteins (TJs). BBB disruption is a well-established feature of IS, accelerating ischemic tissue damage and worsening prognosis. In recent years, mesenchymal stem cells (MSCs) and their small extracellular vesicles (MSCs-sEVs) have emerged as promising therapeutic interventions for several neurological disorders, including IS. However, its effects on BBB repair after IS are not completely understood. In this review, we will discuss novel experimental evidence of MSCs-sEVs effects in BBB protection and highlight the relevance of molecules reported in MSCs-sEVs, their potential cellular and molecular targets, and putative mechanisms implicated in BBB repair, providing a promising research avenue that may translate into effective therapeutic strategies for IS.
Collapse
Affiliation(s)
- David Arturo Lozano-López
- Pharmacology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México
| | - Luis Daniel Hernández-Ortega
- Molecular Biology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México
| | - Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Mónica Díaz-Coránguez
- Department of Pharmacobiology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Diana Cristina Pinto-Dueñas
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Rolando Castañeda-Arellano
- Pharmacology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México.
| |
Collapse
|
13
|
Chu T, Xiao Z, Xun C, Yang C, Lu M, Wang Y, Chen H, Chen P. Peptidomic profiling of mesenchymal stem cell-derived extracellular vesicles and anti-inflammatory activity of degraded peptides. Int Immunopharmacol 2025; 152:114452. [PMID: 40096816 DOI: 10.1016/j.intimp.2025.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Mesenchymal stem cell derived extracellular vesicles (MSC-EVs) are key paracrine mediators involved in various autoimmune diseases. While current research on EVs predominantly focuses on their protein and nucleic acid components, small peptides received less attention. In this study, we found IFN-γ-treated MSC-EVs, as engineered EVs, exhibit better anti-inflammatory effects both in vitro and in vivo. Through LC-MS/MS and bioinformatics analysis, we identified four peptides-C3-1, C3-2, B2M-1, and IFIT3-1-that are highly expressed in IFN-γ-treated MSCs-EVs. These peptides significantly mitigate the proliferation inhibition of HUVEC cells induced by H₂O₂ and enhance their migratory capacity. Furthermore, they downregulate the expression of inflammatory cytokines TNF-α and IL-6 in H₂O₂-induced oxidative stress models of HUVEC and LPS-induced inflammatory models of RAW264.7 macrophages. The peptides also upregulate p-AKT and HIF-1α, with C3-1 demonstrating superior anti-inflammatory efficacy in both cell models. Consistent with the in vitro findings, in vivo experiments revealed that C3-1 reduces LPS-induced inflammatory cell infiltration in liver tissue and restores hepatocyte structural integrity, as evidenced by HE-stained liver sections. Western blot analysis further confirmed that C3-1 upregulates p-AKT expression and suppresses inflammatory cytokines. Collectively, these findings suggest that C3-1 exerts its anti-inflammatory effects via activation of the AKT signaling pathway and regulation of TNF-α and IL-6. This study not only highlights the anti-inflammatory potential of IFN-γ-treated MSC-derived EVs but also identifies C3-1 as a promising candidate for anti-inflammatory drug development. Notably, this is the first study to identify degraded peptides within EVs, providing a foundation for future research in this area.
Collapse
Affiliation(s)
- Tianqi Chu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Zixuan Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Chengfeng Xun
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Hunan Academy of Forestry, Changsha 410081, China
| | - Chunyan Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Mengqi Lu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuqiu Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Haiyan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; East China Institute of Digital Medical Engineering, Shangrao 334000, China.
| | - Ping Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
14
|
Duan Y, Wu Y, Tian J, Yin Y, Yuan Z, Zhu W, Zhou S, Li C, Feng S. Elucidation of the mechanism Underlying the promotion of ferroptosis and enhanced antitumor immunity by citrus polymethoxyflavones in CRC cells. Front Pharmacol 2025; 16:1571178. [PMID: 40290432 PMCID: PMC12021823 DOI: 10.3389/fphar.2025.1571178] [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: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background Colon cancer is a prevalent condition with a high mortality rate on a global scale. Research has indicated that Citrus polymethoxyflavones (PMFs), a class of flavonoids found in Citrus, possess the potential to demonstrate anti-tumor efficacy. Ferroptosis a form of cell death that is dependent on iron accumulation and lipid peroxidation. Immunotherapy is one of the most commonly used anti-tumor modalities in a clinical setting. Consequently, studies on the pharmacodynamic mechanism of Citrus to determine whether it can modulate tumor immunity through ferroptosis provide new ideas for the clinical treatment of colon cancer. Purpose The objective of this study is to ascertain whether Citrus inhibits PD-L1 through ferroptosis and promotes tumor immunity among patients with colon cancer. Methods The inhibitory effect of PMFs on colon cancer was proved by in vitro experiment and in vivo model. In addition, the occurrence of ferroptosis was detected by measuring key ferroptosis indicators. Bioinformatics analysis was then performed to identify the crossover genes for Citrus polymethoxylflavonoids, colon cancer, and ferroptosis. Finally, key genes were identified by immunocorrelation analysis including WB, Q-PCR and flow cytometry. These experiments were designed to reveal the potential mechanisms of PMFs on ferroptosis and anti-tumor immunity. Results In vitro cell proliferation experiment and the growth of transplanted tumor mice showed that PMFs had inhibitory effect on colon cancer. In addition, the change of ferroptosis index showed that PMFs promoted the occurrence of ferroptosis, followed by Q-PCR and WB detection of NOX4 and TIMP1, the key genes screened by bioinformatics, found that PMFs inhibited PD-L1 by down-regulating TIMP1, thus affecting colon cancer. Flow cytometry showed that CD4+ T expression increased and CD8+ T cell expression decreased after treatment, suggesting that anti-tumor immunity was activated. Conclusion It is conceivable that the tumor immune microenvironment may be subject to regulation during the inhibition of colon cancer through ferroptosis in PMFs. The ferroptosis-related gene TIMP1 has been observed to regulate PD-L1, thereby promoting anti-tumor immunity in colon cancer. However, further investigation is required to ascertain the underlyingprecise mechanisms.
Collapse
Affiliation(s)
- Yingying Duan
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Wu
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiaqi Tian
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuqin Yin
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhongwen Yuan
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenting Zhu
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Suyue Zhou
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chen Li
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Senling Feng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Mafakheri A, Fathi F, Majidpoor J, Moayeri H, Mortezaee K. Secretory exosomes from modified immune cells against cancer. Med Oncol 2025; 42:159. [PMID: 40208472 DOI: 10.1007/s12032-025-02706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025]
Abstract
Extracellular vesicles (EVs) play significant roles in cancer progression through mediating inter/intra cellular communications within tumor microenvironment (TME). EVs are used as non-invasive diagnostic tools, drug delivery systems, and cancer vaccines, considering the anti-tumor potential, safety, biocompatibility and physiochemical stability of endogenous EVs. Modification of immune cells, either genetically or epigenetically, is a growing field of cancer research with the goal of enhancing efficacy of immunotherapy. This review focuses on the possibility of manipulating immune cells including dendritic cells (DCs), natural killer (NK) cells and T cells to secrete EVs that exert immune function either by activating immune responses or altering immune cell behavior to enhance anti-tumor efficacy, and discusses potential obstacles and recommendations for improved functionality of this therapeutic method.
Collapse
Affiliation(s)
- Asrin Mafakheri
- Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Fardin Fathi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Hasan Moayeri
- Department of General Surgery, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
16
|
Liao Z, Zeng J, Lin A, Zou Y, Zhou Z. Pre-treated mesenchymal stem cell-derived exosomes: A new perspective for accelerating spinal cord injury repair. Eur J Pharmacol 2025; 992:177349. [PMID: 39921061 DOI: 10.1016/j.ejphar.2025.177349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Spinal cord injury (SCI) is a devastating event for the central nervous system (CNS), often resulting in the loss of sensory and motor functions. It profoundly affects both the physiological and psychological well-being of patients, reducing their quality of life while also imposing significant economic pressure on families and the healthcare system. Due to the complex pathophysiology of SCI, effective treatments for promoting recovery remain scarce. Mesenchymal stem cell-derived exosomes (MSC-Exos) offer advantages such as low immunogenicity, good biocompatibility, and the ability to cross the blood-spinal cord barrier (BSCB). In preclinical studies, they have progressively shown efficacy in promoting SCI repair and functional recovery. However, the low yield and insufficient targeting of MSC-Exos limit their therapeutic efficacy. Currently, genetic engineering and other preprocessing techniques are being employed to optimize both the yield and functional properties of exosomes, thereby enhancing their therapeutic potential. Therefore, this paper provides an overview of the pathophysiology of SCI and the biogenesis of exosomes. It also summarizes current approaches to optimizing exosome performance. Additionally, it details the mechanisms through which optimized exosomes provide neuroprotection and explores the potential of combined treatments involving MSC-Exos and hydrogels.
Collapse
Affiliation(s)
- Zhiqiang Liao
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Junjian Zeng
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Aiqing Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Yu Zou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Zhidong Zhou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
17
|
Eerdekens H, Pirlet E, Willems S, Bronckaers A, Pincela Lins PM. Extracellular vesicles: innovative cell-free solutions for wound repair. Front Bioeng Biotechnol 2025; 13:1571461. [PMID: 40248643 PMCID: PMC12003306 DOI: 10.3389/fbioe.2025.1571461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Chronic non-healing wounds are often associated with conditions such as diabetes and peripheral vascular disease, pose significant medical and socioeconomic challenges. Cell-based therapies have shown promise in promoting wound healing but have major drawbacks such as immunogenicity and tumor formation. As a result, recent research has shifted to the potential of extracellular vesicles (EVs) derived from these cells. EVs are nanosized lipid bilayer vesicles, naturally produced by all cell types, which facilitate intercellular communication and carry bioactive molecules, offering advantages such as low immunogenicity, negligible toxicity and the potential to be re-engineered. Recent evidence recognizes that during wound healing EVs are released from a wide range of cells including immune cells, skin cells, epithelial cells and platelets and they actively participate in wound repair. This review comprehensively summarizes the latest research on the function of EVs from endogenous cell types during the different phases of wound healing, thereby presenting interesting therapeutic targets. Additionally, it gives a critical overview of the current status of mesenchymal stem cell-derived EVs in wound treatment highlighting their tremendous therapeutic potential as a non-cellular of-the-shelf alternative in wound care.
Collapse
Affiliation(s)
- Hanne Eerdekens
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Elke Pirlet
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Sarah Willems
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Annelies Bronckaers
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Paula M. Pincela Lins
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
- Flemish Institute for Technological Research (VITO), Environmental Intelligence Unit, Mol, Belgium
| |
Collapse
|
18
|
McDonald HG, Reagan AM, Faisal ASM, Goettl R, Wang C, Hoyd R, Spakowicz D, Evers BM, Kim J, Bhakta AS. Survival Disparity and the Unique Genomic and Microbiome Profiles of Colon Cancer in Appalachian Kentucky. J Am Coll Surg 2025; 240:612-624. [PMID: 39868697 DOI: 10.1097/xcs.0000000000001299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND Colon cancer is a leading cause of mortality in Appalachian Kentucky. Studies suggest that the microbiome may influence cancer outcomes. We investigate differential gene expression, the tumor microbiome, and their association as potential drivers of disparities in colon cancer outcomes. STUDY DESIGN This study analyzed patients diagnosed with colon adenocarcinoma between 2010 and 2023. Demographic data were extracted from Kentucky Cancer Registry. Somatic mutations and significantly mutated genes were identified using Fisher's exact t -test. RNASeq data were processed for gene expression analysis and Holm-Bonferroni method was used to adjust p values for multiple comparisons. The STAR aligner (exotic), v2.1 pipeline, and KrakenUniq database were used to classify microbes in human samples. The R package (exotic) was then used to decontaminate the results. RESULTS The final cohort included 2,276 patients, 321 of which had available somatic mutation sequencing data. Demographic differences between Appalachian and non-Appalachian patients included marital status (p = 0.0005), race (p < 0.0001), insurance status (p = 0.0005), BMI (p = 0.001), type 2 diabetes (p < 0.0001), and Charlson Comorbidity Index (p = 0.03). There was no difference in gene mutation frequency. There was differential expression of 228 genes. Differential abundance analysis revealed differences in 381 bacterial species. Importantly, 3 microbiota significantly correlated with survival disparities between Appalachian and non-Appalachian patients: Clostridium cadaveris (adjusted p = 0.009), Ligilactobacillus salivarius (adjusted p = 0.048), and Sutterella wadsworthensis (adjusted p = 0.009). CONCLUSIONS This is the first report of the distinct tumor microbiome in Appalachian Kentucky and its impact on survival. Further studies are needed to better characterize the unique tumor and gut microbiome of Appalachian patients with colon cancer.
Collapse
Affiliation(s)
- Hannah G McDonald
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Anna M Reagan
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Abu Saleh Mosa Faisal
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Ryan Goettl
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Chi Wang
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Rebecca Hoyd
- The Ohio State University Comprehensive Cancer Center, Division of Medical Oncology, Columbus, OH (Hoyd, Spakowicz)
| | - Daniel Spakowicz
- The Ohio State University Comprehensive Cancer Center, Division of Medical Oncology, Columbus, OH (Hoyd, Spakowicz)
| | - B Mark Evers
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Joseph Kim
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Avinash S Bhakta
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| |
Collapse
|
19
|
Agnes CJ, Li L, Bertrand D, Murshed M, Willie BM, Tabrizian M. Assessment of bone regeneration potential for a 6-bromoindirubin-3'-oxime (BIO) encapsulated chitosan based scaffold in a mouse critical sized bone defect model. Int J Biol Macromol 2025; 304:140995. [PMID: 39952511 DOI: 10.1016/j.ijbiomac.2025.140995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Development of an effective treatment to guide bone repair in critical size bone defect applications remains a major unmet challenge. Current clinical gold standards display significant disadvantages, thereby necessitating that research focus on designing and producing a suitable alternative for healing. In this study, we comprehensively assessed the bone regenerative potential of a newly formulated 6-Bromoindirubin-3'-Oxime (BIO) incorporated chitosan-based scaffold using a mouse femoral defect model. Live 3D in vivo micro-CT imaging enabled us to monitor the progression of bone formation over 56 days, without needing additional replicates. Results demonstrated smaller distances between bone ends (1.033 ± 0.512 mm) compared to controls (1.474 ± 0.465 mm) at later timepoints (p = 0.0430), suggesting improved bone formation. This observed effect was supported with serum procollagen type I N-propeptide levels, where BIO scaffolds showed marked increases in collagen synthesis. As vascularization is often-overlooked, blood vessel density at 56 days was also assessed, showing an additional benefit of BIO incorporated scaffolds (9.264 ± 0.578) over controls (6.667 ± 1.300) on angiogenesis. Although BIO's incorporation did not lead to bony bridging or a significant difference in bone volume compared to controls at day 56, our findings suggest the BIO incorporated scaffold's ability to improve healing outcomes through enhancement of Wnt signaling. Further studies aimed at optimizing the dose to target this pathway are warranted, as a means to more completely regenerate bone in challenging healing scenarios.
Collapse
Affiliation(s)
- Celine J Agnes
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada; Shriners Hospital for Children, Montreal, QC, Canada.
| | - Ling Li
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
| | - David Bertrand
- Shriners Hospital for Children, Montreal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
| | - Monzur Murshed
- Shriners Hospital for Children, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| | - Bettina M Willie
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada; Shriners Hospital for Children, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
20
|
Ding X, Sheng Z, Cui J, Cui M, Zhang L, Feng R, Wang Y, Sun W, Zhang X, Shi L, Zhang B. Breast cancer-derived exosomal miR-105-5p facilitates the transformation of NFs into CAFs through LATS2-NF-κB signaling. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40151143 DOI: 10.3724/abbs.2025017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Studies of cell-to-cell activities in the tumor microenvironment (TME) have identified multiple potential targets for oncotherapy. The interplay between tumor cells and neighboring cancer-associated fibroblasts (CAFs) persists in all stages of tumor progression. In this study, we reveal that exosomes from breast cancer cells can be endocytosed into fibroblasts and transform normal fibroblasts (NFs) into CAFs and that the ability of exosomes from highly metastatic breast cancer cells is greater than that of those from poorly metastatic breast cancer cells. Further investigation reveals that exosomes from highly metastatic breast cancer cells contain much more miR-105-5p than those from poorly metastatic breast cells do and that exosomal miR-105-5p facilitates the transformation of NFs to CAFs. A detailed study reveals that RBMY1A1-dependent sorting of miR-105-5p into fibroblasts and subsequent internalization of miR-105-5p promote the transformation of NFs to CAFs by downregulating LATS2 expression and activating NF-κB signaling, which concurrently facilitates the EMT of breast cancer cells. Thus, our results indicate that exosomal miR-105-5p may be a potential target for novel therapeutic strategies to prevent the coevolution of breast cancer cells and CAFs.
Collapse
Affiliation(s)
- Xiaodi Ding
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Zhimei Sheng
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
- Affiliated Hospital of Shandong Second Medical University, Weifang 261041, China
| | - Jiayu Cui
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Meimei Cui
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Liying Zhang
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Ruijun Feng
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Yongming Wang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang 261041, China
| | - Wei Sun
- Affiliated Hospital of Shandong Second Medical University, Weifang 261041, China
| | - Xiurong Zhang
- Department of Pharmacology, Shandong Second Medical University, Weifang 261053, China
| | - Lihong Shi
- Department of Rehabilitation Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Baogang Zhang
- Department of Pathology, Shandong Second Medical University, Weifang 261053, China
| |
Collapse
|
21
|
Srikanth Y, Reddy DH, Anusha VL, Dumala N, Viswanadh MK, Chakravarthi G, Nalluri BN, Yadagiri G, Ramakrishna K. Unveiling the Multifaceted Pharmacological Actions of Indole-3-Carbinol and Diindolylmethane: A Comprehensive Review. PLANTS (BASEL, SWITZERLAND) 2025; 14:827. [PMID: 40094833 PMCID: PMC11902694 DOI: 10.3390/plants14050827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Cruciferae family vegetables are remarkably high in phytochemicals such as Indole-3-carbinol (I3C) and Diindolylmethane (DIM), which are widely known as nutritional supplements. I3C and DIM have been studied extensively in different types of cancers like breast, prostate, endometrial, colorectal, gallbladder, hepatic, and cervical, as well as cancers in other tissues. In this review, we summarized the protective effects of I3C and DIM against cardiovascular, neurological, reproductive, metabolic, bone, respiratory, liver, and immune diseases, infections, and drug- and radiation-induced toxicities. Experimental evidence suggests that I3C and DIM offer protection due to their antioxidant, anti-inflammatory, antiapoptotic, immunomodulatory, and xenobiotic properties. Apart from the beneficial effects, the present review also discusses the possible toxicities of I3C and DIM that are reported in various preclinical investigations. So far, most of the reports about I3C and DIM protective effects against various diseases are only from preclinical studies; this emphasizes the dire need for large-scale clinical trials on these phytochemicals against human diseases. Further, in-depth research is required to improve the bioavailability of these two phytochemicals to achieve the desirable protective effects. Overall, our review emphasizes that I3C and DIM may become potential drug candidates for combating dreadful human diseases.
Collapse
Affiliation(s)
- Yadava Srikanth
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Dontiboina Harikrishna Reddy
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Vinjavarapu Lakshmi Anusha
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Naresh Dumala
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Matte Kasi Viswanadh
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Guntupalli Chakravarthi
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Buchi N. Nalluri
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Ganesh Yadagiri
- Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kakarla Ramakrishna
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| |
Collapse
|
22
|
Zhang Y, Yan W, Wu L, Yu Z, Quan Y, Xie X. Different exosomes are loaded in hydrogels for the application in the field of tissue repair. Front Bioeng Biotechnol 2025; 13:1545636. [PMID: 40099037 PMCID: PMC11911322 DOI: 10.3389/fbioe.2025.1545636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Exosomes are double-membrane vesicular nanoparticles in the category of extracellular vesicles, ranging in size from 30 to 150 nm, and are released from cells through a specific multi-step exocytosis process. Exosomes have emerged as promising tools for tissue repair due to their ability to transfer bioactive molecules that promote cell proliferation, differentiation, and tissue regeneration. However, the therapeutic application of exosomes is hindered by their rapid clearance from the body and limited retention at the injury site. To overcome these challenges, hydrogels, known for their high biocompatibility and porous structure, have been explored as carriers for exosomes. Hydrogels can provide a controlled release mechanism, prolonging the retention time of exosomes at targeted tissues, thus enhancing their therapeutic efficacy. This review focuses on the combination of different exosomes with hydrogels in the context of tissue repair. We first introduce the sources and functions of exosomes, particularly those from mesenchymal stem cells, and their roles in regenerative medicine. We then examine various types of hydrogels, highlighting their ability to load and release exosomes. Several strategies for encapsulating exosomes in hydrogels are discussed, including the impact of hydrogel composition and structure on exosome delivery efficiency. Finally, we review the applications of exosomes-loaded hydrogels in the repair of different tissues, such as skin, bone, cartilage, and nerve, and explore the challenges and future directions in this field. The combination of exosomes with hydrogels offers significant promise for advancing tissue repair strategies and regenerative therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Xie
- College of Life Sciences, Northwest University, Xi’an, China
| |
Collapse
|
23
|
Cui X, Zhu Y, Zeng L, Zhang M, Uddin A, Gillespie TW, McCullough LE, Zhao S, Torres MA, Wan Y. Pharmacological Dissection Identifies Retatrutide Overcomes the Therapeutic Barrier of Obese TNBC Treatments through Suppressing the Interplay between Glycosylation and Ubiquitylation of YAP. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407494. [PMID: 39868848 PMCID: PMC11923992 DOI: 10.1002/advs.202407494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/13/2025] [Indexed: 01/28/2025]
Abstract
Triple-negative breast cancer (TNBC) in obese patients remains challenging. Recent studies have linked obesity to an increased risk of TNBC and malignancies. Through multiomic analysis and experimental validation, a dysfunctional Eukaryotic Translation Initiation Factor 3 Subunit H (EIF3H)/Yes-associated protein (YAP) proteolytic axis is identified as a pivotal junction mediating the interplay between cancer-associated adipocytes and the response to anti-cancer drugs in TNBC. Mechanistically, cancer-associated adipocytes drive metabolic reprogramming resulting in an upregulated hexosamine biosynthetic pathway (HBP). This aberrant upregulation of HBP promotes YAP O-GlcNAcylation and the subsequent recruitment of EIF3H deubiquitinase, which stabilizes YAP, thus promoting tumor growth and chemotherapy resistance. It is found that Retatrutide, an anti-obesity agent, inhibits HBP and YAP O-GlcNAcylation leading to increased YAP degradation through the deprivation of EIF3H-mediated deubiquitylation of YAP. In preclinical models of obese TNBC, Retatrutide downregulates HBP, decreases YAP protein levels, and consequently decreases tumor size and enhances chemotherapy efficacy. This effect is particularly pronounced in obese mice bearing TNBC tumors. Overall, these findings reveal a critical interplay between adipocyte-mediated metabolic reprogramming and EIF3H-mediated YAP proteolytic control, offering promising therapeutic strategies to mitigate the adverse effects of obesity on TNBC progression.
Collapse
Affiliation(s)
- Xin Cui
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGA30322USA
| | - Yueming Zhu
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGA30322USA
- Winship Cancer InstituteEmory University School of MedicineAtlantaGA30322USA
| | - Lidan Zeng
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGA30322USA
| | - Mengyuan Zhang
- Department of Biochemistry and Molecular BiologyInstitute of BioinformaticsUniversity of GeorgiaAthensGA30602USA
| | - Amad Uddin
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGA30322USA
| | - Theresa W. Gillespie
- Winship Cancer InstituteEmory University School of MedicineAtlantaGA30322USA
- Department of SurgeryEmory University School of MedicineAtlantaGA30322USA
- Department of Hematology and Medical OncologyEmory University School of MedicineAtlantaGA30322USA
| | - Lauren E. McCullough
- Winship Cancer InstituteEmory University School of MedicineAtlantaGA30322USA
- Department of EpidemiologyEmory University Rollins School of Public HealthAtlantaGA30322USA
| | - Shaying Zhao
- Department of Biochemistry and Molecular BiologyInstitute of BioinformaticsUniversity of GeorgiaAthensGA30602USA
| | - Mylin A. Torres
- Winship Cancer InstituteEmory University School of MedicineAtlantaGA30322USA
- Department of Radiation OncologyEmory University School of MedicineAtlantaGA30322USA
| | - Yong Wan
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGA30322USA
- Winship Cancer InstituteEmory University School of MedicineAtlantaGA30322USA
- Department of Hematology and Medical OncologyEmory University School of MedicineAtlantaGA30322USA
| |
Collapse
|
24
|
Ma N, Huang L, Zhou Q, Zhang X, Luo Q, Song G. Mechanical stretch promotes the migration of mesenchymal stem cells via Piezo1/F-actin/YAP axis. Exp Cell Res 2025; 446:114461. [PMID: 39988125 DOI: 10.1016/j.yexcr.2025.114461] [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: 09/22/2024] [Revised: 01/15/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Mesenchymal stem cells (MSCs) have self-renewal ability and the potential for multi-directional differentiation, and their clinical application has promising prospects, but improving the migration ability of MSCs in vivo is one of the challenges. We previously determined mechanical stretch at 1 Hz with 10 % strain for 8 h can significantly promote MSC migration, however, the molecular mechanism remains poorly understood. Here, we reported that the expression and activity of yes-associated protein (YAP) are upregulated after mechanical stretch. As a classical inhibitor of the YAP-TEAD activity and YAP protein, the treatment of verteporfin (VP) suppressed mechanical stretch-promoted MSC migration. We also observed F-actin polymerization after mechanical stretch. Next, we used Latrunculin A (Lat A), the most widely used reagent to depolymerize actin filaments, to treat MSCs and we found that Lat A treatment inhibits MSC migration by suppressing YAP expression and activity. In addition, the protein expression of Piezo1 was also upregulated after mechanical stretch. Knockdown of Piezo1 suppressed mechanical stretch-promoted MSC migration by restraining F-actin polymerization. Together, these findings demonstrate the role of Piezo1/F-actin/YAP signaling pathway in MSC migration under mechanical stretch, providing new experimental evidence for an in-depth understanding the mechanobiological mechanism of MSC migration.
Collapse
Affiliation(s)
- Ning Ma
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Lei Huang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Qianxu Zhou
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Xiaomei Zhang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
25
|
Poorkazem H, Saber M, Moradmand A, Yakhkeshi S, Seydi H, Hajizadeh-Saffar E, Shekari F, Hassani SN. Comparative effects of various extracellular vesicle subpopulations derived from clonal mesenchymal stromal cells on cultured fibroblasts in wound healing-related process. Int J Biochem Cell Biol 2025; 180:106737. [PMID: 39828140 DOI: 10.1016/j.biocel.2025.106737] [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: 10/23/2024] [Revised: 12/25/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Non-healing wounds pose significant challenges and require effective therapeutic interventions. Extracellular vesicles (EVs) have emerged as promising cell-free therapeutic agents in tissue regeneration. However, the functional differences between different subpopulations of EVs in wound healing remain understudied. This study aimed to evaluate the effects of two distinct subpopulations of clonal mesenchymal stromal cells (cMSC)-derived EVs (cMSC-EVs), namely 20 K and 110K-cMSC-EVs, primarily on in vitro wound healing process, providing fast and cost-effective alternatives to animal models. METHODS In vitro assays were conducted to compare the effects of 20 K and 110K-cMSC-EVs, isolated through high-speed centrifugation and differential ultracentrifugation, respectively. For evaluation the main mechanisms of wound healing, including cell proliferation, cell migration, angiogenesis, and contraction. Human dermal fibroblasts (HDF) were considered as the main cells for analysis of these procedures. Moreover, gene expression analysis was performed to assess the impact of these EV subpopulations on the related process of wound healing on HDF. RESULTS The results demonstrated that both 20 K and 110K-cMSC-EVs exhibited beneficial effects on cell proliferation, cell migration, angiogenesis, and gel contraction. RT-qPCR revealed that both EV types downregulated interleukin 6 (IL6), induced proliferation by upregulating proliferating cell nuclear antigen (PCNA), and regulated remodeling by upregulating matrix metallopeptidase 1 (MMP1) and downregulating collagen type 1 (COL1). DISCUSSION This study highlights the effects of both 20 K and 110K-cMSC-EVs on the potency of HDFs in wound healing-related process. As the notable finding, 20K-cMSC-EVs offer a more feasible and cost-effective subpopulation for isolation and follow the GMP standard, recommended to utilize this fraction for therapeutic application.
Collapse
Affiliation(s)
- Hedie Poorkazem
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Saber
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Azadeh Moradmand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
26
|
Grossini E, Surico D, Venkatesan S, Ola Pour MM, Aquino CI, Remorgida V. Extracellular Vesicles and Pregnancy-Related Hypertensive Disorders: A Descriptive Review on the Possible Implications "From Bench to Bedside". BIOLOGY 2025; 14:240. [PMID: 40136497 PMCID: PMC11939443 DOI: 10.3390/biology14030240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
Pregnancy involves extracellular vesicles (EVs) through mechanisms that are poorly understood to date. Furthermore, it is not surprising that EVs may also be involved in the pathophysiology of pre-eclampsia (PE) and gestational hypertension, two clinical conditions with high morbidity and mortality, given their capacity to mediate intracellular communications and regulate inflammation and angiogenesis. We searched major online scientific search engines (PubMed, Google Scholar, Scopus, WES, Embase, etc.) using the terms "Preeclampsia", "Pregnancy", "Hypertension", "Pregnancy-related hypertension", "Extracellular vesicles", "Biomarkers", "Gestation" AND "Obstetrics". Finding potential early biomarkers of risk or illness progression would be essential for the optimum care of expectant mothers with the aforementioned conditions. Nevertheless, none of the various screening assays that have been discovered recently have shown high predictive values. The analysis of EVs in the peripheral blood starting from the first trimester of pregnancy may hold great promise for the possible correlation with gestational hypertension problems and represent a marker of the early stages of the disease. EVs use may be a novel therapeutic approach for the management of various illnesses, as well. In order to define EVs' function in the physiopathology of pregnancy-associated hypertension and PE, as well as their potential as early biomarkers and therapeutic tools, we have compiled the most recent data in this review.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Daniela Surico
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Mohammad Mostafa Ola Pour
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Carmen Imma Aquino
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| | - Valentino Remorgida
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| |
Collapse
|
27
|
Esmaili M, Jafari N, Ahmadzadeh F, Toosi SMV, Abediankenari S. Effect of conditioned medium from miRNA-34a transfected gastric cancer-associated fibroblast on peripheral blood mononuclear cells. BMC Immunol 2025; 26:9. [PMID: 40000950 PMCID: PMC11854116 DOI: 10.1186/s12865-025-00688-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblast (CAF) cells play an important role in gastric malignancy. MiRNA dysregulation has been detected in CAF cells, which is related to the tumor progression ability of these cells. Therefore, this study aimed to evaluate the function of miRNA34a in CAF cells in gastric carcinoma. METHOD We transiently transfected miRNA-34a mimic in CAF cells and examined the effect of the overexpressed miRNA on PD-L1 expression using real-time PCR. Next, we evaluated the role of transfected CAF-conditioned medium (CM) on the immune response and viability of gastric cancer cell lines. RESULTS We have shown that miRNA-34a significantly reduced PD-L1 expression in CAF cells (p < 0.05). However, the conditioned medium of transfected cells had no significant effect on the immune response. We also found that CM of miRNA-34a transfected CAF cells significantly suppressed gastric cancer cell line viability relative to the control group (P < 0.05). CONCLUSION We indicated that CM of miRNA-34a transfected CAF can reduce gastric cancer cell line proliferation. Additionally, miRNA-34a in these cells may improve immune response via PD-L1 reduction. Thus, miRNA-34a could be a potential therapeutic agent in gastric cancer treatment. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Mozhgan Esmaili
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Ahmadzadeh
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
28
|
Cassani M, Niro F, Fernandes S, Pereira-Sousa D, Faes Morazzo S, Durikova H, Wang T, González-Cabaleiro L, Vrbsky J, Oliver-De La Cruz J, Klimovic S, Pribyl J, Loja T, Skladal P, Caruso F, Forte G. Regulation of Cell-Nanoparticle Interactions through Mechanobiology. NANO LETTERS 2025; 25:2600-2609. [PMID: 39772635 PMCID: PMC11849000 DOI: 10.1021/acs.nanolett.4c04290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/21/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Bio-nano interactions have been extensively explored in nanomedicine to develop selective delivery strategies and reduce systemic toxicity. To enhance the delivery of nanocarriers to cancer cells and improve the therapeutic efficiency, different nanomaterials have been developed. However, the limited clinical translation of nanoparticle-based therapies, largely due to issues associated with poor targeting, requires a deeper understanding of the biological phenomena underlying cell-nanoparticle interactions. In this context, we investigate the molecular and cellular mechanobiology parameters that control such interactions. We demonstrate that the pharmacological inhibition or the genetic ablation of the key mechanosensitive component of the Hippo pathway, i.e., yes-associated protein, enhances nanoparticle internalization by 1.5-fold. Importantly, this phenomenon occurs independently of nanoparticle properties, such as size, or cell properties such as surface area and stiffness. Our study reveals that the internalization of nanoparticles in target cells can be controlled by modulating cell mechanosensing pathways, potentially enhancing nanotherapy specificity.
Collapse
Affiliation(s)
- Marco Cassani
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | - Francesco Niro
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- School of
Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London WC2R 2LS, U.K.
- Faculty of
Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech
Republic
| | - Soraia Fernandes
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | - Daniel Pereira-Sousa
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- Faculty of
Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech
Republic
| | - Sofia Faes Morazzo
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- Faculty of
Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech
Republic
| | - Helena Durikova
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | - Tianzheng Wang
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | - Lara González-Cabaleiro
- Departamento
de Química Física, Universidade
de Vigo, Campus Universitario As Lagoas
Marcosende, Vigo 36310, Spain
| | - Jan Vrbsky
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
| | - Jorge Oliver-De La Cruz
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for
Science and Technology (BIST), 08028 Barcelona, Spain
| | - Simon Klimovic
- Nanobiotechnology
Core Facility, CEITEC Masaryk University, 62500 Brno, Czech Republic
- Department
of Biochemistry, Faculty of Science, Masaryk
University, 62500 Brno, Czech Republic
| | - Jan Pribyl
- Nanobiotechnology
Core Facility, CEITEC Masaryk University, 62500 Brno, Czech Republic
| | - Tomas Loja
- Molecular
Medicine, CEITEC Masaryk University, 62500 Brno, Czech Republic
| | - Petr Skladal
- Department
of Biochemistry, Faculty of Science, Masaryk
University, 62500 Brno, Czech Republic
| | - Frank Caruso
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | - Giancarlo Forte
- International
Clinical Research Center, St. Anne’s
University Hospital, 65691 Brno, Czech Republic
- School of
Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London WC2R 2LS, U.K.
| |
Collapse
|
29
|
Luo X, McAndrews KM, Kalluri R. Natural and Bioengineered Extracellular Vesicles in Diagnosis, Monitoring and Treatment of Cancer. ACS NANO 2025; 19:5871-5896. [PMID: 39869032 PMCID: PMC12002402 DOI: 10.1021/acsnano.4c11630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Extracellular vesicles (EVs) are cell derived nanovesicles which are implicated in both physiological and pathological intercellular communication, including the initiation, progression, and metastasis of cancer. The exchange of biomolecules between stromal cells and cancer cells via EVs can provide a window to monitor cancer development in real time for better diagnostic and interventional strategies. In addition, the process of secretion and internalization of EVs by stromal and cancer cells in the tumor microenvironment (TME) can be exploited for delivering therapeutics. EVs have the potential to provide a targeted, biocompatible, and efficient delivery platform for the treatment of cancer and other diseases. Natural as well as engineered EVs as nanomedicine have immense potential for disease intervention. Here, we provide an overview of current knowledge of EVs' function in cancer progression, diagnostic and therapeutic applications for EVs in the cancer setting, as well as current EV engineering strategies.
Collapse
Affiliation(s)
- Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Kathleen M. McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
30
|
Abbasi R, Alamdari-Mahd G, Maleki-Kakelar H, Momen-Mesgin R, Ahmadi M, Sharafkhani M, Rezaie J. Recent advances in the application of engineered exosomes from mesenchymal stem cells for regenerative medicine. Eur J Pharmacol 2025; 989:177236. [PMID: 39753159 DOI: 10.1016/j.ejphar.2024.177236] [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: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Exosomes, cell-derived vesicles produced by cells, are fascinating and drawing growing interest in biomedical exploration due to their exceptional properties. There is intriguing evidence that exosomes are involved in major biological processes, including diseases and regeneration. Exosomes from mesenchymal stem cells (MSCs) have shown promising outcomes in regenerative medicine. Numerous studies suggest that exosomes have several advantages over conventional synthetic nanocarriers, opening novel frontiers for innovative drug delivery. Regenerative medicine has demonstrated the profound therapeutic outcomes of engineered or loaded exosomes from MSCs. Different methods are being used to modify or/load exosomes. These exosomes can improve cell signaling pathways for bone and cartilage diseases, liver diseases, nerve tissues, kidney diseases, skin tissue, and cardiovascular diseases. Despite extensive research, clinical translation of these exosomes remains a challenge. The optimization of cargo loading methods, efficiency, physiological stability, and the isolation and characterization of exosomes present some challenges. The upcoming examination should include the development of large-scale, quality-controllable production approaches, the modification of drug loading approaches, and numerous in vivo investigations and clinical trials. Here, we provided an informative overview of the extracellular vesicles and modification/loading methods of exosomes. We discuss the last exosome research on regeneration disorders, highlighting the therapeutic applications of MSCs-derived exosomes. We also highlight future directions and challenges, underscoring the significance of addressing the main questions in the field.
Collapse
Affiliation(s)
- Reza Abbasi
- Department of Biology, Urmia University, Urmia, Iran
| | - Ghazal Alamdari-Mahd
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | | | - Mahdi Ahmadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohaddeseh Sharafkhani
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
31
|
Budayr OM, Miller BC, Nguyen J. Harnessing extracellular vesicle-mediated crosstalk between T cells and cancer cells for therapeutic applications. J Control Release 2025; 378:266-280. [PMID: 39657892 PMCID: PMC11830559 DOI: 10.1016/j.jconrel.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Small extracellular vesicles (EVs) are a diverse group of lipid-based particles that are ≤200 nm in diameter and contain an aqueous core. EVs have been shown to mediate intercellular communications between a wide array of immune cells; the downstream effects are diverse and have potential implications for the development of novel immunotherapeutic treatments. Despite a high volume of studies addressing the role EVs play in the immune system, our understanding of the crosstalk between T cells and cancer cells remains limited. Here, we discuss how EVs derived from cancer cells modulate T cell functions and conversely, how T cell derived EVs are crucial in modulating adaptive immune functions. In the context of cancer, tumor derived EVs (TD-EVs) halt T cell-mediated immunity by interfering with effector functions and enhancing regulatory T cell (Treg) functions. In contrast, EVs derived from effector T cells can serve to stimulate anticancer immunity, curbing metastasis and tumor growth. These findings highlight important aspects of how EVs can both mediate the therapeutic effects of T cells as well as impair T cell-mediated immunity. This calls for a deeper understanding of EV-mediated effects in order to advance them as next-generation therapeutics and nanocarriers.
Collapse
Affiliation(s)
- Omar M Budayr
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian C Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
32
|
Han J, Li GC, Fang SY, Cui YM, Yang HH. Dermal Fibroblast-Derived Exosomes Promotes Bone-to-Tendon Interface Healing of Chronic Rotator Cuff Tear in Rabbit Model. Arthroscopy 2025:S0749-8063(25)00061-1. [PMID: 39914613 DOI: 10.1016/j.arthro.2025.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/05/2025] [Accepted: 01/19/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE To investigate the efficacy of exosomes derived from dermal fibroblasts (DF-Ex) on bone-to-tendon interface (BTI) healing in a chronic rotator cuff tear (RCT) model of rabbit. METHODS After extraction of DF-Ex, the characterization of DF-Ex was identified in the in vitro study. In the in vivo experiment, 48 rabbits were randomly allocated into 3 groups. To create chronic RCT models, transected tendons were left untreated for 6 weeks and then were repaired in a transosseous manner. Different materials were injected into repair site according to the allocated group (group A: saline, group B: fibrin glue only, group C: DF-Ex with fibrin glue; n = 16 for each). Genetic and immunofluorescence analyses were conducted at 4 weeks post-surgery. Furthermore, genetic, histologic, and biomechanical analyses were conducted at 12 weeks post-surgery. RESULTS In vitro analyses revealed the exosomal marker proteins CD9, CD63, and ALIX were positively expressed in DF-Ex, whereas negative control Calnexin was nearly absent. In vivo analyses showed that group C had the highest mRNA expression levels of COL1A1, COL3A1, and ACAN among all groups (P < .001, P = .007, and P = .002, respectively) at 4 weeks postsurgery. Meanwhile, there were more preliminary fibrocartilaginous matrix (aggrecan+/collagen II+) formation in group C. At 12 weeks postsurgery, group C had better collagen fiber continuity and orientation, denser collagen fibers, more mature bone-to-tendon junction, and greater fibrocartilage layer formation compared with the other groups (all P < .05). Moreover, group C also had greater load-to-failure value (53.3 ± 6.1 N/kg, P < .001). CONCLUSIONS Topical DF-Ex administration effectively promoted BTI healing by upregulating the COL1A1, COL3A1, and ACAN mRNA expression levels at an early stage and enhancing the structural and biomechanical properties at 12 weeks after surgical repair of a chronic RCT model of rabbit. CLINICAL RELEVANCE The study could be a transitional study to investigate the efficacy of DF-Ex on BTI healing for surgical repair of chronic RCTs as a powerful biological agent in humans.
Collapse
Affiliation(s)
- Jian Han
- Department of Orthopaedic Surgery, The First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Guan-Cong Li
- Department of Orthopaedic Surgery, The First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Shen-Yun Fang
- Department of Orthopaedic Surgery, The First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Yun-Mei Cui
- Department of Pediatrics, The First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Hong-Hang Yang
- Department of Orthopaedic Surgery, The First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China.
| |
Collapse
|
33
|
Louka A, Spacho N, Korovesis D, Adamis K, Papadopoulos C, Kalaitzaki E, Tavernarakis N, Neochoritis CG, Eleftheriadis N. Crafting Molecular Tools for 15-Lipoxygenase-1 in a Single Step. Angew Chem Int Ed Engl 2025; 64:e202418291. [PMID: 39523872 PMCID: PMC11795718 DOI: 10.1002/anie.202418291] [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: 09/23/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Small molecule modulators are powerful tools for selectively probing and manipulating proteins in native biological systems. However, the development of versatile modulators that exhibit desired properties is hindered by the lack of a rapid and robust synthetic strategy. Here, we develop a facile and reliable one-step methodology for the generation of multifunctional toolboxes encompassing a wide variety of chemical modulators with different desired features. These modulators bind irreversibly to the protein target via a selective warhead. Key elements are introduced onto the warhead in a single step using multi-component reactions. To illustrate the power of this new technology, we synthesized a library of diverse modulators designed to explore a highly challenging and poorly understood protein, human 15-lipoxygenase-1. Modulators made include; activity-based/photoaffinity probes, chemosensors, photocrosslinkers, as well as light-controlled and high-affinity inhibitors. The efficacy of our compounds was successfully established through the provision of on demand inhibition and labeling of our target protein in vitro, in cellulo and in vivo; thus, proving that this technology has promising potential for applications in many complex biological systems.
Collapse
Affiliation(s)
- Anastasia Louka
- Department of ChemistryUniversity of CreteVoutes70013 HeraklionGreece
| | - Ntaniela Spacho
- Department of ChemistryUniversity of CreteVoutes70013 HeraklionGreece
| | - Dimitris Korovesis
- Institute of Molecular Biology and BiotechnologyFoundation for Research and Technology-HellasNikolaou Plastira 10070013HeraklionGreece
| | | | | | | | - Nektarios Tavernarakis
- Institute of Molecular Biology and BiotechnologyFoundation for Research and Technology-HellasNikolaou Plastira 10070013HeraklionGreece
- Division of Basic SciencesSchool of MedicineUniversity of CreteVoutes70013 HeraklionGreece
| | | | | |
Collapse
|
34
|
Wu X, Yuan P, Wei N, Ma C, Fu M, Wu W. Extracellular vesicles derived from "serum and glucose" deprived HUCMSCs promoted skin wound healing through enhanced angiogenesis. Mol Cell Biochem 2025; 480:1255-1273. [PMID: 38967721 DOI: 10.1007/s11010-024-05058-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Extracellular vesicles (EVs) produced from MSCs were currently considered as a novel therapeutic agent for skin tissue regeneration and repair. Preconditioning stem cells may activate more molecular pathways and release more bioactive agents. In this study, we obtained EVs from normal (N-EVs) and serum- and glucose-deprived (SGD-EVs) human umbilical cord mesenchymal stem cells (HUCMSCs), and showed that SGD-EVs promoted the migration, proliferation, and tube formation of HUVECs in vitro. In vivo experiments utilizing a rat model show that both N-EVs and SGD-EVs boosted angiogenesis of skin defects and accelerated skin wound healing, while treating wounds with SGD-EVs led to faster skin healing and enhanced angiogenesis. miRNA sequencing showed that miR-29a-3p was abundant in SGD-EVs, and overexpressing miR-29a-3p enhanced the angiogenic ability of HUVECs, while inhibiting miR-29a-3p presented the opposite effect. Further studies demonstrated that miR-29a-3p directly targeted CTNNBIP1, which mediated angiogenesis of HUCMSCs-derived EVs through inhibiting CTNNBIP1 to activate Wnt/β-catenin signaling pathway. Taken together, these findings suggested that SGD-EVs promote angiogenesis via transferring miR-29a-3p, and activation of Wnt/β-catenin signaling pathway played a crucial role in SGD-EVs-induced VEGFA production during wound angiogenesis. Our results offered a new avenue for modifying EVs to enhance tissue angiogenesis and augment its role in skin repair.
Collapse
Affiliation(s)
- Xiaopeng Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, 710069, China
| | - Pingping Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Na Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, 710069, China
| | - Chaoqun Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Mingdi Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Wei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
35
|
Li C, Xu T, Hou G, Wang Y, Fu Q. DNA nanotechnology-based strategies for gastric cancer diagnosis and therapy. Mater Today Bio 2025; 30:101459. [PMID: 39866794 PMCID: PMC11762204 DOI: 10.1016/j.mtbio.2025.101459] [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: 11/01/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Gastric cancer (GC) is a formidable adversary in the field of oncology. The low early diagnosis rate of GC results in a low overall survival rate. Therefore, early accurate diagnosis and effective treatment are the key to reduce the mortality of GC. With the advent of nanotechnology, researchers continue to explore new possibilities for accurate diagnosis and effective treatment. One such breakthrough is the application of DNA nanotechnology. In this paper, the application of exciting DNA nanomaterials in the diagnosis and treatment of GC is discussed in depth. Firstly, the biomarkers related to GC and the diagnostic strategies related to DNA nanotechnology are summarized. Second, the latest research progress of DNA nanomaterials in the GC targeted therapy are summarized. Finally, the challenges and opportunities of DNA nanomaterials in the research and clinical application of GC are prospected.
Collapse
Affiliation(s)
- Congcong Li
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Tongyang Xu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Guopeng Hou
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Yin Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Qinrui Fu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| |
Collapse
|
36
|
Ajongbolo AO, Langhans SA. YAP/TAZ-associated cell signaling - at the crossroads of cancer and neurodevelopmental disorders. Front Cell Dev Biol 2025; 13:1522705. [PMID: 39936032 PMCID: PMC11810912 DOI: 10.3389/fcell.2025.1522705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
YAP/TAZ (Yes-associated protein/paralog transcriptional co-activator with PDZ-binding domain) are transcriptional cofactors that are the key and major downstream effectors of the Hippo signaling pathway. Both are known to play a crucial role in defining cellular outcomes, including cell differentiation, cell proliferation, and apoptosis. Aside from the canonical Hippo signaling cascade with the key components MST1/2 (mammalian STE20-like kinase 1/2), SAV1 (Salvador homologue 1), MOB1A/B (Mps one binder kinase activator 1A/B) and LATS1/2 (large tumor suppressor kinase 1/2) upstream of YAP/TAZ, YAP/TAZ activation is also influenced by numerous other signaling pathways. Such non-canonical regulation of YAP/TAZ includes well-known growth factor signaling pathways such as the epidermal growth factor receptor (EGFR)/ErbB family, Notch, and Wnt signaling as well as cell-cell adhesion, cell-matrix interactions and mechanical cues from a cell's microenvironment. This puts YAP/TAZ at the center of a complex signaling network capable of regulating developmental processes and tissue regeneration. On the other hand, dysregulation of YAP/TAZ signaling has been implicated in numerous diseases including various cancers and neurodevelopmental disorders. Indeed, in recent years, parallels between cancer development and neurodevelopmental disorders have become apparent with YAP/TAZ signaling being one of these pathways. This review discusses the role of YAP/TAZ in brain development, cancer and neurodevelopmental disorders with a special focus on the interconnection in the role of YAP/TAZ in these different conditions.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
- Biological Sciences Graduate Program, University of Delaware, Newark, DE, United States
| | - Sigrid A. Langhans
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
| |
Collapse
|
37
|
Zhu Z, Ding R, Yu W, Liu Y, Zhou Z, Liu CY. YAP/TEAD4/SP1-induced VISTA expression as a tumor cell-intrinsic mechanism of immunosuppression in colorectal cancer. Cell Death Differ 2025:10.1038/s41418-025-01446-2. [PMID: 39875519 DOI: 10.1038/s41418-025-01446-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Hyperactivation of the YAP/TEAD transcriptional complex in cancers facilitates the development of an immunosuppressive tumor microenvironment. Herein, we observed that the transcription factor SP1 physically interacts with and stabilizes the YAP/TEAD complex at regulatory genomic loci in colorectal cancer (CRC). In response to serum stimulation, PKCζ (protein kinase C ζ) was found to phosphorylate SP1 and enhance its interaction with TEAD4. As a result, SP1 enhanced the transcriptional activity of YAP/TEAD and coregulated the expression of a group of YAP/TEAD target genes. The immune checkpoint V-domain Ig suppressor of T-cell activation (VISTA) was identified as a direct target of the SP1-YAP/TEAD4 complex and found to be widely expressed in CRC cells. Importantly, YAP-induced VISTA upregulation in human CRC cells was found to strongly suppress the antitumor function of CD8+ T cells. Consistently, elevated VISTA expression was found to be correlated with hyperactivation of the SP1-YAP/TEAD axis and associated with poor prognosis of CRC patients. In addition, we found by serendipity that enzymatic deglycosylation significantly improved the anti-VISTA antibody signal intensity, resulting in more accurate detection of VISTA in clinical tumor samples. Overall, our study identified SP1 as a positive modulator of YAP/TEAD for the transcriptional regulation of VISTA and developed a protein deglycosylation strategy to better detect VISTA expression in clinical samples. These findings revealed a new tumor cell-intrinsic mechanism of YAP/TAZ-mediated cancer immune evasion.
Collapse
Affiliation(s)
- Zhehui Zhu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Rui Ding
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yun Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| | - Chen-Ying Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China.
| |
Collapse
|
38
|
Hou Y, Zhao Y, Shi Z, Pan Y, Shi K, Zhao C, Liu S, Chen Y, Zhao L, Wu J, Ge G, Jie W. Establishment of a nomogram model based on immune-related genes using machine learning for aortic dissection diagnosis and immunomodulation assessment. Int J Med Sci 2025; 22:873-886. [PMID: 39991758 PMCID: PMC11843136 DOI: 10.7150/ijms.100572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
The clinical manifestation of aortic dissection (AD) is complex and varied, making early diagnosis crucial for patient survival. This study aimed to identify immune-related markers to establish a nomogram model for AD diagnosis. Three datasets from GEO-GSE52093, GSE147026 and GSE153434-were combined and used for identification of immune-related causative genes using weighted gene co-expression network analysis, and 136 immune-related genes were obtained. Then, 15 pivotal genes were screened by the protein-protein interaction network. Through machine learning including the Least Absolute Shrinkage and Selection Operator algorithm, random forest algorithm, and multivariate logistic regression, four key feature genes were obtained-CXCL1, ITGA5, PTX3, and TIMP1-and the diagnostic scores based on these four genes were proved to be effective in distinguishing between AD patients and healthy donors. External dataset (GSE98770 and GSE190635) validation revealed this nomogram displayed strong predictive significance. Further analysis revealed that these genes are related with neutrophils, resting NK cells, resting mast cells, activated mast cells, activated dendritic cells, central memory CD4 T cells, γδ T cells, natural killer T cells, and myeloid-derived suppressor cells in AD. Finally, these four genes were validated to be upregulated in AD patients' tissue and serum samples compared with controls. These results suggest that this nomogram model, using machine learning identified four immune-related genes CXCL1, ITGA5, PTX3, and TIMP1, displays superior diagnostic ability in distinguishing AD and healthy individuals, and immune cells commonly associated with these hub genes may be therapeutic targets for AD.
Collapse
Affiliation(s)
- Yanjun Hou
- Department of Cardiovascular Surgery, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Yangyang Zhao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou 571199, China
- Emergency and Trauma College, Hainan Medical University, Haikou 571199, China
| | - Zhensu Shi
- Department of Cardiovascular Surgery, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Yipeng Pan
- Department of Transplantation, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Kaijia Shi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou 571199, China
| | - Chaoyang Zhao
- Department of Cardiovascular Surgery, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Shengnan Liu
- Department of Cardiovascular Surgery, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Yongkun Chen
- Department of Cardiovascular Surgery, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Lini Zhao
- Department of Pharmacy, the Second Affiliated Hospital, Hainan Medical University, Haikou, 570311, China
| | - Jizhen Wu
- Department of Quality Control, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Guangquan Ge
- Department of Cardiovascular Surgery, the Second Affiliated Hospital, Hainan Medical University, Haikou 570311, China
| | - Wei Jie
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
39
|
Chideriotis S, Anastasiadi AT, Tzounakas VL, Fortis SP, Kriebardis AG, Valsami S. Morphogens and Cell-Derived Structures (Exosomes and Cytonemes) as Components of the Communication Between Cells. Int J Mol Sci 2025; 26:881. [PMID: 39940651 PMCID: PMC11816454 DOI: 10.3390/ijms26030881] [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: 12/20/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Morphogens, which are non-classical transcription factors, according to several studies, display a crucial role in tissue patterning, organ architecture establishment, and human disease pathogenesis. Recent advances have expanded the morphogen participation to a wide range of human diseases. There are many genetic syndromes caused by mutations of components of morphogen signaling pathways. The aberrant morphogen pathways also promote cancer cell maintenance, renewal, proliferation, and migration. On the other hand, exosomes and their application in the biomedical field are of evolving significance. The evidence that membrane structures participate in the creation of morphogenic gradience and biodistribution of morphogen components renders them attractive as new therapeutic tools. This intercellular morphogen transport is performed by cell-derived structures, mainly exosomes and cytonemes, and extracellular substances like heparan sulphate proteoglycans and lipoproteins. The interaction between morphogens and Extracellular Vesicles has been observed at first in the most studied insect, Drosophila, and afterwards analogous findings have been proved in vertebrates. This review presents the protagonists and mechanisms of lipid-modified morphogens (Hedgehog and Wnt/β-catenin) biodistribution.
Collapse
Affiliation(s)
| | - Alkmini T. Anastasiadi
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece; (A.T.A.); (V.L.T.)
| | - Vassilis L. Tzounakas
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece; (A.T.A.); (V.L.T.)
| | - Sotirios P. Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (S.P.F.); (A.G.K.)
| | - Anastasios G. Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (S.P.F.); (A.G.K.)
| | - Serena Valsami
- Hematology Laboratory, Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
40
|
Kim JH, Kim JE, Kang SJ, Yoon JK. Exosomes and Exosome-Mimetics for Atopic Dermatitis Therapy. Tissue Eng Regen Med 2025:10.1007/s13770-024-00695-5. [PMID: 39832066 DOI: 10.1007/s13770-024-00695-5] [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: 12/14/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Exosomes and exosome mimetics are used as alternatives to cell therapy. They have shown potential in treating skin disorders by fortifying the skin barrier, mediating angiogenesis, and regulating the immune response while minimizing side effects. Currently, numerous studies have applied exosome therapy to treat atopic dermatitis (AD) caused by a weakened skin barrier and chronic inflammation. Research on exosomes and exosome mimetics represents a promising avenue for tissue regeneration, potentially paving the way for new therapeutic options. However, the efficacy of the therapy remains poorly understood. Also, the potential of exosome mimetics as alternatives to exosomes in skin therapy remains underexplored. METHODS Here, we reviewed the pathological features and current therapies of AD. Next, we reviewed the application of exosomes and exosome mimetics in regenerative medicine. Finally, we highlighted the therapeutic effects of exosomes based on their cell source and assessed whether exosome mimetics are viable alternatives. RESULTS AND CONCLUSION Exosome therapy may treat AD due to its skin regenerative properties, and exosome mimetics may offer an equally effective yet more efficient alternative. Research on exosomes and exosome mimetics represents a promising avenue for tissue regeneration, potentially paving the way for new therapeutic options.
Collapse
Affiliation(s)
- Jae Hoon Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Ju-El Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Seong-Jun Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea.
| |
Collapse
|
41
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
42
|
Wang J, Hua D, Li M, Liu N, Zhang Y, Zhao Y, Jiang S, Hu X, Wang Y, Zhu H. The Role of Zuo Jin Wan in Modulating the Tumor Microenvironment of Colorectal Cancer. Comb Chem High Throughput Screen 2025; 28:523-532. [PMID: 38284730 DOI: 10.2174/0113862073281374231228041841] [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: 09/11/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
INTRODUCTION Traditional Chinese medicine (TCM) can modulate the immune function of tumor patients in various ways. Zuojin Wan (ZJW, a 6:1 ratio of Huang Lian and Wu Zhu Yu) can modulate the microenvironment of ulcerative colitis, but its role in regulating the colorectal cancer (CRC) microenvironment remains unclear. Exploring the role of ZJW in CRC immunomodulation may improve the antitumor effect of existing immunotherapeutic strategies. MATERIAL AND METHODS The active compounds of each herb in ZJW were obtained from the HIT2.0 database with literature evidence. Single-cell RNA sequencing data of CRC were obtained from published studies (PMID: 32451460, 32103181, and 32561858). Pathway enrichment was analyzed using the reactome database, and intergenic correlation analysis was performed using the corrplot R software package. ZJW-regulated gene expression was verified by RT-qPCR. RESULTS Huang Lian and Wu Zhu Yu contain 19 and 4 compounds, respectively. Huang Lian targets 146 proteins, and Wu Zhu Yu targets 28 proteins based on evidence from the literature. ZJW regulates a range of biological processes associated with immune function, including cytokine signaling and Toll-Like Receptor 4 (TLR4) cascade. ZJW regulates malignant CRC cells, immune cells (including T-cells, B-cells, mast cells, NK/NKT cells, and myeloid cells), and other nonimmune cells (including endothelial cells, enteric glial cells, and pericytes). We confirmed that ZJW significantly downregulated the expression of TIMP1 and MTDHin CRC cell lines. CONCLUSIONS ZJW regulates a range of cells in the CRC microenvironment, including malignant CRC, immune cells, and stromal cells. In CRC cell lines, downregulation of TIMP1 and MTDH by ZJW may play an important role in the immunomodulation in CRC.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dongming Hua
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengyao Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ningning Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiyang Zhao
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shasha Jiang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huirong Zhu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
43
|
Li B, Chen H, Hang R. Osseointegration-Related Exosomes for Surface Functionalization of Titanium Implants. Biomater Res 2024; 28:0124. [PMID: 39711824 PMCID: PMC11661649 DOI: 10.34133/bmr.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Despite that the clinical application of titanium-based implants has achieved great success, patients' own diseases and/or unhealthy lifestyle habits often lead to implant failure. Many studies have been carried out to modify titanium implants to promote osseointegration and implant success. Recent studies showed that exosomes, proactively secreted extracellular vesicles by mammalian cells, could selectively target and modulate the functions of recipient cells such as macrophages, nerve cells, endothelial cells, and bone marrow mesenchymal stem cells that are closely involved in implant osseointegration. Accordingly, using exosomes to functionalize titanium implants has been deemed as a novel and effective way to improve their osseointegration ability. Herein, recent advances pertaining to surface functionalization of titanium implants with exosomes are analyzed and discussed, with focus on the role of exosomes in regulating the functions of osseointegration-related cells, and their immobilization strategies as well as resultant impact on osseointegration ability.
Collapse
Affiliation(s)
- Boqiong Li
- Department of Materials Science and Engineering,
Jinzhong University, Jinzhong 030619, China
| | - Huanming Chen
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
44
|
Wei B, Wei M, Huang H, Fan T, Zhang Z, Song X. Mesenchymal Stem Cell-Derived Exosomes: A Promising Therapeutic Strategy for Age-Related Diseases. Cell Prolif 2024:e13795. [PMID: 39704104 DOI: 10.1111/cpr.13795] [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: 08/28/2024] [Revised: 11/09/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
The global increase in the aging population has led to a concurrent rise in the incidence of age-related diseases, posing substantial challenges to healthcare systems and affecting the well-being of the elderly. Identifying and securing effective treatments has become an urgent priority. In this context, mesenchymal stem cell-derived exosomes (MSC-Exos) have emerged as a promising and innovative modality in the field of anti-aging medicine, offering a multifaceted therapeutic approach. MSC-Exos demonstrate significant potential due to their immunomodulatory and anti-inflammatory properties, their ability to inhibit oxidative stress, and their reparative effects on senescent tissues. These attributes make them valuable in combating a range of conditions associated with aging, such as cardiovascular diseases, neurodegeneration, skin aging, and osteoarthritis. The integration of exosomes with membrane-penetrating peptides introduces a novel strategy for the delivery of biomolecules, surmounting traditional cellular barriers and enhancing therapeutic efficacy. This review provides a comprehensive synthesis of the current understanding of MSC-Exos, underscoring their role as a novel and potent therapeutic strategy against the intricate challenges of age-related diseases.
Collapse
Affiliation(s)
- Bohua Wei
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China
| | - Mengting Wei
- School of Stomatology, China Medical University, Shenyang, Liaoning Province, China
| | - Haonan Huang
- China Medical University, Shenyang, Liaoning Province, China
| | - Ting Fan
- Department of Computer, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning Province, China
| | - Zhichang Zhang
- Department of Computer, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning Province, China
| | - Xiaoyu Song
- The College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
45
|
Gharebaghi A, Afshar S, Tapak L, Ranjbar H, Saidijam M, Dinu I. Utilizing an In-silico Approach to Pinpoint Potential Biomarkers for Enhanced Early Detection of Colorectal Cancer. Cancer Inform 2024; 23:11769351241307163. [PMID: 39687502 PMCID: PMC11648020 DOI: 10.1177/11769351241307163] [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: 06/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Objectives Colorectal cancer (CRC) is a prevalent disease characterized by significant dysregulation of gene expression. Non-invasive tests that utilize microRNAs (miRNAs) have shown promise for early CRC detection. This study aims to determine the association between miRNAs and key genes in CRC. Methods Two datasets (GSE106817 and GSE23878) were extracted from the NCBI Gene Expression Omnibus database. Penalized logistic regression (PLR) and artificial neural networks (ANN) were used to identify relevant miRNAs and evaluate the classification accuracy of the selected miRNAs. The findings were validated through bipartite miRNA-mRNA interactions. Results Our analysis identified 3 miRNAs: miR-1228, miR-6765-5p, and miR-6787-5p, achieving a total accuracy of over 90%. Based on the results of the mRNA-miRNA interaction network, CDK1 and MAD2L1 were identified as target genes of miR-6787-5p. Conclusions Our results suggest that the identified miRNAs and target genes could serve as non-invasive biomarkers for diagnosing colorectal cancer, pending laboratory confirmation.
Collapse
Affiliation(s)
- Alireza Gharebaghi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leili Tapak
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
- Modeling of Noncommunicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Ranjbar
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Irina Dinu
- School of Public Health, Health Academy, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
46
|
Lu J, Zhang W, Zhu Y, Luo P, Tong X, Xie S, Jiang L, Guo X, Huang J, Gu M, Ding X, Xian S, Huang R, Ji S, Xia Z. Revealing the Therapeutic Potential of Stem Cells in Burn Healing: A Deeper Understanding of the Therapeutic Mechanisms of Epidermal Stem Cells and Mesenchymal Stem Cells. Stem Cells Int 2024; 2024:1914585. [PMID: 39717868 PMCID: PMC11666318 DOI: 10.1155/2024/1914585] [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: 09/14/2023] [Revised: 07/20/2024] [Accepted: 08/19/2024] [Indexed: 12/25/2024] Open
Abstract
Background: Burns are a global public health issue and a major cause of disability and death around the world. Stem cells, which are the undifferentiated cells with the potential for indefinite proliferation and multilineage differentiation, have the ability to replace injured skin and facilitate the wound repair process through paracrine mechanisms. In light of this, the present study aims to conduct a bibliometric analysis in order to identify research hotspots of stem cell-related burns and assess global research tendencies. Methods: To achieve this objective, we retrieved scientific publications on burns associated with stem cells covering the period from January 1, 1978, to October 13, 2022, from the Web of Science Core Collection (WoSCC). Bibliometric analyses, including production and collaboration analyses between countries, institutions, authors, and journals, as well as keyword and topic analyses, were conducted using the bibliometrix R package, CiteSpace, and VOSviewer. Results: A total of 1648 burns associated with stem cell documents were published and listed on WOSCC. The most contributive country, institution, journal, and author were the United States, LV Prasad Eye Institute, Burns, and Scheffer C.G. Tseng, respectively. More importantly, combined with historical direct citation network, trend topic analysis, keyword co-occurrence network, and substantial literature analysis, we eventually summarized the research hotspots and frontiers on burns associated stem cell reasearch. Conclusion: The present study obtained deep insight into the developing trends and research hotspots on burns associated with stem cells, which arouses growing concerns and implies increasing clinical implications. The mechanism and therapeutics of epidermal stem cells (ESCs) for burn wounds and the mechanism of mesenchymal stem cells (MSCs) and MSC-derived exosomes for burns wounds are two research hotspots in this field.
Collapse
Affiliation(s)
- Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Wei Zhang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Pengfei Luo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Sujie Xie
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Luofeng Jiang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Jie Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Minyi Gu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xinran Ding
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| |
Collapse
|
47
|
Chen H. UDP-glucuronosyltransferases 2A3 as a biomarker for ulcerative colitis and colon cancer. Front Genet 2024; 15:1419755. [PMID: 39717480 PMCID: PMC11663933 DOI: 10.3389/fgene.2024.1419755] [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: 04/18/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Background Ulcerative colitis has a serious impact on the quality of life of patients and is more likely to progress to colon cancer. Therefore, early diagnosis and timely intervention are of considerable importance. Methods Gene expression data of active ulcerative colitis were downloaded from the Gene Expression Omnibus (GEO) database, and genes with significant differential expression were identified. Biochemical markers with diagnostic significance were selected through machine learning methods. The expression differences of the selected markers between colon adenocarcinoma (COAD) and healthy control groups in The Cancer Genome Atlas (TCGA) database were analyzed to evaluate their diagnostic value. In addition, the correlation between the selected markers and clinical indicators, as well as their predictive efficacy for the survival of COAD patients, was explored. Results Through machine learning and LASSO regression analysis, UGT2A3 was finally determined as a diagnostic marker for ulcerative colitis. It demonstrated high diagnostic accuracy in both the training set and the external validation set. Furthermore, UGT2A3 was significantly downregulated in COAD tissues compared to normal control tissues. The ROC curve suggested that UGT2A3 could serve as a diagnostic marker for COAD with excellent performance, achieving an AUC of 0.969. Immune infiltration analysis indicated a significant negative correlation between the expression of UGT2A3 and neutrophils. Correlation analysis suggested a link between UGT2A3 and the pathological classification of colon cancer. Survival analysis showed that UGT2A3 is negatively correlated with OS, PPS, and RFS in colon cancer. Conclusion The author identified UGT2A3 as a diagnostic marker for ulcerative colitis through bioinformatics methods, and verified its significant downregulation in colon cancer, as well as its predictive role in the survival of COAD patients. These findings suggest that UGT2A3 may serve not only as a diagnostic marker for ulcerative colitis and colon cancer but also as a potential prognostic indicator for colon cancer.
Collapse
Affiliation(s)
- Hao Chen
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
48
|
Liu X, Hyun Kim J, Li X, Liu R. Application of mesenchymal stem cells exosomes as nanovesicles delivery system in the treatment of breast cancer. Int J Pharm 2024; 666:124732. [PMID: 39304093 DOI: 10.1016/j.ijpharm.2024.124732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
As people's living standards continue to improve and human life span expectancy increases, the incidence and mortality rates of breast cancer are continuously rising. Early detection of breast cancer and targeted therapy for different breast cancer subtypes can significantly reduce the mortality rate and alleviate the suffering of patients. Exosomes are extracellular vesicles secreted by various cells in the body. They participate in physiological and pathological responses by releasing active substances and play an important role in regulating intercellular communication. In recent years, research on exosomes has gradually expanded, and their special membrane structure and targetable characteristics are being increasingly applied in various clinical studies. Mesenchymal stem cells (MSCs)-derived exosomes play an important role in regulating the progression of breast cancer. In this review, we summarize the current treatment methods for breast cancer, the connection between MSCs, exosomes, and breast cancer, as well as the application of exosomes derived from MSCs from different sources in cancer treatment. We highlight how the rational design of modified MSCs-derived exosomes (MSCs-Exos) delivery systems can overcome the uncertainties of stem cell therapy and overcome the clinical translation challenges of nanomaterials. This work aims to promote future research on the application of MSCs-Exos in breast cancer treatment.
Collapse
Affiliation(s)
- Xiaofan Liu
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea; Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - June Hyun Kim
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Rui Liu
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea.
| |
Collapse
|
49
|
He Y, Li R, Yu Y, Xu Z, Gao J, Wang C, Huang C, Qi Z. HucMSC extracellular vesicles increasing SATB 1 to activate the Wnt/β-catenin pathway in 6-OHDA-induced Parkinson's disease model. IUBMB Life 2024; 76:1154-1174. [PMID: 39082886 DOI: 10.1002/iub.2893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 11/22/2024]
Abstract
Parkinson's disease (PD) is a degenerative disorder of the nervous system characterized by the loss of dopaminergic neurons and damage of neurons in the substantia nigra (SN) and striatum, resulting in impaired motor functions. This study aims to investigate how extracellular vesicles (EVs) derived from human umbilical cord mesenchymal stem cells (HucMSC) regulate Special AT-rich sequence-binding protein-1 (SATB 1) and influence Wnt/β-catenin pathway and autophagy in PD model. The PD model was induced by damaging SH-SY5Y cells and mice using 6-OHDA. According to the study, administering EVs every other day for 14 days improved the motor behavior of 6-OHDA-induced PD mice and reduced neuronal damage, including dopaminergic neurons. Treatment with EVs for 12 hours increased the viability of 6-OHDA-induced SH-SY5Y cells. The upregulation of SATB 1 expression with EV treatment resulted in the activation of the Wnt/β-catenin pathway in PD model and led to overexpression of β-catenin. Meanwhile, the expression of LC3 II was decreased, indicating alterations in autophagy. In conclusion, EVs could mitigate neuronal damage in the 6-OHDA-induced PD model by upregulating SATB 1 and activating Wnt/β-catenin pathway while also regulating autophagy. Further studies on the potential therapeutic applications of EVs for PD could offer new insights and strategies.
Collapse
Affiliation(s)
- Ying He
- Medical College, Guangxi University, Nanning, China
- Department of Pharmacy, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Ruicheng Li
- Medical College, Guangxi University, Nanning, China
| | - Yuxi Yu
- Medical College, Guangxi University, Nanning, China
| | - Zhiran Xu
- Translational Medicine Research Center, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaxin Gao
- Medical College, Guangxi University, Nanning, China
| | - Cancan Wang
- Medical College, Guangxi University, Nanning, China
| | - Chusheng Huang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, China
| |
Collapse
|
50
|
Huang S, Li S, Li G, Wang C, Guo X, Zhang J, Liu J, Xu Y, Wang Y. Fabrication and Characterization of Phyllanthus Emblica Extract-Polyvinyl Alcohol/Carboxymethyl Cellulose Sodium Antioxidant Hydrogel and Its Application in Wound Healing. Pharmaceutics 2024; 16:1531. [PMID: 39771510 PMCID: PMC11678665 DOI: 10.3390/pharmaceutics16121531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Phyllanthus emblica is a medicinal and edible plant from the Euphorbiaceae family, notable for its rich content of polyphenols and flavonoids, which provide significant antioxidant properties. To exploit the full antioxidant potential of Phyllanthus emblica, this study developed a hydrogel system incorporating polyvinyl alcohol (PVA) and carboxymethyl cellulose sodium (CMC-Na), integrated with Phyllanthus emblica extract, for the purpose of wound healing. Methods: The extraction process of active ingredients of Phyllanthus emblica was optimized and assessed the antioxidant composition and activity of the extract. A series of hydrogel performance evaluations were performed on the Phyllanthus emblica extract-loaded PVA/CMC-Na hydrogel (AEPE composite hydrogel). Additionally, the wound healing efficacy was evaluated through cell culture experiments and wound healing assays using BALB/C mice. Results: The findings indicated that the extraction of Phyllanthus emblica with 95% ethanol yielded an extract rich in polyphenols, primarily gallic acid and ellagic acid, demonstrating high free radical scavenging capacity and robust antioxidant activity. The hydrogel matrix containing 12% PVA and 1% CMC-Na exhibited excellent physicochemical properties. The optimized AEPE composite hydrogel enabled sustained drug release over a 24 h period, exhibited low cytotoxicity and promoted cell migration. In a mouse dorsal wound healing model, the AEPE composite hydrogel showed pronounced anti-inflammatory and antioxidation effects, enhanced collagen deposition, and ultimately accelerated wound healing. Conclusions: The AEPE composite hydrogel demonstrated strong antioxidant characteristics and significant wound healing potential. Thus, this study could broaden the application prospects of Phyllanthus emblica in wound healing.
Collapse
Affiliation(s)
- Shanqin Huang
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Shanglun Li
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Guoyan Li
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Chenyu Wang
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Xiaohan Guo
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Jing Zhang
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Jing Liu
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Ying Xu
- College of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (S.H.)
| | - Yanchun Wang
- Henan Provincial People’s Hospital, Zhengzhou 450003, China
- School of Medical, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|