1
|
Zhang J, Lu E, Deng L, Zhu Y, Lu X, Li X, Li F, Yan Y, Han JY, Li Y, Zhang Y. Immunological roles for resistin and related adipokines in obesity-associated tumors. Int Immunopharmacol 2024; 142:112911. [PMID: 39232363 DOI: 10.1016/j.intimp.2024.112911] [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: 05/24/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024]
Abstract
Rationale Obesity is an independent risk factor for the occurrence and development of tumors. Obesity is influenced by signaling of adipokines, which are secreted factors from adipocytes and resident immune cells within adipose tissues that mediate lipid metabolism. More recently, adipokines have been implicated in chronic inflammation as well as in tumor formation and growth. Among them, resistin has received increasing attention in research related to the growth and expansion of solid tumors and hematological cancers through various signaling pathways. Objective and findings We reviewed the physiological, biochemical, and immune functions of adipose tissue, with a focus on the structure and expression of resistin and adipokines within multiple adipose cell types, their signaling pathways and putative effects on tumor cells, as well as their in vivo regulation. Current evidence indicates that adipokines such as resistin act as pro-inflammatory factors to stimulate immune cells which, in turn, promotes tumor angiogenesis, connective tissue proliferation, and matrix fibrosis. Concurrently, in states of metabolic dysfunction and lipotoxicity in obese individuals, the numbers and functions of immune cells are compromised, leading to an immunosuppressive environment that fosters tumor cell survival and weak cancer immune monitoring. Conclusion Adipokines such as resistin are important to the development of obesity-related tumors. Clarifying the roles for obesity-related factors in immune regulation and tumor progression may lead to the discovery of novel anti-tumor strategies for targeting obesity factors such as resistin to limit tumor growth and manage obesity, or both.
Collapse
Affiliation(s)
- Jingxin Zhang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Enting Lu
- Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Deng
- Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yaoxuan Zhu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiaoqing Lu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xinyuan Li
- School of Nursing, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fangmei Li
- Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yan Yan
- Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yin Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| | - Yi Zhang
- Department of Gynecology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
2
|
Miao S, Xuan Q, Xie H, Jiang Y, Sun M, Huang W, Li J, Qi H, Li A, Wang Q, Liu Z, Wang R. An Integrated Nomogram Combining Deep Learning and Radiomics for Predicting Malignancy of Pulmonary Nodules Using CT-Derived Nodules and Adipose Tissue: A Multicenter Study. Cancer Med 2024; 13:e70372. [PMID: 39494854 PMCID: PMC11533136 DOI: 10.1002/cam4.70372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Correctly distinguishing between benign and malignant pulmonary nodules can avoid unnecessary invasive procedures. This study aimed to construct a deep learning radiomics clinical nomogram (DLRCN) for predicting malignancy of pulmonary nodules. METHODS One thousand and ninety-eight patients with 6-30 mm pulmonary nodules who received histopathologic diagnosis at 3 centers were included and divided into a primary cohort (PC), an internal test cohort (I-T), and two external test cohorts (E-T1, E-T2). The DLRCN was built by integrating adipose tissue radiomics features, intranodular and perinodular deep learning features, and clinical characteristics for diagnosing malignancy of pulmonary nodules. The least absolute shrinkage and selection operator (LASSO) was used for feature selection. The performance of DLRCN was assessed with respect to its calibration curve, area under the curve (AUC), and decision curve analysis (DCA). Furthermore, we compared it with three radiologists. The net reclassification improvement (NRI), integrated discrimination improvement (IDI), and subgroup analysis were also taken into account. RESULTS The incorporation of adipose tissue radiomics features led to significant NRI and IDI (NRI = 1.028, p < 0.05, IDI = 0.137, p < 0.05). In the I-T, E-T1, and E-T2, the AUCs of DLRCN were 0.946 (95% CI: 0.936, 0.955), 0.948 (95% CI: 0.933, 0.963) and 0.962 (95% CI: 0.945, 0.979), The calibration curve revealed good predictive accuracy between the actual probability and predicted probability (p > 0.05). DCA showed that the DLRCN was clinically useful. Under equal specificity, the sensitivity of DLRCN increased by 8.6% compared to radiologist assessments. The subgroup analysis conducted on adipose tissue radiomics features further demonstrated their supplementary value in determining the malignancy of pulmonary nodules. CONCLUSION The DLRCN demonstrated good performance in predicting the malignancy of pulmonary nodules, which was comparable to radiologist assessments. The adipose tissue radiomics features have notably enhanced the performance of DLRCN.
Collapse
Affiliation(s)
- Shidi Miao
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Qifan Xuan
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Hanbing Xie
- Department of Internal MedicineHarbin Medical University Cancer Hospital, Harbin Medical UniversityHarbinChina
| | - Yuyang Jiang
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Mengzhuo Sun
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Wenjuan Huang
- Department of Internal MedicineHarbin Medical University Cancer Hospital, Harbin Medical UniversityHarbinChina
| | - Jing Li
- Department of GeriatricsThe Second Affiliated Hospital, Harbin Medical UniversityHarbinChina
| | - Hongzhuo Qi
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Ao Li
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Qiujun Wang
- Department of General PracticeThe Second Affiliated Hospital, Harbin Medical UniversityHarbinChina
| | - Zengyao Liu
- Department of Interventional MedicineThe First Affiliated Hospital, Harbin Medical UniversityHarbinChina
| | - Ruitao Wang
- Department of Internal MedicineHarbin Medical University Cancer Hospital, Harbin Medical UniversityHarbinChina
| |
Collapse
|
3
|
Akla N, Veilleux C, Annabi B. The Chemopreventive Impact of Diet-Derived Phytochemicals on the Adipose Tissue and Breast Tumor Microenvironment Secretome. Nutr Cancer 2024:1-17. [PMID: 39300732 DOI: 10.1080/01635581.2024.2401647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Cancer cells-derived extracellular vesicles can trigger the transformation of adipose-derived mesenchymal stem cells (ADMSC) into a pro-inflammatory, cancer-associated adipocyte (CAA) phenotype. Such secretome-mediated crosstalk between the adipose tissue and the tumor microenvironment (TME) therefore impacts tumor progression and metastatic processes. In addition, emerging roles of diet-derived phytochemicals, especially epigallocatechin-3-gallate (EGCG) among other polyphenols, in modulating exosome-mediated metabolic and inflammatory signaling pathways have been highlighted. Here, we discuss how selected diet-derived phytochemicals could alter the secretome signature as well as the crosstalk dynamics between the adipose tissue and the TME, with a focus on breast cancer. Their broader implication in the chemoprevention of obesity-related cancers is also discussed.
Collapse
Affiliation(s)
- Naoufal Akla
- Laboratoire d'Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montreal, Canada
| | - Carolane Veilleux
- Laboratoire d'Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montreal, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montreal, Canada
| |
Collapse
|
4
|
WANG YUN, LI XIAOJIANG, LIU DALONG, WANG ZHIFENG, XIA JICHEN, WANG LIJUN, ZHANG XUDONG. Research progress on the role of adipocyte exosomes in cancer progression. Oncol Res 2024; 32:1649-1660. [PMID: 39308520 PMCID: PMC11413817 DOI: 10.32604/or.2024.043482] [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: 07/03/2023] [Accepted: 04/07/2024] [Indexed: 09/25/2024] Open
Abstract
Exosomes, minute vesicles ubiquitously released by diverse cell types, serve as critical mediators in intercellular communication. Their pathophysiological relevance, especially in malignancies, has garnered significant attention. A meticulous exploration of the exosomal impact on cancer development has unveiled avenues for innovative and clinically valuable techniques. The cargo conveyed by exosomes exerts transformative effects on both local and distant microenvironments, thereby influencing a broad spectrum of biological responses in recipient cells. These membrane-bound extracellular vesicles (EVs) play a pivotal role in delivering bioactive molecules among cells and organs. Cellular and biological processes in recipient cells, ranging from stromal cell reprogramming to immunological responses, extracellular matrix formation, and modulation of cancer cell activation, expansion, and metastasis, are subject to exosome-mediated cell-to-cell communication. Moreover, exosomes have been implicated in endowing cancer cells with resistance to treatment. Extensive research has explored the potential of exosomes as therapeutic targets and diagnostic indicators. This comprehensive review seeks to provide an in-depth understanding of the pivotal components and roles of exosomes in tumorigenesis, growth, progression, and therapeutic responses. The insights into the multifaceted involvement of exosomes in malignant cancers are essential for the scientific community, fostering the development of novel therapeutic and diagnostic strategies in the relentless pursuit of cancer.
Collapse
Affiliation(s)
- YUN WANG
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130021, China
| | - XIAOJIANG LI
- Department of Orthopaedics, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130021, China
| | - DALONG LIU
- Department of Orthopaedics, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130021, China
| | - ZHIFENG WANG
- Department of Internal Medicine, Changchun Chaoyang District Hospital of Traditional Chinese Medicine, Changchun, 130061, China
| | - JICHEN XIA
- Department of Orthopedics and Traumatology, Jilin Integrated Traditional Chinese and Western Medicine Hospital of Jilin Province, Jilin, 132012, China
| | - LIJUN WANG
- Department of Oncology, Liaoyuan Second People’s Hospital, Liaoyuan, 136299, China
| | - XUDONG ZHANG
- Department of Brain Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130021, China
| |
Collapse
|
5
|
Qi H, Xuan Q, Liu P, An Y, Huang W, Miao S, Wang Q, Liu Z, Wang R. Deep Learning Radiomics Features of Mediastinal Fat and Pulmonary Nodules on Lung CT Images Distinguish Benignancy and Malignancy. Biomedicines 2024; 12:1865. [PMID: 39200329 PMCID: PMC11352131 DOI: 10.3390/biomedicines12081865] [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: 07/19/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
This study investigated the relationship between mediastinal fat and pulmonary nodule status, aiming to develop a deep learning-based radiomics model for diagnosing benign and malignant pulmonary nodules. We proposed a combined model using CT images of both pulmonary nodules and the fat around the chest (mediastinal fat). Patients from three centers were divided into training, validation, internal testing, and external testing sets. Quantitative radiomics and deep learning features from CT images served as predictive factors. A logistic regression model was used to combine data from both pulmonary nodules and mediastinal adipose regions, and personalized nomograms were created to evaluate the predictive performance. The model incorporating mediastinal fat outperformed the nodule-only model, with C-indexes of 0.917 (training), 0.903 (internal testing), 0.942 (external testing set 1), and 0.880 (external testing set 2). The inclusion of mediastinal fat significantly improved predictive performance (NRI = 0.243, p < 0.05). A decision curve analysis indicated that incorporating mediastinal fat features provided greater patient benefits. Mediastinal fat offered complementary information for distinguishing benign from malignant nodules, enhancing the diagnostic capability of this deep learning-based radiomics model. This model demonstrated strong diagnostic ability for benign and malignant pulmonary nodules, providing a more accurate and beneficial approach for patient care.
Collapse
Affiliation(s)
- Hongzhuo Qi
- School of Computer Science and Technology, Harbin University of Science and Technology, Harbin 150080, China; (H.Q.); (Y.A.); (S.M.)
| | - Qifan Xuan
- School of Computer Science and Technology, Harbin University of Science and Technology, Harbin 150080, China; (H.Q.); (Y.A.); (S.M.)
| | - Pingping Liu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, China; (P.L.); (W.H.); (R.W.)
| | - Yunfei An
- School of Computer Science and Technology, Harbin University of Science and Technology, Harbin 150080, China; (H.Q.); (Y.A.); (S.M.)
| | - Wenjuan Huang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, China; (P.L.); (W.H.); (R.W.)
| | - Shidi Miao
- School of Computer Science and Technology, Harbin University of Science and Technology, Harbin 150080, China; (H.Q.); (Y.A.); (S.M.)
| | - Qiujun Wang
- Department of General Practice, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China;
| | - Zengyao Liu
- Department of Interventional Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin 150086, China;
| | - Ruitao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, China; (P.L.); (W.H.); (R.W.)
| |
Collapse
|
6
|
Lin H, Zhou J, Ding T, Zhu Y, Wang L, Zhong T, Wang X. Therapeutic potential of extracellular vesicles from diverse sources in cancer treatment. Eur J Med Res 2024; 29:350. [PMID: 38943222 PMCID: PMC11212438 DOI: 10.1186/s40001-024-01937-x] [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: 02/07/2024] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
Cancer, a prevalent and complex disease, presents a significant challenge to the medical community. It is characterized by irregular cell differentiation, excessive proliferation, uncontrolled growth, invasion of nearby tissues, and spread to distant organs. Its progression involves a complex interplay of several elements and processes. Extracellular vesicles (EVs) serve as critical intermediaries in intercellular communication, transporting critical molecules such as lipids, RNA, membrane, and cytoplasmic proteins between cells. They significantly contribute to the progression, development, and dissemination of primary tumors by facilitating the exchange of information and transmitting signals that regulate tumor growth and metastasis. However, EVs do not have a singular impact on cancer; instead, they play a multifaceted dual role. Under specific circumstances, they can impede tumor growth and influence cancer by delivering oncogenic factors or triggering an immune response. Furthermore, EVs from different sources demonstrate distinct advantages in inhibiting cancer. This research examines the biological characteristics of EVs and their involvement in cancer development to establish a theoretical foundation for better understanding the connection between EVs and cancer. Here, we discuss the potential of EVs from various sources in cancer therapy, as well as the current status and future prospects of engineered EVs in developing more effective cancer treatments.
Collapse
Affiliation(s)
- Haihong Lin
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Jun Zhou
- Department of Laboratory Medicine, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550000, China
| | - Tao Ding
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Yifan Zhu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Lijuan Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China.
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| | - Xiaoling Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China.
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
7
|
Jansen J, Garmyn M, Güvenç C. The Effect of Body Mass Index on Melanoma Biology, Immunotherapy Efficacy, and Clinical Outcomes: A Narrative Review. Int J Mol Sci 2024; 25:6433. [PMID: 38928137 PMCID: PMC11204248 DOI: 10.3390/ijms25126433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Recent studies indicate that a higher body mass index (BMI) might correlate with improved responses to melanoma treatment, especially with immune checkpoint inhibitors (ICIs), despite the general association of obesity with an increased risk of cancer and higher mortality rates. This review examines the paradoxical relationship between BMI and clinical outcomes in melanoma patients by exploring molecular links, the efficacy of immunotherapy, and patient survival outcomes. Our comprehensive literature search across the PubMed and Embase databases revealed a consistent pattern: increased BMI is associated with a better prognosis in melanoma patients undergoing ICI treatment. This "obesity paradox" might be explained by the metabolic and immunological changes in obesity, which could enhance the effectiveness of immunotherapy in treating melanoma. The findings highlight the complexity of the interactions between obesity and melanoma, suggesting that adipose tissue may modulate the immune response and treatment sensitivity favorably. Our review highlights the need for personalized treatment strategies that consider the metabolic profiles of patients and calls for further research to validate BMI as a prognostic factor in clinical settings. This nuanced approach to the obesity paradox in melanoma could significantly impact treatment planning and patient management.
Collapse
Affiliation(s)
| | | | - Canan Güvenç
- Department of Dermatology, University Hospitals Leuven, 3000 Leuven, Belgium; (J.J.); (M.G.)
| |
Collapse
|
8
|
Arora A, Sharma V, Gupta R, Aggarwal A. Isolation and Characterization of Extracellular Vesicles Derived from Ex Vivo Culture of Visceral Adipose Tissue. Bio Protoc 2024; 14:e5011. [PMID: 38873012 PMCID: PMC11166539 DOI: 10.21769/bioprotoc.5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/15/2024] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of nanoparticles possessing a lipid bilayer membrane that plays a significant role in intercellular communication by transferring their cargoes, consisting of peptides, proteins, fatty acids, DNA, and RNA, to receiver cells. Isolation of EVs is cumbersome and time-consuming due to their nano size and the co-isolation of small molecules along with EVs. This is why current protocols for the isolation of EVs are unable to provide high purity. So far, studies have focused on EVs derived from cell supernatants or body fluids but are associated with a number of limitations. Cell lines with a high passage number cannot be considered as representative of the original cell type, and EVs isolated from those can present distinct properties and characteristics. Additionally, cultured cells only have a single cell type and do not possess any cellular interactions with other types of cells, which normally exist in the tissue microenvironment. Therefore, studies involving the direct EVs isolation from whole tissues can provide a better understanding of intercellular communication in vivo. This underscores the critical need to standardize and optimize protocols for isolating and characterizing EVs from tissues. We have developed a differential centrifugation-based technique to isolate and characterize EVs from whole adipose tissue, which can be potentially applied to other types of tissues. This may help us to better understand the role of EVs in the tissue microenvironment in both diseased and normal conditions. Key features • Isolation of tissue-derived extracellular vesicles from ex vivo culture of visceral adipose tissue or any whole tissue. • Microscopic visualization of extracellular vesicles' morphology without dehydration steps, with minimum effect on their shape. • Flow cytometry approach to characterize the extracellular vesicles using specific protein markers, as an alternative to the time-consuming western blot.
Collapse
Affiliation(s)
- Ankita Arora
- Department of Anatomy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vinit Sharma
- Department of Anatomy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajesh Gupta
- Department of GI Surgery, HPB and Liver Transplantation, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anjali Aggarwal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
9
|
Ye Z, Chen W, Li G, Huang J, Lei J. Tissue-derived extracellular vesicles in cancer progression: mechanisms, roles, and potential applications. Cancer Metastasis Rev 2024; 43:575-595. [PMID: 37851319 DOI: 10.1007/s10555-023-10147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
Extracellular vesicles (EVs) are small lipid bilayer-enclosed vesicles that mediate vital cellular communication by transferring cargo between cells. Among these, tissue-derived extracellular vesicles (Ti-EVs) stand out due to their origin from the tissue microenvironment, providing a more accurate reflection of changes in this setting. This unique advantage makes Ti-EVs valuable in investigating the intricate relationship between extracellular vesicles and cancer progression. Despite considerable research efforts exploring the association between Ti-EVs and cancers, a comprehensive clustering or grouping of these studies remains lacking. In this review, we aim to fill this gap by presenting a comprehensive synthesis of the mechanisms underlying Ti-EV generation, release, and transport within cancer tissues. Moreover, we delve into the pivotal roles that Ti-EVs play in cancer progression, shedding light on their potential as diagnostic and therapeutic tools. The review culminates in the construction of a comprehensive functional spectrum of Ti-EVs, providing a valuable reference for future research endeavors. By summarizing the current state of knowledge on Ti-EVs and their significance in tumor biology, this work contributes to a deeper understanding of cancer microenvironment dynamics and opens up avenues for harnessing Ti-EVs in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Ziyang Ye
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Muttiah B, Ng SL, Lokanathan Y, Ng MH, Law JX. Extracellular Vesicles in Breast Cancer: From Intercellular Communication to Therapeutic Opportunities. Pharmaceutics 2024; 16:654. [PMID: 38794316 PMCID: PMC11125876 DOI: 10.3390/pharmaceutics16050654] [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: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer, a multifaceted and heterogeneous disease, poses significant challenges in terms of understanding its intricate resistance mechanisms and devising effective therapeutic strategies. This review provides a comprehensive overview of the intricate landscape of extracellular vesicles (EVs) in the context of breast cancer, highlighting their diverse subtypes, biogenesis, and roles in intercellular communication within the tumour microenvironment (TME). The discussion spans various aspects, from EVs and stromal cells in breast cancer to their influence on angiogenesis, immune response, and chemoresistance. The impact of EV production in different culture systems, including two dimensional (2D), three dimensional (3D), and organoid models, is explored. Furthermore, this review delves into the therapeutic potential of EVs in breast cancer, presenting emerging strategies such as engineered EVs for gene delivery, nanoplatforms for targeted chemotherapy, and disrupting tumour derived EVs as a treatment approach. Understanding these complex interactions of EV within the breast cancer milieu is crucial for identifying resistance mechanisms and developing new therapeutic targets.
Collapse
Affiliation(s)
- Barathan Muttiah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
11
|
Li W, Zhu J, Li J, Jiang Y, Sun J, Xu Y, Pan H, Zhou Y, Zhu J. Research advances of tissue-derived extracellular vesicles in cancers. J Cancer Res Clin Oncol 2024; 150:184. [PMID: 38598014 PMCID: PMC11006789 DOI: 10.1007/s00432-023-05596-z] [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: 11/16/2023] [Accepted: 12/23/2023] [Indexed: 04/11/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) can mediate cell-to-cell communication and affect various physiological and pathological processes in both parent and recipient cells. Currently, extensive research has focused on the EVs derived from cell cultures and various body fluids. However, insufficient attention has been paid to the EVs derived from tissues. Tissue EVs can reflect the microenvironment of the specific tissue and the cross-talk of communication among different cells, which can provide more accurate and comprehensive information for understanding the development and progression of diseases. METHODS We review the state-of-the-art technologies involved in the isolation and purification of tissue EVs. Then, the latest research progress of tissue EVs in the mechanism of tumor occurrence and development is presented. And finally, the application of tissue EVs in the clinical diagnosis and treatment of cancer is anticipated. RESULTS We evaluate the strengths and weaknesses of various tissue processing and EVs isolation methods, and subsequently analyze the significance of protein characterization in determining the purity of tissue EVs. Furthermore, we focus on outlining the importance of EVs derived from tumor and adipose tissues in tumorigenesis and development, as well as their potential applications in early tumor diagnosis, prognosis, and treatment. CONCLUSION When isolating and characterizing tissue EVs, the most appropriate protocol needs to be specified based on the characteristics of different tissues. Tissue EVs are valuable in the diagnosis, prognosis, and treatment of tumors, and the potential risks associated with tissue EVs need to be considered as therapeutic agents.
Collapse
Affiliation(s)
- Wei Li
- Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, People's Republic of China
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Jingyao Zhu
- School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Jiayuan Li
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Yiyun Jiang
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Jiuai Sun
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Yan Xu
- Research Laboratory for Functional Nanomaterial, National Engineering Research Center for Nanotechnology, Shanghai, 200241, People's Republic of China
| | - Hongzhi Pan
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China.
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 200120, People's Republic of China.
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| | - Jun Zhu
- School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
- Research Laboratory for Functional Nanomaterial, National Engineering Research Center for Nanotechnology, Shanghai, 200241, People's Republic of China.
| |
Collapse
|
12
|
Long Y, Mao C, Liu S, Tao Y, Xiao D. Epigenetic modifications in obesity-associated diseases. MedComm (Beijing) 2024; 5:e496. [PMID: 38405061 PMCID: PMC10893559 DOI: 10.1002/mco2.496] [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: 08/03/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
The global prevalence of obesity has reached epidemic levels, significantly elevating the susceptibility to various cardiometabolic conditions and certain types of cancer. In addition to causing metabolic abnormalities such as insulin resistance (IR), elevated blood glucose and lipids, and ectopic fat deposition, obesity can also damage pancreatic islet cells, endothelial cells, and cardiomyocytes through chronic inflammation, and even promote the development of a microenvironment conducive to cancer initiation. Improper dietary habits and lack of physical exercise are important behavioral factors that increase the risk of obesity, which can affect gene expression through epigenetic modifications. Epigenetic alterations can occur in early stage of obesity, some of which are reversible, while others persist over time and lead to obesity-related complications. Therefore, the dynamic adjustability of epigenetic modifications can be leveraged to reverse the development of obesity-associated diseases through behavioral interventions, drugs, and bariatric surgery. This review provides a comprehensive summary of the impact of epigenetic regulation on the initiation and development of obesity-associated cancers, type 2 diabetes, and cardiovascular diseases, establishing a theoretical basis for prevention, diagnosis, and treatment of these conditions.
Collapse
Affiliation(s)
- Yiqian Long
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Chao Mao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaChina
| | - Shuang Liu
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaChina
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Department of Thoracic SurgerySecond Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Desheng Xiao
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
13
|
Cho YE, Chen S, Crouch K, Yun J, Klingelhutz A. Impact of Aging and a High-Fat Diet on Adipose-Tissue-Derived Extracellular Vesicle miRNA Profiles in Mice. Biomedicines 2024; 12:100. [PMID: 38255206 PMCID: PMC10813715 DOI: 10.3390/biomedicines12010100] [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: 11/04/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Middle-aged adults have the highest obesity rates, leading to significant health complications in later years. Obesity triggers the release of altered molecules, including extracellular vesicles (EVs) from excess adipose tissue (AT), contributing to various health complications. In this study, we assessed the effects of age and a high-fat diet on AT-derived EV miRNA profiles to understand their potential roles in aging and obesity. METHOD C57BL/6 male mice were subjected to a normal chow diet (NCD) or a high-fat diet (HFD) for either 10-12 weeks (young mice, n = 10) or 50-61 weeks (middle-aged mice, n = 12). After evaluating metabolic characteristics, peri-gonadal white AT was isolated and cultured to obtain EVs. AT-derived EV miRNAs were profiled using a NanoString miRNA panel (n = 599). RESULTS Middle-aged mice exhibited obesity regardless of diet. Young mice fed an HFD showed similar metabolic traits to middle-aged mice. In the NCD group, 131 differentially expressed miRNAs (DE-miRNAs) emerged in middle-aged mice compared to young mice, including miR-21, miR-148a, and miR-29a, associated with cancer, neuro/psychological disorders, and reproductive diseases. In the HFD group, 55 DE-miRNAs were revealed in middle-aged mice compared to young mice. These miRNAs were associated with significantly suppressed IGF1R activity. CONCLUSION This study demonstrates the potential significant impact of miRNAs of AT EVs on aging- and obesity-related diseases.
Collapse
Affiliation(s)
- Young-Eun Cho
- College of Nursing, The University of Iowa, 50 Newton Road, Iowa City, IA 52242, USA
| | - Shaoshuai Chen
- College of Nursing, The University of Iowa, 50 Newton Road, Iowa City, IA 52242, USA
| | - Keith Crouch
- College of Nursing, The University of Iowa, 50 Newton Road, Iowa City, IA 52242, USA
| | - Joseph Yun
- Predictiv Care, Inc., 800 West El Camino Real, Mountain View, CA 94040, USA
| | - Aloysius Klingelhutz
- Department of Microbiology and Immunology, College of Medicine, The University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| |
Collapse
|
14
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
15
|
Zhai S, Li X, Lin T. Obese Mouse Fat Cell-derived Extracellular Vesicles Transport miR-99a-5p to Mitigate the Proliferation and Migration of Non-small Cell Lung Cancer Cells. Comb Chem High Throughput Screen 2024; 27:214-226. [PMID: 36927435 DOI: 10.2174/1386207326666230316103604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/29/2022] [Accepted: 01/20/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVE Fat cells-derived extracellular vesicles (FC-EVs) play a role in regulating the tumor microenvironment in cancers by transporting RNAs. MicroRNAs (miRNAs) are vital regulators of cancer development. This study was conducted to explore the role of FC-EVs in the proliferation and migration of non-small cell lung cancer (NSCLC) cells, providing targets for NSCLC treatment. METHODS The obese mouse model was established via high-fat diet (HFD), followed by separation and characterization of FC-EVs (HFD-EVs). The levels of miR-99a-5p, precursor-miR-99a-5p, and heparan sulfate-glucosamine 3-sulfotransferase 3B1 (HS3ST3B1) were measured by RT-qPCR or Western blot assay. Cell proliferation and migration were evaluated by 3-(4, 5-dimethylthiazol- 2-yl)-2, 5-diphenyltetrazolium bromide and wound healing assays. The expression of Cy3-labeled miR-99a-5p in A549 cells (one NSCLC cell line) was observed via confocal microscopy. The binding of miR-99a-5p to HS3ST3B1 was analyzed by the dual luciferase assay. Rescue experiments were performed to confirm the role of HS3ST3B1 in NSCLC cells. RESULTS miR-99a-5p was upregulated in adipose tissues, FCs, and HFD-EVs. HFD-EVs mitigated the proliferation and migration of NSCLC cells. HFD-EVs transported miR-99a-5p into A549 cells, which upregulated miR-99a-5p expression and inhibited HS3ST3B1 expression in A549 cells. HS3ST3B1 overexpression reversed the inhibition of HFD-EVs on the proliferation and migration of NSCLC cells. CONCLUSION HFD-EVs transported miR-99a-5p into NSCLC cells and inhibited HS3ST3B1, thereby inhibiting proliferation and migration of NSCLC cells.
Collapse
Affiliation(s)
- Shengping Zhai
- Department of General Practice, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xiaoping Li
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Tiantian Lin
- Respiratory Intensive Care Unit, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| |
Collapse
|
16
|
Alvarez-Artime A, Garcia-Soler B, Gonzalez-Menendez P, Fernandez-Vega S, Cernuda-Cernuda R, Hevia D, Mayo JC, Sainz RM. Castration promotes the browning of the prostate tumor microenvironment. Cell Commun Signal 2023; 21:267. [PMID: 37770940 PMCID: PMC10536697 DOI: 10.1186/s12964-023-01294-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Adipose tissue has gained attention due to its potential paracrine role. Periprostatic adipose tissue surrounds the prostate and the prostatic urethra, and it is an essential player in prostate cancer progression. Since obesity is directly related to human tumor progression, and adipose tissue depots are one of the significant components of the tumor microenvironment, the molecular mediators of the communication between adipocytes and epithelial cells are in the spotlight. Although periprostatic white adipose tissue contributes to prostate cancer progression, brown adipose tissue (BAT), which has beneficial effects in metabolic pathologies, has been scarcely investigated concerning cancer progression. Given that adipose tissue is a target of androgen signaling, the actual role of androgen removal on the periprostatic adipose tissue was the aim of this work. METHODS Surgical castration of the transgenic adenocarcinoma of the mouse prostate (TRAMP) was employed. By histology examination and software analysis, WAT and BAT tissue was quantified. 3T3-like adipocytes were used to study the role of Casodex® in modifying adipocyte differentiation and to investigate the function of the secretome of adipocytes on the proliferation of androgen-dependent and independent prostate cancer cells. Finally, the role of cell communication was assayed by TRAMP-C1 xenograft implanted in the presence of 3T3-like adipocytes. RESULTS Androgen removal increases brown/beige adipose tissue in the fat immediately surrounding the prostate glands of TRAMP mice, concomitant with an adjustment of the metabolism. Castration increases body temperature, respiratory exchange rate, and energy expenditure. Also, in vitro, it is described that blocking androgen signaling by Casodex® increases the uncoupling protein 1 (UCP1) marker in 3T3-like adipocytes. Finally, the effect of brown/beige adipocyte secretome was studied on the proliferation of prostate cancer cells in vivo and in vitro. The secretome of brown/beige adipocytes reduces the proliferation of prostate cancer cells mediated partly by the secretion of extracellular vesicles. CONCLUSIONS Consequently, we concluded that hampering androgen signaling plays a crucial role in the browning of the periprostatic adipose tissue. Also, the presence of brown adipocytes exhibits the opposite effect to that of white adipocytes in vitro regulating processes that govern the mechanisms of cell proliferation of prostate cancer cells. And finally, promoting the browning of adipose tissue in the periprostatic adipose tissue might be a way to handle prostate cancer cell progression. Video Abstract.
Collapse
Affiliation(s)
- Alejandro Alvarez-Artime
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Belen Garcia-Soler
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Pedro Gonzalez-Menendez
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Sheila Fernandez-Vega
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Rafael Cernuda-Cernuda
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - David Hevia
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Juan C Mayo
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain.
| | - Rosa M Sainz
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain.
| |
Collapse
|
17
|
Sepúlveda F, Mayorga-Lobos C, Guzmán K, Durán-Jara E, Lobos-González L. EV-miRNA-Mediated Intercellular Communication in the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:13085. [PMID: 37685891 PMCID: PMC10487525 DOI: 10.3390/ijms241713085] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023] Open
Abstract
Cancer research has prioritized the study of the tumor microenvironment (TME) as a crucial area of investigation. Understanding the communication between tumor cells and the various cell types within the TME has become a focal point. Bidirectional communication processes between these cells support cellular transformation, as well as the survival, invasion, and metastatic dissemination of tumor cells. Extracellular vesicles are lipid bilayer structures secreted by cells that emerge as important mediators of this cell-to-cell communication. EVs transfer their molecular cargo, including proteins and nucleic acids, and particularly microRNAs, which play critical roles in intercellular communication. Tumor-derived EVs, for example, can promote angiogenesis and enhance endothelial permeability by delivering specific miRNAs. Moreover, adipocytes, a significant component of the breast stroma, exhibit high EV secretory activity, which can then modulate metabolic processes, promoting the growth, proliferation, and migration of tumor cells. Comprehensive studies investigating the involvement of EVs and their miRNA cargo in the TME, as well as their underlying mechanisms driving tumoral capacities, are necessary for a deeper understanding of these complex interactions. Such knowledge holds promise for the development of novel diagnostic and therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Francisca Sepúlveda
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610615, Chile; (F.S.); (C.M.-L.); (K.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
| | - Cristina Mayorga-Lobos
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610615, Chile; (F.S.); (C.M.-L.); (K.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
- Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380492, Chile
| | - Kevin Guzmán
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610615, Chile; (F.S.); (C.M.-L.); (K.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
- Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380492, Chile
| | - Eduardo Durán-Jara
- Subdepartamento de Genética Molecular, Instituto de Salud Pública de Chile, Santiago 7780050, Chile;
| | - Lorena Lobos-González
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610615, Chile; (F.S.); (C.M.-L.); (K.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
| |
Collapse
|
18
|
Lei Y, Lin L, Cheng S, Shao Q, Ding C, Zuo R, Chen W, Liao Q, Liu G. Acute inflammatory reaction during anti-angiogenesis therapy combined with immunotherapy as a possible indicator of the therapeutic effect: Three case reports and literature review. Front Oncol 2023; 13:1072480. [PMID: 37124541 PMCID: PMC10140593 DOI: 10.3389/fonc.2023.1072480] [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: 10/17/2022] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
The posterior line treatment of unresectable advanced or metastatic gastrointestinal (GI) tumors has always been a challenging point. In particular, for patients with microsatellite stable (MSS)/mismatch repair proficient (pMMR) 0GI tumors, the difficulty of treatment is exacerbated due to their insensitivity to immune drugs. Accordingly, finding a new comprehensive therapy to improve the treatment effect is urgent. In this study, we report the treatment histories of three patients with MSS/pMMR GI tumors who achieved satisfactory effects by using a comprehensive treatment regimen of apatinib combined with camrelizumab and TAS-102 after the failure of first- or second-line regimens. The specific contents of the treatment plan were as follows: apatinib (500 mg/d) was administered orally for 10 days, followed by camrelizumab (200 mg, ivgtt, day 1, 14 days/cycle) and TAS-102 (20 mg, oral, days 1-21, 28 days/cycle). Apatinib (500 mg/d) was maintained during treatment. Subsequently, we discuss the possible mechanism of this combination and review the relevant literature, and introduce clinical trials on anti-angiogenesis therapy combined with immunotherapy.
Collapse
Affiliation(s)
- Yihui Lei
- The School of Clinical Medical, Fujian Medical University, Fuzhou, Fujian, China
| | - Li Lin
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shuyu Cheng
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Qiming Shao
- The School of Clinical Medical, Fujian Medical University, Fuzhou, Fujian, China
| | - Chenchun Ding
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Renjie Zuo
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Weiping Chen
- The School of Clinical Medical, Fujian Medical University, Fuzhou, Fujian, China
| | - Quan Liao
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Guoyan Liu
- The School of Clinical Medical, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
19
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
20
|
Barone I, Gelsomino L, Accattatis FM, Giordano F, Gyorffy B, Panza S, Giuliano M, Veneziani BM, Arpino G, De Angelis C, De Placido P, Bonofiglio D, Andò S, Giordano C, Catalano S. Analysis of circulating extracellular vesicle derived microRNAs in breast cancer patients with obesity: a potential role for Let-7a. J Transl Med 2023; 21:232. [PMID: 37004031 PMCID: PMC10064709 DOI: 10.1186/s12967-023-04075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND The incidence of obesity, a known risk factor for several metabolic and chronic diseases, including numerous malignancies, has risen sharply in the world. Various clinical studies demonstrate that excessive Body Mass Index (BMI) may worsen the incidence, prognosis, and mortality rates of breast cancer. Thus, understanding the link tying up obesity and breast cancer onset and progression is critically important, as it can impact patients' survival and quality of life. Recently, circulating extracellular vesicle (EV) derived miRNAs have attracted much attention for their diagnostic, prognostic and therapeutic potential in oncology research. Although the potential role of EV-derived miRNAs in the early detection of breast cancer has been repeatedly mentioned, screening of miRNAs packaged within serum EVs has not yet been reported in patients with obesity. METHODS Circulating EVs were isolated from normal weight (NW), and overweight/obese (OW/Ob) breast cancer patients and characterized by Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA), and protein marker expression. Evaluation of EV-associated miRNAs was conducted in a screening (RNA-seq) and a validation (qRT-PCR) cohort. Bioinformatic analysis was performed to uncover significantly enriched biological processes, molecular functions and pathways. ROC and Kaplain-Meier survival analyses were used for clinical significance. RESULTS Comparison of serum EV-derived miRNAs from NW and OW/Ob patients detected seven differentially expressed miRNAs (let-7a-5p, miR-122-5p, miR-30d-5p, miR-126-3p, miR-27b-3p, miR-4772-3p, and miR-10a-5p) in the screening cohort. GO analysis revealed the enrichment of protein phosphorylation, intracellular signal transduction, signal transduction, and vesicle-mediated transport among the top biological processes. In addition, the target genes were significantly enriched in pathways related to PI3K/Akt, growth hormones, and insulin signalings, which are all involved in obesity-related diseases and/or breast cancer progression. In the validation cohort, qRT-PCR confirmed a significant down-regulation of EV-derived let-7a in the serum of OW/Ob breast cancer patients compared to NW patients. Let-7a levels also exhibited a negative correlation with BMI values. Importantly, decreased let-7a miRNA expression was associated with higher tumor grade and poor survival in patients with breast cancer. CONCLUSION These results suggest that serum-EV derived miRNAs may reflect a differential profile in relation to a patient's BMI, which, once validated in larger cohorts of patients, could provide insights into novel specific biomarkers and innovative targets to prevent the progression of obesity-mediated breast cancer.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Felice Maria Accattatis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Balazs Gyorffy
- Departments of Bioinformatics and Pediatrics, Semmelweis University, 1094, Budapest, Hungary
- TTK Cancer Biomarker Research Group, 1117, Budapest, Hungary
| | - Salvatore Panza
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
| |
Collapse
|
21
|
Extracellular Vesicles as Carriers of Adipokines and Their Role in Obesity. Biomedicines 2023; 11:biomedicines11020422. [PMID: 36830957 PMCID: PMC9953604 DOI: 10.3390/biomedicines11020422] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Extracellular vesicles (EVs) have lately arisen as new metabolic players in energy homeostasis participating in intercellular communication at the local and distant levels. These nanosized lipid bilayer spheres, carrying bioactive molecular cargo, have somehow changed the paradigm of biomedical research not only as a non-classic cell secretion mechanism, but as a rich source of biomarkers and as useful drug-delivery vehicles. Although the research about the role of EVs on metabolism and its deregulation on obesity and associated pathologies lagged slightly behind other diseases, the knowledge about their function under normal and pathological homeostasis is rapidly increasing. In this review, we are focusing on the current research regarding adipose tissue shed extracellular vesicles including their characterization, size profile, and molecular cargo content comprising miRNAs and membrane and intra-vesicular proteins. Finally, we will focus on the functional aspects attributed to vesicles secreted not only by adipocytes, but also by other cells comprising adipose tissue, describing the evidence to date on the deleterious effects of extracellular vesicles released by obese adipose tissue both locally and at the distant level by interacting with other peripheral organs and even at the central level.
Collapse
|
22
|
Yanina IY, Nikolaev VV, Zakharova OA, Borisov AV, Dvoretskiy KN, Berezin KV, Kochubey VI, Kistenev YV, Tuchin VV. Measurement and Modeling of the Optical Properties of Adipose Tissue in the Terahertz Range: Aspects of Disease Diagnosis. Diagnostics (Basel) 2022; 12:2395. [PMID: 36292084 PMCID: PMC9600075 DOI: 10.3390/diagnostics12102395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
In this paper, the measurement and modeling of optical properties in the terahertz (THz) range of adipose tissue and its components with temperature changes were performed. Spectral measurements were made in the frequency range 0.25-1 THz. The structural models of main triglycerides of fatty acids are constructed using the B3LYP/6-31G(d) method and the Gaussian03, Revision B.03 program. The optical density (OD) of adipose tissue samples decreases as temperature increases, which can be associated mostly with the dehydration of the sample. Some inclusion of THz wave scattering suppression into the OD decrease can also be expected due to refractive index matching provided by free fatty acids released from adipocytes at thermally induced cell lipolysis. It was shown that the difference between the THz absorption spectra of water and fat makes it possible to estimate the water content in adipose tissue. The proposed model was verified on the basis of molecular modeling and a comparison with experimental data for terahertz spectra of adipose tissue during its heating. Knowing the exact percentage of free and bound water in adipose tissue can help diagnose and monitor diseases, such as diabetes, obesity, and cancer.
Collapse
Affiliation(s)
- Irina Y. Yanina
- Institute of Physics, Saratov State University, 410012 Saratov, Russia
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
| | - Viktor V. Nikolaev
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
| | - Olga A. Zakharova
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
| | - Alexei V. Borisov
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
| | | | - Kirill V. Berezin
- Institute of Physics, Saratov State University, 410012 Saratov, Russia
| | - Vyacheslav I. Kochubey
- Institute of Physics, Saratov State University, 410012 Saratov, Russia
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
| | - Yuri V. Kistenev
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
| | - Valery V. Tuchin
- Institute of Physics, Saratov State University, 410012 Saratov, Russia
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, 634050 Tomsk, Russia
- Institute of Precision Mechanics and Control, FRC “Saratov Scientific Centre of the Russian Academy of Sciences”, 410028 Saratov, Russia
| |
Collapse
|
23
|
Fontana F, Anselmi M, Carollo E, Sartori P, Procacci P, Carter D, Limonta P. Adipocyte-Derived Extracellular Vesicles Promote Prostate Cancer Cell Aggressiveness by Enabling Multiple Phenotypic and Metabolic Changes. Cells 2022; 11:cells11152388. [PMID: 35954232 PMCID: PMC9368412 DOI: 10.3390/cells11152388] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 12/12/2022] Open
Abstract
Background: In recent decades, obesity has widely emerged as an important risk factor for prostate cancer (PCa). Adipose tissue and PCa cells have been shown to orchestrate a complex interaction network to support tumor growth and evolution; nonetheless, the study of this communication has only been focused on soluble factors, although increasing evidence highlights the key role of extracellular vesicles (EVs) in the modulation of tumor progression. Methods and Results: In the present study, we found that EVs derived from 3T3-L1 adipocytes could affect PC3 and DU145 PCa cell traits, inducing increased proliferation, migration and invasion. Furthermore, conditioning of both PCa cell lines with adipocyte-released EVs resulted in lower sensitivity to docetaxel, with reduced phosphatidylserine externalization and decreased caspase 3 and PARP cleavage. In particular, these alterations were paralleled by an Akt/HIF-1α axis-related Warburg effect, characterized by enhanced glucose consumption, lactate release and ATP production. Conclusions: Collectively, these findings demonstrate that EV-mediated crosstalk exists between adipocytes and PCa, driving tumor aggressiveness.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; (M.A.); (P.L.)
- Correspondence:
| | - Martina Anselmi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; (M.A.); (P.L.)
| | - Emanuela Carollo
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (E.C.); (D.C.)
| | - Patrizia Sartori
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (P.S.); (P.P.)
| | - Patrizia Procacci
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (P.S.); (P.P.)
| | - David Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (E.C.); (D.C.)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; (M.A.); (P.L.)
| |
Collapse
|
24
|
Prasad S, Saha P, Chatterjee B, Chaudhary AA, Lall R, Srivastava AK. Complexity of Tumor Microenvironment: Therapeutic Role of Curcumin and Its Metabolites. Nutr Cancer 2022; 75:1-13. [PMID: 35818029 DOI: 10.1080/01635581.2022.2096909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment (TME) is a complex network of cellular and non-cellular components surrounding the tumor. The cellular component includes fibroblasts, adipocytes, endothelial cells, and immune cells, while non-cellular components are tumor vasculature, extracellular matrix and signaling molecules. The tumor cells have constant close interaction with their surrounding TME components that facilitate their growth, survival, and metastasis. Targeting a complex TME network and its interaction with the tumor can offer a novel strategy to disrupt cancer cell progression. Curcumin, from turmeric rhizome, is recognized as a safe and effective natural therapeutic agent against multiple diseases including cancer. Here the effects of curcumin and its metabolites on tumor-TME interaction modulating ability have been described. Curcumin and its metabolites regulate TME by inhibiting the growth of its cellular components such as cancer-associated adipocytes, cancer-associated fibroblast, tumor endothelial cells, tumor-stimulating immune cells, and inducing anticancer immune cells. They also inhibit the interplay of tumor cells to TME by suppressing non-cellular components such as extracellular matrix, and associated tumor promoting signaling-pathways. In addition, curcumin inhibits the inflammatory environment, suppresses angiogenic factors, and increases antioxidant status in TME. Overall, curcumin has the capability to regulate TME components and their interaction with tumor cells.
Collapse
Affiliation(s)
| | - Priyanka Saha
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Bilash Chatterjee
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh, Saudi Arabia
| | - Rajiv Lall
- Noble Pharma, LLC, Menomonie, Wisconsin, USA
| | - Amit K Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
25
|
Thresholds of visceral fat area and percent of body fat to define sarcopenic obesity and its clinical consequences in Chinese cancer patients. Clin Nutr 2022; 41:737-745. [DOI: 10.1016/j.clnu.2022.01.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/08/2022] [Accepted: 01/29/2022] [Indexed: 11/19/2022]
|
26
|
Zhang Y, Qin P, Xu X, Li M, Huang H, Yan J, Zhou Y. Mediator Complex Subunit 19 Promotes the Development of Hepatocellular Carcinoma by Regulating the AKT/mTOR Signaling Pathway. Front Oncol 2022; 11:792285. [PMID: 35047403 PMCID: PMC8761619 DOI: 10.3389/fonc.2021.792285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/10/2021] [Indexed: 01/13/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common malignant tumors, the pathogenesis of which remains unclear. Mediator complex subunit 19 (MED19), a subunit of the Mediator complex, is a multi-protein co-activator necessary for DNA transcription factors to induce RNA polymerase II transcription. In the current study, we aimed to study the role of MED19 in HCC and elucidate its mechanism. Methods MED19 expression in HCC tissues was determined. The relationship between MED19 and the clinical prognosis was explored. The influence of MED19 on HCC cell viability, migration, invasion, and apoptosis was studied. The expression of AKT/mTOR pathway genes and proteins was detected by qRT-PCR and western blot. The correlation between MED19 and immune infiltration was investigated. Results MED19 was upregulated in HCC tissues compared with tumor-adjacent tissues, and was associated with a poor prognosis. Furthermore, high MED19 expression was correlated with race, gender, etc. Knockdown of MED19 inhibited cell proliferation, migration, invasion, and promoted apoptosis. Knockdown of MED19 decreased p-AKT and p-mTOR protein expression. Additionally, the downstream effectors of the AKT/mTOR pathway, p70S6K1 and 4EBP1, were affected by MED19. Notably, MED19 expression was positively correlated with the infiltration levels of B cells, CD4+ T cells, CD8+ T cells, macrophages, etc. Conclusion MED19 is significantly upregulated in HCC tissues and cells. MED19 may promote the progression of HCC in vitro and may be related to immune infiltration. Together, our data show that MED19 could be considered as a new possible biomarker as well as a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Microbiology, Guilin Medical University, Guilin, China.,Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Peifang Qin
- Department of Microbiology, Guilin Medical University, Guilin, China.,Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Xingfeng Xu
- Department of Physiology, Guilin Medical University, Guilin, China
| | - Mao Li
- Department of Physiology, Guilin Medical University, Guilin, China
| | - Haitao Huang
- Department of Microbiology, Guilin Medical University, Guilin, China.,Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Jianguo Yan
- Department of Physiology, Guilin Medical University, Guilin, China
| | - Yali Zhou
- Department of Microbiology, Guilin Medical University, Guilin, China.,Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| |
Collapse
|
27
|
Brena D, Huang MB, Bond V. Extracellular vesicle-mediated transport: Reprogramming a tumor microenvironment conducive with breast cancer progression and metastasis. Transl Oncol 2021; 15:101286. [PMID: 34839106 PMCID: PMC8636863 DOI: 10.1016/j.tranon.2021.101286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles’ (EVs) role in breast tumor microenvironment and pre-metastatic niche development. Breast cancer EV-mediated transmission of pro-metastatic and drug-resistant phenotypes. Precision medicine with EVs as biomarkers and delivery vehicles for drug and anticancer genetic material.
Breast cancer metastatic progression to critical secondary sites is the second leading cause of cancer-related mortality in women. While existing therapies are highly effective in combating primary tumors, metastatic disease is generally deemed incurable with a median survival of only 2, 3 years. Extensive efforts have focused on identifying metastatic contributory targets for therapeutic antagonism and prevention to improve patient survivability. Excessive breast cancer release of extracellular vesicles (EVs), whose contents stimulate a metastatic phenotype, represents a promising target. Complex breast cancer intercellular communication networks are based on EV transport and transference of molecular information is in bulk resulting in complete reprogramming events within recipient cells. Other breast cancer cells can acquire aggressive phenotypes, endothelial cells can be induced to undergo tubule formation, and immune cells can be neutralized. Recent advancements continue to implicate the critical role EVs play in cultivating a tumor microenvironment tailored to cancer proliferation, metastasis, immune evasion, and conference of drug resistance. This literature review serves to frame the role of EV transport in breast cancer progression and metastasis. The following five sections will be addressed: (1) Intercellular communication in developing a tumor microenvironment & pre-metastatic niche. (2) Induction of the epithelial-to-mesenchymal transition (EMT). (3). Immune suppression & evasion. (4) Transmission of drug resistance mechanisms. (5) Precision medicine: clinical applications of EVs.
Collapse
Affiliation(s)
- Dara Brena
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Ming-Bo Huang
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| | - Vincent Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| |
Collapse
|
28
|
La Civita E, Liotti A, Cennamo M, Crocetto F, Ferro M, Liguoro P, Cimmino A, Imbimbo C, Beguinot F, Formisano P, Terracciano D. Peri-Prostatic Adipocyte-Released TGFβ Enhances Prostate Cancer Cell Motility by Upregulation of Connective Tissue Growth Factor. Biomedicines 2021; 9:biomedicines9111692. [PMID: 34829922 PMCID: PMC8615771 DOI: 10.3390/biomedicines9111692] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022] Open
Abstract
Periprostatic adipose tissue (PPAT) has emerged as a key player in the prostate cancer (PCa) microenvironment. In this study, we evaluated the ability of PPAT to promote PCa cell migration, as well as the molecular mechanisms involved. METHODS We collected conditioned mediums from in vitro differentiated adipocytes isolated from PPAT taken from PCa patients during radical prostatectomy. Migration was studied by scratch assay. RESULTS Culture with CM of human PPAT (AdipoCM) promotes migration in two different human androgen-independent (AI) PCa cell lines (DU145 and PC3) and upregulated the expression of CTGF. SB431542, a well-known TGFβ receptor inhibitor, counteracts the increased migration observed in presence of AdipoCM and decreased CTGF expression, suggesting that a paracrine secretion of TGFβ by PPAT affects motility of PCa cells. CONCLUSIONS Collectively, our study showed that factors secreted by PPAT enhanced migration through CTGF upregulation in AI PCa cell lines. These findings reveal the potential of novel therapeutic strategies targeting adipocyte-released factors and TGFβ/CTGF axis to fight advanced PCa dissemination.
Collapse
Affiliation(s)
- Evelina La Civita
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
| | - Antonietta Liotti
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
| | - Michele Cennamo
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
| | - Felice Crocetto
- Department of Neurosciences, Sciences of Reproduction and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (C.I.)
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, 20141 Milan, Italy;
| | - Pasquale Liguoro
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
| | - Amelia Cimmino
- Institute of Genetics and Biophysic, National Research Council, 80131 Naples, Italy;
| | - Ciro Imbimbo
- Department of Neurosciences, Sciences of Reproduction and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (C.I.)
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
- Correspondence: (P.F.); (D.T.)
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (A.L.); (M.C.); (P.L.); (F.B.)
- Correspondence: (P.F.); (D.T.)
| |
Collapse
|
29
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kamperi N, Kanara I, Kodukula K, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Steliou K, Tamvakopoulos C, Vavvas DG, Zamboni RJ, Sampani K. Pathogenic mitochondrial dysfunction and metabolic abnormalities. Biochem Pharmacol 2021; 193:114809. [PMID: 34673016 DOI: 10.1016/j.bcp.2021.114809] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Abstract
Herein we trace links between biochemical pathways, pathogenesis, and metabolic diseases to set the stage for new therapeutic advances. Cellular and acellular microorganisms including bacteria and viruses are primary pathogenic drivers that cause disease. Missing from this statement are subcellular compartments, importantly mitochondria, which can be pathogenic by themselves, also serving as key metabolic disease intermediaries. The breakdown of food molecules provides chemical energy to power cellular processes, with mitochondria as powerhouses and ATP as the principal energy carrying molecule. Most animal cell ATP is produced by mitochondrial synthase; its central role in metabolism has been known for >80 years. Metabolic disorders involving many organ systems are prevalent in all age groups. Progressive pathogenic mitochondrial dysfunction is a hallmark of genetic mitochondrial diseases, the most common phenotypic expression of inherited metabolic disorders. Confluent genetic, metabolic, and mitochondrial axes surface in diabetes, heart failure, neurodegenerative disease, and even in the ongoing coronavirus pandemic.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.
| | - Douglas V Faller
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Ioannis P Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, NY, USA
| | - David N Harpp
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Natalia Kamperi
- Center for Clinical, Experimental Surgery and Translational Research Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | | | - Anastasios N Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Carl A Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Whitney R Powers
- Department of Health Sciences, Boston University, Boston, MA, USA; Department of Anatomy, Boston University School of Medicine, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA; PhenoMatriX, Inc., Natick, MA, USA
| | - Constantin Tamvakopoulos
- Center for Clinical, Experimental Surgery and Translational Research Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Demetrios G Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Robert J Zamboni
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Konstantina Sampani
- Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|