1
|
Wei G, Shen FJ, Liu JL, Zhao JH, Yang FY, Feng RQ, Lu J, Zhang CY, Wang FW, Chen BD, Ding X, Yang JK. Uncoupling protein 1 deficiency leads to transcriptomic differences in livers of pregnancy female mice and aggravates hepatic steatosis. Arch Biochem Biophys 2025; 768:110395. [PMID: 40122441 DOI: 10.1016/j.abb.2025.110395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Pregnancy requires the coordination of metabolically active organs to support maternal nutrition and fetal growth. However, the metabolic cross-talk between adipose tissue and liver in females during pregnancy is still less clear. In this study, we evaluated the metabolic adaptations and phenotypes of liver in response to pregnancy-associated metabolic stress, particularly in the context of genetic ablation of Uncoupling protein 1 (Ucp1)-mediated catabolic circuit. Our results revealed that Ucp1 deficiency (UCP1 knockout, KO) mice during late pregnancy exhibited significantly deteriorated metabolic phenotypes, including hepatic steatosis and whole-body glucose and lipid homeostasis, as compared to Ucp1 deficiency or normal pregnancy mice. However, non-pregnant Ucp1 deficiency mice displayed nearly normal metabolic phenotypes and structure alterations similar to those of littermate controls. Moreover, transcriptomic analyses by RNA sequencing (RNA-seq) clearly revealed that Ucp1 deficiency led to a significant liver metabolic remodeling of differentially express genes (DEGs) before and especially during pregnancy. Consistently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses demonstrated the potential altered functions and signaling pathways, including metabolic dysfunctions in ribosome, oxidative phosphorylation, etc. Importantly, as derived from trend analyses of DEGs, our results further revealed the distinct expression pattern of each subcluster, which coincided with potential biological functions and relevant signaling pathways. The findings in the present study might provide valuable insights into the molecular mechanism of metabolic dysfunction-associated fatty liver disease (MAFLD) during pregnancy. Additionally, our data may provide a novel animal model of MAFLD, thus facilitating its potential therapies. NEW & NOTEWORTHY: Genetic ablation of Ucp1 during pregnancy increases hepatic steatosis and deteriorated whole-body glucose and lipid homeostasis. Moreover, changes in hepatic gene expression are closely associated with metabolic dysfunctions in ribosome and oxidative phosphorylation. This work highlights the therapeutic potential of targeting UCP1- mediated catabolic circuit between adipose and liver during pregnancy, and the utility of RNA-seq analysis to reveal valuable information for the distinct expression pattern of each subcluster that contribute to pregnancy-dependent MASLD progression.
Collapse
Affiliation(s)
- Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| | - Feng-Jie Shen
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jun-Li Liu
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Jian-Hua Zhao
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Fang-Yuan Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Ruo-Qi Feng
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Chen-Yang Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Feng-Wei Wang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Bei-Dong Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100005, China.
| | - Xin Ding
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100020, China.
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
2
|
Attané C, Muller C. From Fat Providers to Cancer Therapy: Adipocytes as Unexpected Allies. Cancer Res 2025; 85:1750-1752. [PMID: 40202825 DOI: 10.1158/0008-5472.can-25-1511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
Adipocytes from white adipose tissue support cancer progression by supplying fatty acids (FA) to tumor cells, whereas cold-activated brown adipose tissue has been shown to inhibit tumor growth by disrupting cancer cell metabolism. In a groundbreaking study published in Nature Biotechnology, Nguyen and colleagues developed adipose manipulation transplantation (AMT), a strategy that genetically reprograms white adipocytes to outcompete tumors for key nutrients. Using CRISPR activation technology, researchers enhanced adipocyte glucose and FA consumption by inducing a stable browning phenotype. In vitro, browned adipocytes reduced glycolysis and FA oxidation in cancer cells, inhibiting their proliferation. Implantation of engineered adipose organoids adjacent to tumors suppressed tumor growth, reduced angiogenesis, and altered metabolic gene expression in xenograft models. AMT also prevented tumor development in genetic mouse models of cancer, suggesting a role in cancer prevention. Finally, modified human mammary adipocytes inhibited the growth of patient-derived breast cancer organoids. This therapy, based on autologous fat transplantation, could offer a reversible and patient-specific approach. Challenges remain, including metabolic plasticity in cancer cells and the fragility of mature adipocytes in cell culture. AMT represents a paradigm shift in cancer therapy, leveraging adipocytes as metabolic competitors rather than tumor facilitators, opening new avenues for metabolism-targeted cancer treatments.
Collapse
Affiliation(s)
- Camille Attané
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, Université de Toulouse, Université Toulouse III - Paul Sabatier (UPS), Équipe Labélisée Ligue Nationale contre le Cancer, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, Université de Toulouse, Université Toulouse III - Paul Sabatier (UPS), Équipe Labélisée Ligue Nationale contre le Cancer, Toulouse, France
| |
Collapse
|
3
|
Park JS, Choi SB, Kim J, Jang WS, Ham WS. Impact of β 3-adrenergic receptor agonist on kidney cancer risk in patients with overactive bladder. BJU Int 2025. [PMID: 40344609 DOI: 10.1111/bju.16771] [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] [Indexed: 05/11/2025]
Abstract
OBJECTIVES To determine the effect of β3-adrenergic receptor (AR) agonist on the risk of kidney cancer in patients with overactive bladder (OAB). PATIENTS AND METHODS A nationwide population cohort study was conducted using data from the Korean National Health Insurance System database between January 2016 and December 2023. Validation analyses were performed using clinical data from patients with OAB treated with mirabegron or anticholinergics at a tertiary referral hospital between January 2014 and December 2023. The main exposure was intake of β3-AR agonist or anticholinergics, and the main outcome was incidence of kidney cancer. RESULTS Of the 1 419 148 patients (61.6% male; median [interquartile range] age, 64 [53-73] years), 3229 developed kidney cancer after OAB treatment. The incidence rate of kidney cancer was 0.7 per 1000 person-years in the mirabegron group and 0.5 per 1000 person-years in the anticholinergic group. Among the validation data of 3108 patients (49.3% male; mean [standard deviation] age, 63.9 [13.3] years), 45 (1.4%) developed kidney cancer after OAB treatment. The mirabegron group had a higher incidence of kidney cancer (1.8%) than the anticholinergic group (0.7%) (P = 0.025). CONCLUSIONS Use of β3-AR agonists was associated with an increased risk of kidney cancer compared with anticholinergics. While these findings suggest a potential association between mirabegron use and kidney cancer, further studies are needed to confirm causality. Clinicians should exercise caution when prescribing mirabegron in patients with risk factors for kidney cancer.
Collapse
Affiliation(s)
- Jee Soo Park
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Urology, Sorokdo National Hospital, Goheung, Korea
| | - Soo Beom Choi
- Division of Urban Society Research, Seoul Institute, Seoul, Korea
| | - Jongchan Kim
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Urology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
| | - Won Sik Jang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Leng X, Zhou C, Wu J, Zheng H, Wang J, Li Q, Huang Y, Liu J. The relationship between renal cell carcinoma pathological types and perirenal fat area. BMC Cancer 2025; 25:841. [PMID: 40340924 PMCID: PMC12060561 DOI: 10.1186/s12885-025-14164-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
INTRODUCTION To explore whether there is a relationship between perirenal fat area (PFA) and the pathological types of renal cell carcinoma (RCC). METHODS Two hundred ninety-seven cases of RCC patients were included in our study, which is a retrospective analysis. Based on pathological type, we divided the 297 RCC patients into two groups: the clear cell renal cell carcinoma (ccRCC) group (236 cases) and the non-clear cell renal cell carcinoma (non-ccRCC) group (61 cases). Computed tomography (CT) images at the renal vein level were used to measure PFA. A multivariate logistic regression model was employed to examine the connection between various pathological types of RCC and PFA. RESULTS Significant differences were observed between ccRCC and non-ccRCC patients in PFA (P = 0.007), contralateral PFA (P = 0.011), weight (P = 0.002), BMI (P < 0.001), pathological stage 1 (P = 0.010), and pathological stage 2 (P = 0.002). To study the link between pathological subtypes and PFA, a multivariate logistic regression model was employed. Stratifying patients by tumor location in the kidney, the multivariate logistic regression analysis showed that when the tumor is located outside the polar lines of the kidney (OPLK), for every 1 cm2 increase in PFA, the probability of developing ccRCC increases by 5% [1.05 (1.01, 1.10) P = 0.0153]. Furthermore, after stratifying patients by tumor location and pathological stage, it was found that in T1 stage patients with tumors located OPLK, for every 1 cm2 increase in PFA, the probability of developing ccRCC increases by 6% [1.06 (1.01, 1.11) P = 0.0300]. CONCLUSION When the tumor is located OPLK in T1 stage patients, PFA is positively correlated with ccRCC. Perirenal adipose tissue may be a risk factor for ccRCC.
Collapse
Affiliation(s)
- Xin Leng
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jiulong Wu
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Hongfang Zheng
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Jianliang Wang
- Department of Radiology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Qiaoxing Li
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Jianhu Liu
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China.
| |
Collapse
|
5
|
Zhou X, Li R, Lai M, Lai C. Exploring molecular and cellular mechanisms of Pre-Metastatic niche in renal cell carcinoma. Mol Cancer 2025; 24:121. [PMID: 40264130 PMCID: PMC12012986 DOI: 10.1186/s12943-025-02315-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Renal cell carcinoma (RCC) is among the most frequently occurring types of cancer, and its metastasis is a major contributor to its elevated mortality. Before the primary tumor metastasizes to secondary or distant organs, it remodels the microenvironment of these sites, creating a pre-metastatic niche (PMN) conducive to the colonization and growth of metastatic tumors. RCC releases a variety of biomolecules that induce angiogenesis, alter vascular permeability, modulate immune cells to create an immunosuppressive microenvironment, affect extracellular matrix remodeling and metabolic reprogramming, and determine the organotropism of metastasis through different signaling pathways. This review summarizes the principal processes and mechanisms underlying the formation of the premetastatic niche in RCC. Additionally, we emphasize the significance and potential of targeting PMNs for the prevention and treatment of tumor metastasis in future therapeutic approaches. Finally, we summarized the currently potential targeted strategies for detecting and treating PMN in RCC and provide a roadmap for further in-depth studies on PMN in RCC.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ruirui Li
- Institute of Immunology, Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Maode Lai
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Chong Lai
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
6
|
Zhang L, Li Q, Wu M, Feng X, Dai W, Chen P, Chen D, Zheng Z, Lin X, Wei G. TRIM22 governs tumorigenesis and protects against endometrial cancer-associated cachexia by inhibiting inflammatory response and adipose thermogenic activity. Cancer Metab 2025; 13:17. [PMID: 40200303 PMCID: PMC11980105 DOI: 10.1186/s40170-025-00386-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Endometrial cancer (EC) is one of the most common cancers in women, with a short overall survival and poor prognosis. Besides the biologically aggressive EC properties, Cancer-associated cachexia is the main factor. However, the detailed mechanism underlying EC-related cachexia and its harmful effects on EC progression and patient prognosis remains unclear. METHODS For clinical specimen and the vitro experiment, we detected TRIM22 expression level, EC patients' survival time, EC cell functional change, and adipose thermogenic changes to identify the function of TRIM22 in EC progression, EC-associated cachexia, and their molecular mechanisms. Then, for the vivo experiment, we exploited the xenografts in mice to identify the function of TRIM22 again, and to screen the drug therapeutic schedule. RESULTS Herein, we demonstrated that TRIM22 inhibited EC cell growth, invasion, and migration. Interleukin (IL)-6 mediated brown adipose tissue activation and white adipose tissue browning which induced EC-related cachexia. TRIM22 suppressed the EC cells' secretion of IL-6, and IL-6 mediated EC-related cachexia. Mechanistically, TRIM22 inhibited EC progression by suppressing the nucleotide-binding oligomerization domain 2(NOD2)/nuclear factor-kappaB (NF-κB) signaling pathway, with the purpose of impeding the production of IL-6. Moreover, we revealed that TRIM22 inhibited EC-associated cachexia by suppressing the IL-6/IL-6 receptor (IL-6R) signaling pathway. Therapeutically, we demonstrated that combination treatment with a TRIM22 inducer (progesterone) and a thermogenic inhibitor (IL-6R antibody) synergistically augmented the antitumor efficacy of carbotaxol (carboplatin and paclitaxel), in vivo. CONCLUSION Our data reveals that TRIM22-EC-IL-6-cachexia cross-communication has important clinical relevance and that the use of combined therapy holds great promise for enhancing the efficacy of anti-ECs. (Fig. graphical abstract).
Collapse
Affiliation(s)
- Liping Zhang
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Quanrong Li
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Meiting Wu
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiushan Feng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Weichao Dai
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Peifang Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Dezhao Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Zhiqun Zheng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiaoyan Lin
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
7
|
Wei Z, Ye Y, Liu C, Wang Q, Zhang Y, Chen K, Cheng G, Zhang X. MIER2/PGC1A elicits sunitinib resistance via lipid metabolism in renal cell carcinoma. J Adv Res 2025; 70:287-305. [PMID: 38702028 PMCID: PMC11976417 DOI: 10.1016/j.jare.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is one of the most common malignant tumors of the urinary system and accounts for more than 90 % of all renal tumors. Resistance to targeted therapy has emerged as a pivotal factor that contributes to the progressive deterioration of patients with advanced RCC. Metabolic reprogramming is a hallmark of tumorigenesis and progression, with an increasing body of evidence indicating that abnormal lipid metabolism plays a crucial role in the advancement of renal clear cell carcinoma. OBJECTIVES Clarify the precise mechanisms underlying abnormal lipid metabolism and drug resistance. METHODS Bioinformatics screening and analyses were performed to identify hub gene. qRT-PCR, western blot, chromatin immunoprecipitation (ChIP) assays, and other biological methods were used to explore and verify related pathways. Various cell line models and animal models were used to perform biological functional experiments. RESULTS In this study, we identified Mesoderm induction early response 2 (MIER2) as a novel biomarker for RCC, demonstrating its role in promoting malignancy and sunitinib resistance by influencing lipid metabolism in RCC. Mechanistically, MIER2 facilitated P53 deacetylation by binding to HDAC1. Acetylation modification augmented the DNA-binding stability and transcriptional function of P53, while deacetylation of P53 hindered the transcriptional process of PGC1A, leading to intracellular lipid accumulation in RCC. Furthermore, Trichostatin A (TSA), an inhibitor of HDAC1, was found to impede the MIER2/HDAC1/P53/PGC1A pathway, offering potential benefits for patients with sunitinib-resistant renal cell cancer. CONCLUSION Our findings highlight MIER2 as a key player in anchoring HDAC1 and inhibiting PGC1A expression through the deacetylation of P53, thereby inducing lipid accumulation in RCC and promoting drug resistance. Lipid-rich RCC cells compensate for energy production and sustain their own growth in a glycolysis-independent manner, evading the cytotoxic effects of targeted drugs and ultimately culminating in the development of drug resistance.
Collapse
Affiliation(s)
- Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhong Ye
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunxuan Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Shenzhen Huazhong University of Science and Technology Research Institute, China.
| |
Collapse
|
8
|
Wei G, Shen FJ, Liu JL, Zhao JH, Xie RR, Lu J, Zhang CY, Wang Y, Shi TT, Yang FY, Chen SQ, Huang YJ, Yang JK. Resinacein S, a novel triterpenoid from functional mushroom Ganoderma resinaceum, curbs obesity by regulating thermogenesis and energy metabolism. J Food Sci 2025; 90:e70161. [PMID: 40243376 DOI: 10.1111/1750-3841.70161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/12/2025] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Ganoderma mushrooms are popularly used as dietary supplements to promote health around the world. However, their potential applications for the prevention and treatment of obesity needs to be further investigated. In this study, we isolated a novel triterpenoid from Ganoderma resinaceum, Resinacein S (Res S), and determined its absolute configuration. We reported that Res S treatment significantly inhibited the high-fat HF diet-induced body weight gain though increased thermogenesis and energy metabolism. Specifically, treatment with Res S promoted brown adipose tissue activation and browning of inguinal white adipose tissue, improving whole-body glucose and lipid homeostasis. Mechanistically, Res S treatment induced the expression of thermogenic genes and related protein, for example, uncoupling protein 1 and mitochondrial biogenesis in a cell-autonomous manner by activating the AMPK-PGC1α signaling pathway. These findings identify Res S as a potential therapeutic alternative for obesity in the setting of its increasingly high prevalence. HIGHLIGHTS: Resinacein S (Res S) exhibited potent anti-obesity effects in high-fat diet-fed mice; Res S treatment significantly promoted brown adipose tissue activation and browning of inguinal white adipose tissue; Res S treatment stimulated UCP1 expression and enhanced mitochondrial function; Res S induced adipocyte thermogenic activity through activating the AMPK-PGC1α axis.
Collapse
Affiliation(s)
- Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Feng-Jie Shen
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jun-Li Liu
- Henan Key Laboratory of Neural Regeneration, Henan International Joint Laboratory of Neurorestoratology for Senile Dementia, Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Jian-Hua Zhao
- Henan Key Laboratory of Neural Regeneration, Henan International Joint Laboratory of Neurorestoratology for Senile Dementia, Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Rong-Rong Xie
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chen-Yang Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yuan Wang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ting-Ting Shi
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Fang-Yuan Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shu-Qin Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yan-Jie Huang
- College of Life Science, Tarim University, Alar, China
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
La Civita E, Sirica R, Crocetto F, Ferro M, Lasorsa F, Lucarelli G, Imbimbo C, Formisano P, Beguinot F, Terracciano D. FABP4-mediated ERK phosphorylation promotes renal cancer cell migration. BMC Cancer 2025; 25:575. [PMID: 40159492 PMCID: PMC11956428 DOI: 10.1186/s12885-025-13989-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
Clear cell Carcinoma (ccRCC) is the most common and lethal subtype among renal cancers. In the present study we investigated the potential role of fatty acid-binding protein 4 (FABP4), also known as adipocyte FABP (A-FABP) or aP2 on ccRCC progression. Firstly, we found that FABP4 median serum levels were significantly higher in ccRCC patients compared to HD. Based on this result and to evaluate whether FABP4 plays a role on renal cancer malignant phenotype, we analyzed proliferation and migration in 786-O and ACHN cell lines using recombinant FABP4. We found that FABP4 significantly increased cell migration, whereas it had no significant effect on proliferation. As FABP4 is mainly expressed by adipocytes, we measured FABP4 adipocyte conditioned media (Ad-CM) levels showing that Ad-CM from ccRCC (Ad-CM ccRCC) had significantly higher mean values compared to Ad-CM obtained from Healthy Donors (HD). To assess the effects of adipocyte-released FABP-4, on cancer malignant phenotype we evaluated 786-O and ACHN proliferation and migration, using Ad-CM from ccRCC and Ad-CM from HD alone or in combination with FABP4 inhibitor BMS309403. Our results showed that Ad-CM enhanced cell proliferation in ACHN, but not in 786-O and on cell motility in both cell lines and this effect was partially reverted by BMS309403 in both cell lines. Moreover, in both cell lines, FABP4 effect was associated with an increased ERK phosphorylation. Collectively these data support the role of FABP4 in ccRCC progression and its potential use as noninvasive biomarker and therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Evelina La Civita
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Rosa Sirica
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Felice Crocetto
- Department of Neurosciences, Sciences of Reproduction and Odontostomatology, University of Naples "Federico II", Naples, 80131, Italy
| | - Matteo Ferro
- Unit of Urology, Department of Health Science, University of Milan, ASST Santi Paolo and Carlo, Milan, 20142, Italy
| | - Francesco Lasorsa
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, 70124, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, 70124, Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Sciences of Reproduction and Odontostomatology, University of Naples "Federico II", Naples, 80131, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy.
| |
Collapse
|
10
|
Grigoraș A, Amalinei C. The Role of Perirenal Adipose Tissue in Carcinogenesis-From Molecular Mechanism to Therapeutic Perspectives. Cancers (Basel) 2025; 17:1077. [PMID: 40227577 PMCID: PMC11987925 DOI: 10.3390/cancers17071077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Perirenal adipose tissue (PRAT) exhibits particular morphological features, with its activity being mainly related to thermogenesis. However, an expanded PRAT area seems to play a significant role in cardiovascular diseases, diabetes mellitus, and chronic kidney disease pathogenesis. Numerous studies have demonstrated that PRAT may support cancer progression and invasion, mainly in obese patients. The mechanism underlying these processes is of dysregulation of PRAT's secretion of adipokines and pro-inflammatory cytokines, such as leptin, adiponectin, chemerin, apelin, omentin-1, vistatin, nesfatin-1, and other pro-inflammatory cytokines, modulated by tumor cells. Cancer cells may also induce a metabolic reprogramming of perirenal adipocytes, leading to increased lipids and lactate transfer to the tumor microenvironment, contributing to cancer growth in a hypoxic milieu. In addition, the PRAT browning process has been specifically detected in renal cell carcinoma (RCC), being characterized by upregulated expression of brown/beige adipocytes markers (UCP1, PPAR-ɣ, c/EBPα, and PGC1α) and downregulated white fat cells markers, such as LEPTIN, SHOX2, HOXC8, and HOXC9. Considering its multifaceted role in cancer, modulation of PRAT's role in tumor progression may open new directions for oncologic therapy improvement. Considering the increasing evidence of the relationship between PRAT and tumor cells, our review aims to provide a comprehensive analysis of the perirenal adipocytes' impact on tumor progression and metastasis.
Collapse
Affiliation(s)
- Adriana Grigoraș
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Histopathology, Institute of Legal Medicine, 700455 Iasi, Romania
| | - Cornelia Amalinei
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Histopathology, Institute of Legal Medicine, 700455 Iasi, Romania
| |
Collapse
|
11
|
Wang N, Lu S, Cao Z, Li H, Xu J, Zhou Q, Yin H, Qian Q, Zhang X, Tao M, Jiang Q, Zhou P, Zheng L, Han L, Li H, Yin L, Gu Y, Dou X, Sun H, Wang W, Piao HL, Li F, Xu Y, Yang W, Chen S, Liu J. Pyruvate metabolism enzyme DLAT promotes tumorigenesis by suppressing leucine catabolism. Cell Metab 2025:S1550-4131(25)00066-X. [PMID: 40112809 DOI: 10.1016/j.cmet.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/24/2024] [Accepted: 02/22/2025] [Indexed: 03/22/2025]
Abstract
Pyruvate and branched-chain amino acid (BCAA) metabolism are pivotal pathways in tumor progression, yet the intricate interplay between them and its implications for tumor progression remain elusive. Our research reveals that dihydrolipoamide S-acetyltransferase (DLAT), a pyruvate metabolism enzyme, promotes leucine accumulation and sustains mammalian target of rapamycin (mTOR) complex activation in hepatocellular carcinoma (HCC). Mechanistically, DLAT directly acetylates the K109 residue of AU RNA-binding methylglutaconyl-coenzyme A (CoA) hydratase (AUH), a critical enzyme in leucine catabolism, inhibiting its activity and leading to leucine accumulation. Notably, DLAT upregulation correlates with poor prognosis in patients with HCC. Therefore, we developed an AUHK109R-mRNA lipid nanoparticles (LNPs) therapeutic strategy, which effectively inhibits tumor growth by restoring leucine catabolism and inhibiting mTOR activation in vivo. In summary, our findings uncover DLAT's unexpected role as an acetyltransferase for AUH, suppressing leucine catabolism. Restoring leucine catabolism with AUHK109R-mRNA LNP effectively inhibits HCC development, highlighting a novel direction for cancer research.
Collapse
Affiliation(s)
- Ning Wang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Sijia Lu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ziyi Cao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huimin Li
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junting Xu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qian Zhou
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hanrui Yin
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qiqi Qian
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xianjing Zhang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Mijia Tao
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Quanxin Jiang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peihui Zhou
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liaoyuan Zheng
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liu Han
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hongtao Li
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Limin Yin
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yunqing Gu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xuefeng Dou
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haipeng Sun
- Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Wei Wang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Fuming Li
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiwei Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Suzhen Chen
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Junli Liu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
12
|
Xu J, Cai Z, Pang Z, Chen J, Zhu K, Wang D, Tu J. Smilax glabra Flavonoids Inhibit AMPK Activation and Induce Ferroptosis in Obesity-Associated Colorectal Cancer. Int J Mol Sci 2025; 26:2476. [PMID: 40141120 PMCID: PMC11942472 DOI: 10.3390/ijms26062476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Smilax glabra flavonoids (SGF), the active components of Smilax glabra Roxb., have been demonstrated to exhibit antioxidant activity and metabolic benefits in obesity, leading us to further explore their antitumor effects in obesity-related colorectal cancer (CRC). This study investigated the antiproliferative effects of SGF on obesity-related CRC by using a murine colon adenocarcinoma MC38 cell line. The underlying mechanisms were further explored via RNA-Seq and bioinformatics analysis in combination with experimental validation. SGF was proven to possess cytotoxic effects against MC38 cells, indicated by the inhibition of proliferation and migration, especially in an adipocyte-rich environment. In line with this, SGF exhibited much stronger antiproliferative effects on MC38-transplanted tumors in obese mice. Transcriptomics analysis showed that the cytotoxic effects of SGF might be related to the AMPK pathway and ferroptosis. On this basis, SGF was confirmed to induce ferroptosis and dictate ferroptosis sensitivity in a high-fat context mimicked by a two-step conditioned medium (CM) transfer experiment or a Transwell coculture system. The results of Western blotting validated that SGF suppressed the phosphorylation of AMPK, accompanied by alterations in the biomarkers of ferroptosis. These results demonstrate that SGF exerts in vitro and in vivo antiproliferative effects in obesity-associated CRC through inhibiting AMPK activation, thereby driving ferroptosis.
Collapse
Affiliation(s)
- Jianqin Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
| | - Zhaowei Cai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyao Pang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiayan Chen
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Keyan Zhu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
| | - Dejun Wang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jue Tu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
| |
Collapse
|
13
|
Wang J, Du H, Xie W, Bi J, Zhang H, Liu X, Wang Y, Zhang S, Lei A, He C, Yuan H, Zhang J, Li Y, Xu P, Liu S, Zhou Y, Shen J, Wu J, Cai Y, Yang C, Li Z, Liang Y, Zhao Y, Zhang J, Song M. CAR-Macrophage Therapy Alleviates Myocardial Ischemia-Reperfusion Injury. Circ Res 2024; 135:1161-1174. [PMID: 39465245 DOI: 10.1161/circresaha.124.325212] [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: 07/10/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Given the growing acknowledgment of the detrimental effects of excessive myocardial fibrosis on pathological remodeling after myocardial ischemia-reperfusion injury (I/R), targeting the modulation of myocardial fibrosis may offer protective and therapeutic advantages. However, effective clinical interventions and therapies that target myocardial fibrosis remain limited. As a promising chimeric antigen receptor (CAR) cell therapy, whether CAR macrophages (CAR-Ms) can be used to treat I/R remains unclear. METHODS The expression of FAP (fibroblast activation protein) was studied in mouse hearts after I/R. FAP CAR-Ms were generated to target FAP-expressing cardiac fibroblasts in mouse hearts after I/R. The phagocytosis activity of FAP CAR-Ms was tested in vitro. The efficacy and safety of FAP CAR-Ms in treating I/R were evaluated in vivo. RESULTS FAP was significantly upregulated in activated cardiac fibroblasts as early as 3 days after I/R. Upon demonstrating their ability to engulf FAP-overexpressing fibroblasts, we intravenously administered FAP CAR-Ms to mice at 3 days after I/R and found that FAP CAR-Ms significantly improved cardiac function and reduced myocardial fibrosis in mice after I/R. No toxicities associated with FAP CAR-Ms were detected in the heart or other organs at 2 weeks after I/R. Finally, we found that FAP CAR-Ms conferred long-term cardioprotection against I/R. CONCLUSIONS Our proof-of-concept study demonstrates the therapeutic potential of FAP CAR-Ms in alleviating myocardial I/R and potentially opens new avenues for the treatment of a range of heart diseases that include a fibrotic phenotype.
Collapse
Affiliation(s)
- Jiawan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
| | - Heng Du
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
| | - Wanrun Xie
- Liangzhu Laboratory (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
- Center of Gene and Cell Therapy and Genome Medicine of Zhejiang Province, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
| | - Jinmiao Bi
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Hao Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Xu Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Yuhan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Shaolong Zhang
- Liangzhu Laboratory (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
- Center of Gene and Cell Therapy and Genome Medicine of Zhejiang Province, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
| | - Anhua Lei
- Liangzhu Laboratory (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
- Center of Gene and Cell Therapy and Genome Medicine of Zhejiang Province, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
| | - Chuting He
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Hailong Yuan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Jiahe Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Yujing Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Pengfei Xu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Siqi Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Yanan Zhou
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Jianghua Shen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Jingdong Wu
- Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China (J. Wu, Y.C., Y. Zhao)
| | - Yihong Cai
- Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China (J. Wu, Y.C., Y. Zhao)
| | - Chaofan Yang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Zeya Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Yingxin Liang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| | - Yang Zhao
- Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China (J. Wu, Y.C., Y. Zhao)
| | - Jin Zhang
- Beijing Chao-Yang Hospital, Department of Anesthesiology, Beijing, China (J. Wang)
- Liangzhu Laboratory (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital (W.X., S.Z., A.L., Jin Zhang), Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
- Center of Gene and Cell Therapy and Genome Medicine of Zhejiang Province, Hangzhou, China (W.X., S.Z., A.L., Jin Zhang)
| | - Moshi Song
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
- University of Chinese Academy of Sciences, Beijing, China (J. Wang, H.D., J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Y. Liang, M.S.)
- Beijing Institute for Stem Cell and Regenerative Medicine, China (J.B., H.Z., X.L., Y.W., C.H., H.Y., Jiahe Zhang, Y. Li, P.X., S.L., Y. Zhou, J.S., C.Y., Z.L., Y. Liang, M.S.)
| |
Collapse
|
14
|
Ma J, Li J, Chen X, Ma Y. Ojeok-san enhances platinum sensitivity in ovarian cancer by regulating adipocyte paracrine IGF1 secretion. Adipocyte 2024; 13:2282566. [PMID: 37993991 PMCID: PMC10761029 DOI: 10.1080/21623945.2023.2282566] [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: 06/20/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Platinum is a commonly used drug for ovarian cancer (OvCa) treatment, but drug resistance limits its clinical application. This study intended to delineate the effects of adipocytes on platinum resistance in OvCa. METHODS OvCa cells were maintained in the adipocyte-conditioned medium. Cell viability and apoptosis were detected by CCK-8 and flow cytometry, separately. Proliferation and apoptosis-related protein expression were assayed by western blot. The IC50 values of cisplatin and carboplatin were determined using CCK-8. IGF1 secretion and expression were assayed via ELISA and western blot, respectively. A xenograft model was established, and pathological changes were detected by H&E staining. Proliferation and apoptosis-associated protein expression was assessed via IHC. RESULTS Adipocytes promoted the viability and repressed cell apoptosis in OvCa, as well as enhancing platinum resistance, while the addition of IGF-1 R inhibitor reversed the effects of adipocytes on proliferation, apoptosis, and drug resistance of OvCa cells. Treatment with different concentrations of Ojeok-san (OJS) inhibited the adipocyte-induced platinum resistance in OvCa cells by suppressing IGF1. The combined treatment of OJS and cisplatin significantly inhibited tumour growth in vivo with good mouse tolerance. CONCLUSION In summary, OJS inhibited OvCa proliferation and platinum resistance by suppressing adipocyte paracrine IGF1 secretion.
Collapse
Affiliation(s)
- Jiong Ma
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Junyan Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Xuejun Chen
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yanyan Ma
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Zhang H, Zhao J, Yu J, Zhang X, Ran S, Wang S, Ye W, Luo Z, Li X, Hao Y, Zong J, Li R, Lai L, Zheng K, Huang P, Zhou C, Wu J, Li Y, Xia J. Lactate metabolism and lactylation in cardiovascular disease: novel mechanisms and therapeutic targets. Front Cardiovasc Med 2024; 11:1489438. [PMID: 39664763 PMCID: PMC11631895 DOI: 10.3389/fcvm.2024.1489438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Cardiovascular disease (CVD) is responsible for approximately 30% of annual global mortality rates, yet existing treatments for this condition are considered less than ideal. Despite being previously overlooked, lactate, a byproduct of glycolysis, is now acknowledged for its crucial role in the cellular functions of the cardiovascular system. Recent studies have shown that lactate influences the proliferation, differentiation, and activation of immune cells through its modulation of post-translational protein modifications, thereby affecting the development and prognosis of cardiovascular disease. Consequently, there has been a notable increase in interest towards drug targets targeting lactylation in immune cells, prompting further exploration. In light of the swift advancements in this domain, this review article is dedicated to examining lactylation in cardiovascular disease and potential drug targets for regulating lactylation, with the aim of enhancing comprehension of this intricate field.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Song Wang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ran Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kexiao Zheng
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Translational Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, ChineseAcademy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|
16
|
Liu Y, Ouyang Q, Li Q. A novel brown adipocytes-related gene signature predicts and validates prognosis and immune infiltration of clear cell renal cell carcinoma. Am J Cancer Res 2024; 14:4286-4305. [PMID: 39417181 PMCID: PMC11477832 DOI: 10.62347/viqm5219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common kidney cancer. The crosstalk between tumor tissue and adjacent adipose tissue has been appreciated recently. This study examines the predictive usefulness of brown adipocyte-related genes (BARGs) in ccRCC. METHODS The transcriptome and clinical data of ccRCC patients were obtained from TCGA-KIRC and USA-ccRCC cohorts (848 tumor samples; 72 normal samples). Lasso-Cox methods were used to construct the risk prognostic signature model. We used Kaplan-Meier survival analysis to evaluate the prognostic significance of the risk model with ROC curves ascertaining prediction accuracy. The differences in immune cell infiltrates and signature risk scores between different risk categories were analyzed. Finally, biological experiments were performed to explore the functions of candidate genes. RESULTS TCGA-KIRC patients were classified into two clusters that differed significantly regarding overall survival (OS) and tumor microenvironment. After screening BARGs candidates, a signature consisting of PPP1R1A, DPYSL3, and PTPRM was created to calculate risk score. Patients were assigned to the high or low-risk group, and the high-risk group had a significantly worse prognosis. Consistent trend was validated in external USA-ccRCC cohort. Meanwhile, the signature risk score affected immune cell infiltrates within the ccRCC microenvironment, positively correlated with the infiltration of CD4+ T cells, CD8+ T cells, CD56dim, CD56bright NK cells, MDSCs, and macrophage cells, while negatively correlated with neutrophil, iDCs, mast cells, and eosinophil. Finally, knockdown of PPP1R1A and DPYSL3 in renal cancer cells showed impairment in tumor proliferation ability of ccRCC in vitro and in vivo. Conversely, knockdown of PTPRM exhibited a promotive effect. CONCLUSION We developed a predictive BARGs-related risk signature for early diagnosis and classifying ccRCC patients, which offers potential targets for individualized treatment of ccRCC.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University87 Xiangya Road, Changsha 410008, Hunan, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics110 Xiangya Road, Changsha 410078, Hunan, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education110 Xiangya Road, Changsha 410078, Hunan, P. R. China
| | - Qianying Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University87 Xiangya Road, Changsha 410008, Hunan, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics110 Xiangya Road, Changsha 410078, Hunan, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education110 Xiangya Road, Changsha 410078, Hunan, P. R. China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University87 Xiangya Road, Changsha 410008, Hunan, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics110 Xiangya Road, Changsha 410078, Hunan, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education110 Xiangya Road, Changsha 410078, Hunan, P. R. China
| |
Collapse
|
17
|
Chen L, Liu L. Adipose thermogenic mechanisms by cold, exercise and intermittent fasting: Similarities, disparities and the application in treatment. Clin Nutr 2024; 43:2043-2056. [PMID: 39088961 DOI: 10.1016/j.clnu.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Given its nonnegligible role in metabolic homeostasis, adipose tissue has been the target for treating metabolic disorders such as obesity, diabetes and cardiovascular diseases. Besides its lipolytic function, adipose thermogenesis has gained increased interest due to the irreplaceable contribution to dissipating energy to restore equilibrium, and its therapeutic effects have been testified in various animal models. In this review, we will brief about the canonical cold-stimulated adipose thermogenic mechanisms, elucidate on the exercise- and intermittent fasting-induced adipose thermogenic mechanisms, with a focus on the similarities and disparities among these signaling pathways, in an effort to uncover the overlapped and specific targets that may yield potent therapeutic efficacy synergistically in improving metabolic health.
Collapse
Affiliation(s)
- Linshan Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China.
| |
Collapse
|
18
|
Li S, Zhou Z, Gao M, Liao Z, He K, Qu W, Li J, Kamel IR, Chu Q, Zhang Q, Li Z. Incremental value of automatically segmented perirenal adipose tissue for pathological grading of clear cell renal cell carcinoma: a multicenter cohort study. Int J Surg 2024; 110:4221-4230. [PMID: 38573065 PMCID: PMC11254242 DOI: 10.1097/js9.0000000000001358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVES Accurate preoperative prediction of the pathological grade of clear cell renal cell carcinoma (ccRCC) is crucial for optimal treatment planning and patient outcomes. This study aims to develop and validate a deep-learning (DL) algorithm to automatically segment renal tumours, kidneys, and perirenal adipose tissue (PRAT) from computed tomography (CT) images and extract radiomics features to predict the pathological grade of ccRCC. METHODS In this cross-ethnic retrospective study, a total of 614 patients were divided into a training set (383 patients from the local hospital), an internal validation set (88 patients from the local hospital), and an external validation set (143 patients from the public dataset). A two-dimensional TransUNet-based DL model combined with the train-while-annotation method was trained for automatic volumetric segmentation of renal tumours, kidneys, and visceral adipose tissue (VAT) on images from two groups of datasets. PRAT was extracted using a dilation algorithm by calculating voxels of VAT surrounding the kidneys. Radiomics features were subsequently extracted from three regions of interest of CT images, adopting multiple filtering strategies. The least absolute shrinkage and selection operator (LASSO) regression was used for feature selection, and the support vector machine (SVM) for developing the pathological grading model. Ensemble learning was used for imbalanced data classification. Performance evaluation included the Dice coefficient for segmentation and metrics such as accuracy and area under curve (AUC) for classification. The WHO/International Society of Urological Pathology (ISUP) grading models were finally interpreted and visualized using the SHapley Additive exPlanations (SHAP) method. RESULTS For automatic segmentation, the mean Dice coefficient achieved 0.836 for renal tumours and 0.967 for VAT on the internal validation dataset. For WHO/ISUP grading, a model built with features of PRAT achieved a moderate AUC of 0.711 (95% CI, 0.604-0.802) in the internal validation set, coupled with a sensitivity of 0.400 and a specificity of 0.781. While model built with combination features of the renal tumour, kidney, and PRAT showed an AUC of 0.814 (95% CI, 0.717-0.889) in the internal validation set, with a sensitivity of 0.800 and a specificity of 0.753, significantly higher than the model built with features solely from tumour lesion (0.760; 95% CI, 0.657-0.845), with a sensitivity of 0.533 and a specificity of 0.767. CONCLUSION Automated segmentation of kidneys and visceral adipose tissue (VAT) through TransUNet combined with a conventional image morphology processing algorithm offers a standardized approach to extract PRAT with high reproducibility. The radiomics features of PRAT and tumour lesions, along with machine learning, accurately predict the pathological grade of ccRCC and reveal the incremental significance of PRAT in this prediction.
Collapse
Affiliation(s)
- Shichao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Ziling Zhou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Mengmeng Gao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Zhouyan Liao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Kangwen He
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Weinuo Qu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Jiali Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Ihab R Kamel
- Department of Radiology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Qingpeng Zhang
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, and the Musketeers Foundation Institute of Data Science, University of Hong Kong, Hong Kong, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
19
|
Zhao J, Jin D, Huang M, Ji J, Xu X, Wang F, Zhou L, Bao B, Jiang F, Xu W, Lu X, Xiao M. Glycolysis in the tumor microenvironment: a driver of cancer progression and a promising therapeutic target. Front Cell Dev Biol 2024; 12:1416472. [PMID: 38933335 PMCID: PMC11199735 DOI: 10.3389/fcell.2024.1416472] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Even with sufficient oxygen, tumor cells use glycolysis to obtain the energy and macromolecules they require to multiply, once thought to be a characteristic of tumor cells known as the "Warburg effect". In fact, throughout the process of carcinogenesis, immune cells and stromal cells, two major cellular constituents of the tumor microenvironment (TME), also undergo thorough metabolic reprogramming, which is typified by increased glycolysis. In this review, we provide a full-scale review of the glycolytic remodeling of several types of TME cells and show how these TME cells behave in the acidic milieu created by glucose shortage and lactate accumulation as a result of increased tumor glycolysis. Notably, we provide an overview of putative targets and inhibitors of glycolysis along with the viability of using glycolysis inhibitors in combination with immunotherapy and chemotherapy. Understanding the glycolytic situations in diverse cells within the tumor immunological milieu will aid in the creation of subsequent treatment plans.
Collapse
Affiliation(s)
- Junpeng Zhao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Mengxiang Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xuebing Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fei Wang
- Department of Laboratory Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lirong Zhou
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Baijun Bao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Weisong Xu
- Department of Gastroenterology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaomin Lu
- Department of Oncology Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Laboratory Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
20
|
Zheng X, Huang Y, Yang M, Jin L, Zhang X, Zhang R, Wu Y, Yan C, Gao Y, Zeng M, Li F, Zhou X, Zhang N, Liu J, Zha B. Vitamin D is involved in the effects of the intestinal flora and its related metabolite TMAO on perirenal fat and kidneys in mice with DKD. Nutr Diabetes 2024; 14:42. [PMID: 38858392 PMCID: PMC11164932 DOI: 10.1038/s41387-024-00297-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 05/21/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Vitamin D was shown to directly exert a protective effect on diabetic kidney disease (DKD) in our previous study. However, whether it has an effect on perirenal adipose tissue (PRAT) or the intestinal flora and its metabolites (trimethylamine N-oxide, TMAO) is unclear. METHODS DKD mice were received different concentrations of 1,25-(OH)2D3 for 2 weeks. Serum TNF-α levels and TMAO levels were detected. 16S rRNA sequencing was used to analyze gut microbiota. qPCR was used to detect the expression of TLR4, NF-Κb, PGC1α, and UCP-1 in kidney and adipose tissue. Histological changes in kidney and perirenal adipose tissue were observed using HE, PAS, Masson and oil red staining. Immunofluorescence and immunohistochemistry were used to detect the expression of VDR, PGC1α, podocin, and UCP-1 in kidney and adipose tissue. Electron microscopy was used to observe the pathological changes in the kidney. VDR knockout mice were constructed to observe the changes in the gut and adipose tissue, and immunofluorescence and immunohistochemistry were used to detect the expression of UCP-1 and collagen IV in the kidney. RESULTS 1,25-(OH)2D3 could improve the dysbiosis of the intestinal flora of mice with DKD, increase the abundance of beneficial bacteria, decrease the abundance of harmful bacteria, reduce the pathological changes in the kidney, reduce fat infiltration, and downregulate the expression of TLR4 and NF-κB in kidneys. The serum TMAO concentration in mice with DKD was significantly higher than that of the control group, and was significantly positively correlated with the urine ACR. In addition, vitamin D stimulated the expression of the surface markers PGC1α, UCP-1 and VDR in the PRAT in DKD mice, and TMAO downregulated the expression of PRAT and renal VDR. CONCLUSIONS The protective effect of 1,25-(OH)2D3 in DKD mice may affect the intestinal flora and its related metabolite TMAO on perirenal fat and kidneys.
Collapse
Affiliation(s)
- Xiaodi Zheng
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Yuhong Huang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Mengxue Yang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
- Community Health Research Center, Fudan University, Shanghai, 200240, China.
| | - Lulu Jin
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Xuemeng Zhang
- Community Health Research Center, Fudan University, Shanghai, 200240, China
- Pujiang Community Health Service Center, Minhang District, Shanghai, 2011112, China
| | - Rui Zhang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Yueyue Wu
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Cuili Yan
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Yuan Gao
- Pujiang Community Health Service Center, Minhang District, Shanghai, 2011112, China
| | - Miao Zeng
- Department of Infectious Diseases, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Fei Li
- Department of Endocrinology, Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Xue Zhou
- Department of Endocrinology, Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Neng Zhang
- Department of Endocrinology, Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Jun Liu
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| | - Bingbing Zha
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Community Health Research Center, Fudan University, Shanghai, 200240, China
| |
Collapse
|
21
|
Sun Z, Xia Y, Zhang Y, Guo H, Song Y, Ma H. Nonlinear Associations of Visceral and Perirenal Fat with Short-Term Postoperative Complications in Clear Cell Renal Cell Carcinoma Patients. Ann Surg Oncol 2024; 31:2208-2211. [PMID: 38252263 DOI: 10.1245/s10434-024-14931-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024]
Affiliation(s)
- Zehua Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, Shandong, China
| | - Yuanhao Xia
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, Shandong, China
- Department of Radiology, Binzhou Medical University, Yantai, Shandong, China
| | - Yumei Zhang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, Shandong, China
| | - Hao Guo
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, Shandong, China.
| | - Yang Song
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China.
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, Shandong, China.
| |
Collapse
|
22
|
Lin L, Tang Y, Ning K, Li X, Hu X. Investigating the causal associations between metabolic biomarkers and the risk of kidney cancer. Commun Biol 2024; 7:398. [PMID: 38561482 PMCID: PMC10984917 DOI: 10.1038/s42003-024-06114-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Metabolic reprogramming plays an important role in kidney cancer. We aim to investigate the causal effect of 249 metabolic biomarkers on kidney cancer from population-based data. This study extracts data from previous genome wide association studies with large sample size. The primary endpoint is random-effect inverse variance weighted (IVW). After completing 249 times of two-sample Mendelian randomization analysis, those significant metabolites are included for further sensitivity analysis. According to a strict Bonferrion-corrected level (P < 2e-04), we only find two metabolites that are causally associated with renal cancer. They are lactate (OR:3.25, 95% CI: 1.84-5.76, P = 5.08e-05) and phospholipids to total lipids ratio in large LDL (low density lipoprotein) (OR: 0.63, 95% CI: 0.50-0.80, P = 1.39e-04). The results are stable through all the sensitivity analysis. The results emphasize the central role of lactate in kidney tumorigenesis and provide novel insights into possible mechanism how phospholipids could affect kidney tumorigenesis.
Collapse
Affiliation(s)
- Lede Lin
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaxiong Tang
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kang Ning
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiang Li
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xu Hu
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Imran M, Abida, Eltaib L, Siddique MI, Kamal M, Asdaq SMB, Singla N, Al-Hajeili M, Alhakami FA, AlQarni AF, Abdulkhaliq AA, Rabaan AA. Beyond the genome: MALAT1's role in advancing urologic cancer care. Pathol Res Pract 2024; 256:155226. [PMID: 38452585 DOI: 10.1016/j.prp.2024.155226] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
Urologic cancers (UCs), which include bladder, kidney, and prostate tumors, account for almost a quarter of all malignancies. Long non-coding RNAs (lncRNAs) are tissue-specific RNAs that influence cell growth, death, and division. LncRNAs are dysregulated in UCs, and their abnormal expression may allow them to be used in cancer detection, outlook, and therapy. With the identification of several novel lncRNAs and significant exploration of their functions in various illnesses, particularly cancer, the study of lncRNAs has evolved into a new obsession. MALAT1 is a flexible tumor regulator implicated in an array of biological activities and disorders, resulting in an important research issue. MALAT1 appears as a hotspot, having been linked to the dysregulation of cell communication, and is intimately linked to cancer genesis, advancement, and response to treatment. MALAT1 additionally operates as a competitive endogenous RNA, binding to microRNAs and resuming downstream mRNA transcription and operation. This regulatory system influences cell growth, apoptosis, motility, penetration, and cell cycle pausing. MALAT1's evaluation and prognosis significance are highlighted, with a thorough review of its manifestation levels in several UC situations and its association with clinicopathological markers. The investigation highlights MALAT1's adaptability as a possible treatment target, providing fresh ways for therapy in UCs as we integrate existing information The article not only gathers current knowledge on MALAT1's activities but also lays the groundwork for revolutionary advances in the treatment of UCs.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia.
| | - Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Lina Eltaib
- Department of Pharmaceutics, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Muhammad Irfan Siddique
- Department of Pharmaceutics, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Neelam Singla
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Marwan Al-Hajeili
- Department of Medicine, King Abdulaziz University, Jeddah 23624, Saudi Arabia
| | - Fatemah Abdulaziz Alhakami
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Saudi Arabia
| | - Ahmed Farhan AlQarni
- Histopathology Laboratory, Najran Armed Forces Hospital, Najran 66251, Saudi Arabia
| | - Altaf A Abdulkhaliq
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| |
Collapse
|
24
|
Wu Q, Liu Z, Li B, Liu YE, Wang P. Immunoregulation in cancer-associated cachexia. J Adv Res 2024; 58:45-62. [PMID: 37150253 PMCID: PMC10982873 DOI: 10.1016/j.jare.2023.04.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/31/2023] [Accepted: 04/26/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Cancer-associated cachexia is a multi-organ disorder associated with progressive weight loss due to a variable combination of anorexia, systemic inflammation and excessive energy wasting. Considering the importance of immunoregulation in cachexia, it still lacks a complete understanding of the immunological mechanisms in cachectic progression. AIM OF REVIEW Our aim here is to describe the complex immunoregulatory system in cachexia. We summarize the effects and translational potential of the immune system on the development of cancer-associated cachexia and we attempt to conclude with thoughts on precise and integrated therapeutic strategies under the complex immunological context of cachexia. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight the inflammatory factors and additional mediators that have been identified to modulate this syndrome. Second, we decipher the potential role of immune checkpoints in tissue wasting. Third, we discuss the multilayered insights in cachexia through the immunometabolic axis, immune-gut axis and immune-nerve axis.
Collapse
Affiliation(s)
- Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University.
| | - Zhou Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Yu-E Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University.
| |
Collapse
|
25
|
Gao D, Zhang C, Guo H, Xu H, Liu H, Wang Z, Xu B, Gang W. Low-dose polystyrene microplastics exposure impairs fertility in male mice with high-fat diet-induced obesity by affecting prostate function. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123567. [PMID: 38367694 DOI: 10.1016/j.envpol.2024.123567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
The harmful effects of microplastics (MPs) on male fertility are receiving more and more attention. However, the impact of low-dose MPs exposure on the reproductive function of male mice is still unclear. In this study, we exposed male mice to low-dose MPs (25-30 μg/kg body weight/day) or low-dose MPs combined with high-fat diet (HFD) feeding. Our results showed that low-dose MPs exposure or HFD feeding significantly reduced sperm quality and the number of offspring born, while low-dose MPs exposure combined with HFD feeding further enhanced the above effects. The combination of low-dose MPs exposure and HFD feeding resulted in a notable elevation of inflammatory level within the prostate of mice and induced apoptosis of prostate epithelium and a decrease in nutrients (zinc, citrate) in seminal plasma fluid. Our findings in this study could provide valuable clues for better understanding the influence of low-dose MPs exposure on the reproductive system under metabolic disorders and facilitate the development of the prevention of reproductive toxicity caused by MPs exposure.
Collapse
Affiliation(s)
- Dajun Gao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Caoxu Zhang
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Huaqi Guo
- Department of Respiratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Lu, Shanghai, 200011, China.
| | - Huan Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Hui Liu
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, China.
| | - Zhong Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Wei Gang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China; Department of Endocrinology and Metabolism, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
26
|
Ren J, Huang B, Li W, Wang Y, Pan X, Ma Q, Liu Y, Wang X, Liang C, Zhang Y, Wang S, Yang F, Li H, Ning H, Jiang Y, Qin C, Ran A, Xiao B. RNA-binding protein IGF2BP2 suppresses metastasis of clear cell renal cell carcinoma by enhancing CKB mRNA stability and expression. Transl Oncol 2024; 42:101904. [PMID: 38341962 PMCID: PMC10867445 DOI: 10.1016/j.tranon.2024.101904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/15/2024] [Accepted: 02/03/2024] [Indexed: 02/13/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney cancer, with a highly aggressive phenotype and poor prognosis. RNA binding proteins (RBPs) play crucial roles in post-transcriptional gene regulation and have been implicated in tumorigenesis. RBPs have the potential to become a new therapeutic target for ccRCC. In this study, we screened and validated that insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) as an RBP, was down-regulated in ccRCC tissues and cell lines. Functionally, we verified that IGF2BP2 significantly suppressed the migration and invasion ability of ccRCC in vitro and in vivo. Mechanistically, RIP-seq and actinomycin D experiments results showed that IGF2BP2 enhanced the expression of Creatine Kinase B (CKB) by binding to CKB mRNA and enhancing its mRNA stability. Thus, IGF2BP2 inhibited ccRCC metastasis through enhancing the expression of CKB. Taken together, these finding suggests that IGF2BP2 is a novel metastasis suppressor of ccRCC and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Junwu Ren
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Bo Huang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China
| | - Wei Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing 400030, PR China
| | - Yongquan Wang
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing 400038, PR China
| | - Xiaojuan Pan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Qiang Ma
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Yuying Liu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Xiaolin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Ce Liang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Yuying Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Shimin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Feifei Yang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Haiping Li
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Ning
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Changhong Qin
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Ai Ran
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Bin Xiao
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
27
|
Wang P, Wang H, Xiao Y, Zou J, Chen H, Chen L, Wang F, Hu Y, Liu Y. Insights into metabolic characteristics and biological activity changes in Zangju ( Citrus reticulata cv. Manau Gan) peel at different maturity stages through UPLC-MS/MS-based metabolomics. Food Chem X 2024; 21:101197. [PMID: 38357370 PMCID: PMC10865237 DOI: 10.1016/j.fochx.2024.101197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/25/2024] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
In this study, comprehensive and systematic nontargeted metabolomics analysis was performed with the metabolites of Zangju peel (Citrus reticulata cv. Manau Gan, CRZP, which has been cultivated for over 400 years in Derong County, China.) at four different mature stages. A total of 1878 metabolites were identified, among which flavonoids were the most abundant (62.04 %), and identified 62 key differential metabolites significantly affected by maturity. Based on biological activity measurements, CRZP showed better antioxidant activity, lipase inhibition ability, inhibition of adipogenic differentiation in 3TT-L1 cells and promotion of lipid metabolism, with the biological activity of CRZP at different maturity stages being associated with key differential metabolite. Thus, CRZP is natural antioxidants and possess anti-obesity potential, and industrial production needs to consider the Maturity stage of its collection.
Collapse
Affiliation(s)
- Peng Wang
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Haifan Wang
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Yang Xiao
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jialiang Zou
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Hongping Chen
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Lin Chen
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Fu Wang
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Yuan Hu
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| | - Youping Liu
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| |
Collapse
|
28
|
Zhang H, Li Y, Ibáñez CF, Xie M. Perirenal adipose tissue contains a subpopulation of cold-inducible adipocytes derived from brown-to-white conversion. eLife 2024; 13:RP93151. [PMID: 38470102 PMCID: PMC10932542 DOI: 10.7554/elife.93151] [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] [Indexed: 03/13/2024] Open
Abstract
Perirenal adipose tissue (PRAT) is a unique visceral depot that contains a mixture of brown and white adipocytes. The origin and plasticity of such cellular heterogeneity remains unknown. Here, we combine single-nucleus RNA sequencing with genetic lineage tracing to reveal the existence of a distinct subpopulation of Ucp1-&Cidea+ adipocytes that arises from brown-to-white conversion during postnatal life in the periureter region of mouse PRAT. Cold exposure restores Ucp1 expression and a thermogenic phenotype in this subpopulation. These cells have a transcriptome that is distinct from subcutaneous beige adipocytes and may represent a unique type of cold-recruitable adipocytes. These results pave the way for studies of PRAT physiology and mechanisms controlling the plasticity of brown/white adipocyte phenotypes.
Collapse
Affiliation(s)
- Houyu Zhang
- Chinese Institute for Brain Research, Zhongguancun Life Science ParkBeijingChina
- Peking University Academy for Advanced Interdisciplinary StudiesBeijingChina
| | - Yan Li
- Chinese Institute for Brain Research, Zhongguancun Life Science ParkBeijingChina
- Peking University Academy for Advanced Interdisciplinary StudiesBeijingChina
| | - Carlos F Ibáñez
- Chinese Institute for Brain Research, Zhongguancun Life Science ParkBeijingChina
- Peking University School of Life Sciences, Peking-Tsinghua Center for Life SciencesBeijingChina
- PKU-IDG/McGovern Institute for Brain ResearchBeijingChina
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| | - Meng Xie
- PKU-IDG/McGovern Institute for Brain ResearchBeijingChina
- Peking University School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental HealthBeijingChina
- Department of Biosciences and Nutrition, Karolinska InstituteFlemingsbergSweden
| |
Collapse
|
29
|
Sun Z, Zhang Y, Xia Y, Ba X, Zheng Q, Liu J, Kuang X, Xie H, Gong P, Shi Y, Mao N, Wang Y, Liu M, Ran C, Wang C, Wang X, Li M, Zhang W, Fang Z, Liu W, Guo H, Ma H, Song Y. Association between CT-based adipose variables, preoperative blood biochemical indicators and pathological T stage of clear cell renal cell carcinoma. Heliyon 2024; 10:e24456. [PMID: 38268833 PMCID: PMC10803934 DOI: 10.1016/j.heliyon.2024.e24456] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/26/2024] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is corelated with tumor-associated material (TAM), coagulation system and adipocyte tissue, but the relationships between them have been inconsistent. Our study aimed to explore the cut-off intervals of variables that are non-linearly related to ccRCC pathological T stage for providing clues to understand these discrepancies, and to effectively preoperative risk stratification. Methods This retrospective analysis included 218 ccRCC patients with a clear pathological T stage between January 1st, 2014, and November 30th, 2021. The patients were categorized into two cohorts based on their pathological T stage: low T stage (T1 and T2) and high T stage (T3 and T4). Abdominal and perirenal fat variables were measured based on preoperative CT images. Blood biochemical indexes from the last time before surgery were also collected. The generalized sum model was used to identify cut-off intervals for nonlinear variables. Results In specific intervals, fibrinogen levels (FIB) (2.63-4.06 g/L) and platelet (PLT) counts (>200.34 × 109/L) were significantly positively correlated with T stage, while PLT counts (<200.34 × 109/L) were significantly negatively correlated with T stage. Additionally, tumor-associated material exhibited varying degrees of positive correlation with T stage at different cut-off intervals (cut-off value: 90.556 U/mL). Conclusion Preoperative PLT, FIB and TAM are nonlinearly related to pathological T stage. This study is the first to provide specific cut-off intervals for preoperative variables that are nonlinearly related to ccRCC T stage. These intervals can aid in the risk stratification of ccRCC patients before surgery, allowing for developing a more personalized treatment planning.
Collapse
Affiliation(s)
- Zehua Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Yumei Zhang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Yuanhao Xia
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
- Department of Radiology, Binzhou Medical University, Yantai, 264000, Shandong, China
| | - Xinru Ba
- Department of Radiology, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - Qingyin Zheng
- Department of Otolaryngology-Head & Neck Surgery, Case Western Reserve University, Cleveland, OH, 44106, United States
| | - Jing Liu
- Department of Pediatrics, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Xiaojing Kuang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, Shandong, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Peiyou Gong
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Yinghong Shi
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Yongtao Wang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Ming Liu
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Chao Ran
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Chenchen Wang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Xiaoni Wang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Min Li
- Department of Radiology, Yantai Traditional Chinese Medicine Hospital, Yantai, 264000, Shandong, China
| | - Wei Zhang
- Department of Radiology, Yantai Penglai People's Hospital, Yantai, 265600, Shandong, China
| | - Zishuo Fang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 610000, China
| | - Wanchen Liu
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Hao Guo
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Yang Song
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, China
| |
Collapse
|
30
|
Wang Q, Su Y, Sun R, Xiong X, Guo K, Wei M, Yang G, Ru Y, Zhang Z, Li J, Zhang J, Qiao Q, Li X. MIIP downregulation drives colorectal cancer progression through inducing peri-cancerous adipose tissue browning. Cell Biosci 2024; 14:12. [PMID: 38245780 PMCID: PMC10800076 DOI: 10.1186/s13578-023-01179-0] [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: 05/05/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND The enrichment of peri-cancerous adipose tissue is a distinctive feature of colorectal cancer (CRC), accelerating disease progression and worsening prognosis. The communication between tumor cells and adjacent adipocytes plays a crucial role in CRC advancement. However, the precise regulatory mechanisms are largely unknown. This study aims to explore the mechanism of migration and invasion inhibitory protein (MIIP) downregulation in the remodeling of tumor cell-adipocyte communication and its role in promoting CRC. RESULTS MIIP expression was found to be decreased in CRC tissues and closely associated with adjacent adipocyte browning. In an in vitro co-culture model, adipocytes treated with MIIP-downregulated tumor supernatant exhibited aggravated browning and lipolysis. This finding was further confirmed in subcutaneously allografted mice co-injected with adipocytes and MIIP-downregulated murine CRC cells. Mechanistically, MIIP interacted with the critical lipid mobilization factor AZGP1 and regulated AZGP1's glycosylation status by interfering with its association with STT3A. MIIP downregulation promoted N-glycosylation and over-secretion of AZGP1 in tumor cells. Subsequently, AZGP1 induced adipocyte browning and lipolysis through the cAMP-PKA pathway, releasing free fatty acids (FFAs) into the microenvironment. These FFAs served as the primary energy source, promoting CRC cell proliferation, invasion, and apoptosis resistance, accompanied by metabolic reprogramming. In a tumor-bearing mouse model, inhibition of β-adrenergic receptor or FFA uptake, combined with oxaliplatin, significantly improved therapeutic efficacy in CRC with abnormal MIIP expression. CONCLUSIONS Our data demonstrate that MIIP plays a regulatory role in the communication between CRC and neighboring adipose tissue by regulating AZGP1 N-glycosylation and secretion. MIIP reduction leads to AZGP1 oversecretion, resulting in adipose browning-induced CRC rapid progression and poor prognosis. Inhibition of β-adrenergic receptor or FFA uptake, combined with oxaliplatin, may represent a promising therapeutic strategy for CRC with aberrant MIIP expression.
Collapse
Affiliation(s)
- Qinhao Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Yuanyuan Su
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- Department of Pharmacology, Medical College, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Ruiqi Sun
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Xin Xiong
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Kai Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Mengying Wei
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Guodong Yang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yi Ru
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhengxiang Zhang
- Department of Pharmacology, Medical College, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Jing Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jing Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Xia Li
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
31
|
Xu W, Ye S, Liu W, Guo H, Zhang L, Wei S, Anwaier A, Chang K, Malafaia G, Zhang H, Ye D, Wei G. Single-cell RNA-seq analysis decodes the kidney microenvironment induced by polystyrene microplastics in mice receiving a high-fat diet. J Nanobiotechnology 2024; 22:13. [PMID: 38167034 PMCID: PMC10762848 DOI: 10.1186/s12951-023-02266-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
In recent years, the environmental health issue of microplastics has aroused an increasingly significant concern. Some studies suggested that exposure to polystyrene microplastics (PS-MPs) may lead to renal inflammation and oxidative stress in animals. However, little is known about the essential effects of PS-MPs with high-fat diet (HFD) on renal development and microenvironment. In this study, we provided the single-cell transcriptomic landscape of the kidney microenvironment induced by PS-MPs and HFD in mouse models by unbiased single-cell RNA sequencing (scRNA-seq). The kidney injury cell atlases in mice were evaluated after continued PS-MPs exposure, or HFD treated for 35 days. Results showed that PS-MPs plus HFD treatment aggravated the kidney injury and profibrotic microenvironment, reshaping mouse kidney cellular components. First, we found that PS-MPs plus HFD treatment acted on extracellular matrix organization of renal epithelial cells, specifically the proximal and distal convoluted tubule cells, to inhibit renal development and induce ROS-driven carcinogenesis. Second, PS-MPs plus HFD treatment induced activated PI3K-Akt, MAPK, and IL-17 signaling pathways in endothelial cells. Besides, PS-MPs plus HFD treatment markedly increased the proportions of CD8+ effector T cells and proliferating T cells. Notably, mononuclear phagocytes exhibited substantial remodeling and enriched in oxidative phosphorylation and chemical carcinogenesis pathways after PS-MPs plus HFD treatment, typified by alterations tissue-resident M2-like PF4+ macrophages. Multispectral immunofluorescence and immunohistochemistry identified PF4+ macrophages in clear cell renal cell carcinoma (ccRCC) and adjacent normal tissues, indicating that activate PF4+ macrophages might regulate the profibrotic and pro-tumorigenic microenvironment after renal injury. In conclusion, this study first systematically revealed molecular variation of renal cells and immune cells in mice kidney microenvironment induced by PS-MPs and HFD with the scRNA-seq approach, which provided a molecular basis for decoding the effects of PS-MPs on genitourinary injury and understanding their potential profibrotic and carcinogenesis in mammals.
Collapse
Affiliation(s)
- Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Shiqi Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Huaqi Guo
- Department of Pulmonary and Critical Care Medicine, The Ninth People's Hospital of Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Linhui Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Shiyin Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Kun Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Guilherme Malafaia
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute - Urutaí Campus, Rodovia Geraldo Silva Nascimento, 2,5 Km, Zona Rural, Urutaí, GO, Brazil.
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, People's Republic of China.
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Beijing Diabetes Institute, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
32
|
Chen W, Jiang J, Gao J, Wang G, Wang R, Lv J, Ben J. Roles and signaling pathways of CITED1 in tumors: overview and novel insights. J Int Med Res 2024; 52:3000605231220890. [PMID: 38190845 PMCID: PMC10775745 DOI: 10.1177/03000605231220890] [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: 08/07/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
CBP/p300 interacting transactivator with Glu/Asp-rich carboxy-terminal domain 1 (CITED1) is a transcriptional activator belonging to the non-DNA-binding transcription co-regulator family. It regulates diverse pathways, including the transforming growth factor/bone morphogenetic protein/SMAD, estrogen, Wnt-β-catenin, and androgen-AR signaling pathways, by binding to CBP/p300 co-activators through its conserved transactivation domain CR2. CITED1 plays an important role in embryonic development and a certain regulatory role in the occurrence and development of various tumors. In this article, the biological characteristics, expression regulation, participating signaling pathways, and potential roles of CITED1 in the clinical diagnosis and treatment of tumors are reviewed.
Collapse
Affiliation(s)
- Wenting Chen
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, China
| | - Jianing Jiang
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, China
| | - Jinqi Gao
- Department of Intervention, The Second Hospital Affiliated to Dalian Medical University, Dalian, China
| | - Gang Wang
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruoyu Wang
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, China
| | - Jinyan Lv
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jing Ben
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
33
|
Xie L, Hu W, Zhang H, Ding Y, Zeng Q, Liao X, Wang D, Xie W, Hui HX, Deng T. Single-nucleus RNA sequencing reveals heterogeneity among multiple white adipose tissue depots. LIFE METABOLISM 2023; 2:load045. [PMID: 39872854 PMCID: PMC11748973 DOI: 10.1093/lifemeta/load045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 01/30/2025]
Abstract
Regardless of its anatomical site, adipose tissue shares a common energy-storage role but exhibits distinctive properties. Exploring the cellular and molecular heterogeneity of white adipose tissue (WAT) is crucial for comprehending its function and properties. However, existing single-nucleus RNA sequencing (snRNA-seq) studies of adipose tissue heterogeneity have examined only one or two depots. In this study, we employed snRNA-seq to test five representative depots including inguinal, epididymal, mesenteric, perirenal, and pericardial adipose tissues in mice under physiological conditions. By analyzing the contents of main cell categories and gene profiles of various depots, we identified their distinctive physiological properties. Immune cells and fibro-adipogenic progenitor cells (FAPs) showed dramatic differences among WAT depots, while adipocytes seemed to be conserved. The heightened presence of regulatory macrophages and B cells in pericardial adipose tissues implied their potential contribution to the preservation of coronary vascular function. Moreover, the selective aggregation of pericytes within mesenteric adipose tissue was likely associated with the maintenance of intestinal barrier homeostasis. Using a combination of RNA sequencing and snRNA-seq analysis, the major subpopulations of FAPs derived from these depots determined the site characteristics of FAPs to a certain extent. Our work establishes a systematic and reliable foundation for investigating the heterogeneity of WAT depots and elucidating the unique roles these depots play in coordinating the function of adjacent organs.
Collapse
Affiliation(s)
- Limin Xie
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Wanyu Hu
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Haowei Zhang
- The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yujin Ding
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Qin Zeng
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiyan Liao
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Dandan Wang
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Wanqin Xie
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410028, China
| | - Hannah Xiaoyan Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Tuo Deng
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
34
|
Wang H, Zhang W, Ding Z, Ke H, Su D, Wang Q, Xu K. SEMA3G functions as a novel prognostic biomarker associated with Wnt pathway in clear cell renal cell carcinoma. Cell Signal 2023; 111:110868. [PMID: 37633476 DOI: 10.1016/j.cellsig.2023.110868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/13/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Renal cell cancer (RCC) is one of the most common cancer, and the incidence of clear cell renal cell cancer rank at the first among multiple subtypes of RCC. Tumor heterogeneity and limited therapies expedite researches and studies on prognostic biomarkers and molecular mechanism. SEMA3G mediates various bimolecular processes but few studies have assessed the influence of SEMA3G on ccRCC. The expression of SEMA3G at mRNA level in ccRCC was analyzed using 4 TCGA datasets. The expression at protein level was verified by immunohistochemistry and western blot. Biological pathway was explored by GSEA and western blot. At both mRNA and protein level, SEMA3G expressed significantly lower in ccRCC tissues compared with normal renal tissues, and the expression was highly associated with clinical stage and pathological grade. Low expression of SEMA3G indicated a poorer overall survival and disease specific survival. Transwell and wound-healing assays showed that overexpressed SEMA3G inhibited the cell motility of renal cancer cells. Upregulated SEMA3G suppressed the invasion and proliferation of both 769-P and 786-O cells. Wnt signaling pathway was tested to work in the interfering of SEMA3G on tumorigenesis and progression of ccRCC. The results provide novel insight into the role of SEMA3G in ccRCC, suggesting the prognostic value and potential suppressor role of SEMA3G.
Collapse
Affiliation(s)
- Huanrui Wang
- Department of Urology, Peking University People's Hospital, Beijing 100045, China
| | - Weiyu Zhang
- Department of Urology, Peking University People's Hospital, Beijing 100045, China
| | - Zehua Ding
- Department of Urology, Peking University People's Hospital, Beijing 100045, China
| | - Hanwei Ke
- Department of Urology, Peking University People's Hospital, Beijing 100045, China
| | - Dongyu Su
- Department of Urology, Peking University People's Hospital, Beijing 100045, China
| | - Qi Wang
- Department of Urology, Peking University People's Hospital, Beijing 100045, China
| | - Kexin Xu
- Department of Urology, Peking University People's Hospital, Beijing 100045, China.
| |
Collapse
|
35
|
Zhang Y, Sun Z, Ma H, Wang C, Zhang W, Liu J, Li M, Zhang Y, Guo H, Ba X. Prediction of Fuhrman nuclear grade for clear cell renal carcinoma by a multi-information fusion model that incorporates CT-based features of tumor and serum tumor associated material. J Cancer Res Clin Oncol 2023; 149:15855-15865. [PMID: 37672076 DOI: 10.1007/s00432-023-05353-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023]
Abstract
PURPOSE Prediction of Fuhrman nuclear grade is crucial for making informed herapeutic decisions in clear cell renal cell carcinoma (ccRCC). The current study aimed to develop a multi-information fusion model utilizing computed tomography (CT)-based features of tumors and preoperative biochemical parameters to predict the Fuhrman nuclear grade of ccRCC in a non-invasive manner. METHODS 218 ccRCC patients confirmed by histopathology were retrospectively analyzed. Univariate and multivariate logistic regression analyses were performed to identify independent predictors and establish a model for predicting the Fuhrman grade in ccRCC. The predictive performance of the model was evaluated using receiver operating characteristic (ROC) curves, calibration, the 10-fold cross-validation method, bootstrapping, the Hosmer-Lemeshow test, and decision curve analysis (DCA). RESULTS R.E.N.A.L. Nephrometry Score (RNS) and serum tumor associated material (TAM) were identified as independent predictors for Fuhrman grade of ccRCC through multivariate logistic regression. The areas under the ROC curve (AUC) for the multi-information fusion model composed of the above two factors was 0.810, higher than that of the RNS (AUC 0.694) or TAM (AUC 0.764) alone. The calibration curve and Hosmer-Lemeshow test showed the integrated model had a good fitting degree. The 10-fold cross-validation method (AUC 0.806) and bootstrap test (AUC 0.811) showed the good stability of the model. DCA demonstrated that the model had superior clinical utility. CONCLUSION A multi-information fusion model based on CT features of tumor and routine biochemical indicators, can predict the Fuhrman grade of ccRCC using a non-invasive approach. This model holds promise for assisting clinicians in devising personalized management strategies.
Collapse
Affiliation(s)
- Yumei Zhang
- Department of Radiology, Laishan Branch of Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Zehua Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Chenchen Wang
- Department of Radiology, Laishan Branch of Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Wei Zhang
- Department of Radiology, Yantai Penglai People's Hospital, Yantai, 265600, Shandong, China
| | - Jing Liu
- Department of Pathology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China
| | - Min Li
- Department of Radiology, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Yuxia Zhang
- Department of Obstetrics and Gynecology, Yanzhou Hospital of TCM, Yanzhou, 272100, Shandong, China
| | - Hao Guo
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, 264000, Shandong, China.
| | - Xinru Ba
- Department of Radiology, Yantaishan Hospital, Yantai, 264000, Shandong, China.
| |
Collapse
|
36
|
Tan S, Wang Z, Li N, Guo X, Zhang Y, Ma H, Peng X, Zhao Y, Li C, Gao L, Li T, Liang X, Ma C. Transcription factor Zhx2 is a checkpoint that programs macrophage polarization and antitumor response. Cell Death Differ 2023; 30:2104-2119. [PMID: 37582865 PMCID: PMC10482862 DOI: 10.1038/s41418-023-01202-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023] Open
Abstract
Macrophages are usually educated to tumor-associated macrophages (TAMs) in cancer with pro-tumor functions by tumor microenvironment (TME) and TAM reprogramming has been proposed as a potential tumor immunotherapy strategy. We recently demonstrated the critical role of Zinc-fingers and homeoboxes 2 (Zhx2) in macrophages' metabolic programming. However, whether Zhx2 is responsible for macrophage polarization and TAMs reprogramming is largely unknown. Here, we show that Zhx2 controls macrophage polarization under the inflammatory stimulus and TME. Myeloid-specific deletion of Zhx2 suppresses LPS-induced proinflammatory polarization but promotes IL-4 and TME-induced anti-inflammatory and pro-tumoral phenotypes in murine liver tumor models. Factors in TME, especially lactate, markedly decrease the expression of Zhx2 in TAMs, leading to the switch of TAMs to pro-tumor phenotype and consequent cancer progression. Notably, reduced ZHX2 expression in TAM correlates with poor survival of HCC patients. Mechanistic studies reveal that Zhx2 associates with NF-κB p65 and binds to the Irf1 promoter, leading to transcriptional activation of Irf1 in macrophages. Zhx2 functions in maintaining macrophage polarization by regulating Irf1 transcription, which may be a potential target for macrophage-based cancer immunotherapy.
Collapse
Affiliation(s)
- Siyu Tan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), Qingdao, China
| | - Na Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Xiaowei Guo
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Hongxin Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| | - Xueqi Peng
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Ying Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China.
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
37
|
Zhang K, Zhang J, Kan C, Tian H, Ma Y, Huang N, Han F, Hou N, Sun X. Role of dysfunctional peri-organ adipose tissue in metabolic disease. Biochimie 2023; 212:12-20. [PMID: 37019205 DOI: 10.1016/j.biochi.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Metabolic disease is a complex disorder defined by a group with interrelated factors. There is growing evidence that obesity can lead to a variety of metabolic diseases, including diabetes and cardiovascular disease. Excessive adipose tissue (AT) deposition and ectopic accumulation can lead to increased peri-organ AT thickness. Dysregulation of peri-organ (perivascular, perirenal, and epicardial) AT is strongly associated with metabolic disease and its complications. The mechanisms include secretion of cytokines, activation of immunocytes, infiltration of inflammatory cells, involvement of stromal cells, and abnormal miRNA expression. This review discusses the associations and mechanisms by which various types of peri-organ AT affect metabolic diseases while addressing it as a potential future treatment strategy.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongzhan Tian
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yanhui Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| |
Collapse
|
38
|
Yang L, Tu PH, Zhang CX, Xie RR, Dong M, Jing Y, Chen X, Wei G, Song HD. Influence of two anti-tumor drugs, pazopanib, and axitinib, on the development and thyroid-axis of zebrafish ( Danio rerio) embryos/larvae. Front Endocrinol (Lausanne) 2023; 14:1204678. [PMID: 37600710 PMCID: PMC10433177 DOI: 10.3389/fendo.2023.1204678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction In recent years, the potential toxicities of different pharmaceuticals toward the thyroid system have received increasing attention. In this study, we aim to evaluate the toxic effects of pazopanib and axitinib, two anti-tumor drugs with widespread clinical use, on thyroid function in the zebrafish model. Methods We measured levels of thyroid-related hormones using the commercial Enzyme-Linked Immunosorbent Assay (ELISA) kit. Whole-mount in situ hybridization (WISH) analysis was employed to detect target gene expression changes. Morphology of the thyroid were evaluated by using transgenic Tg (tg: EGFP) fish line under a confocal microscope. The relative mRNA expression of key genes was verified through quantitative real-time polymerase chain reaction (RT‒qPCR). The size and number of the follicles was quantified whereby Hematoxylin-Eosin (H & E) staining under a light microscope. Results The results revealed that fertilized zebrafish embryos were incubated in pazopanib or axitinib for 96 hours, development and survival were significantly affected, which was accompanied by significant disturbances in thyroid endocrine system (e.g., increased thyroid-stimulating hormone (TSH) content and decreased triiodothyronine (T3) and thyroxine (T4) content, as well as transcription changes of genes associated with the hypothalamus-pituitary-thyroid (HPT) axis. Moreover, based on whole-mount in situ hybridization staining of tg and histopathological examination of zebrafish embryos treated with pazopanib and axitinib, we observed a significantly abnormal development of thyroid follicles in the Tg (tg: EGFP) zebrafish transgenic line. Conclusion Collectively, these findings indicate that pazopanib and axitinib may have toxic effects on thyroid development and function, at least partially, by influencing the regulation of the HPT axis. Thus, we believe that the potential thyroid toxicities of pazopanib and axitinib in their clinical applications should receive greater attention.
Collapse
Affiliation(s)
- Liu Yang
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ping-hui Tu
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cao-xu Zhang
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Rong-rong Xie
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Mei Dong
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Jing
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xia Chen
- Department of Endocrinology, Shanghai Gongli Hospital, Shanghai, China
| | - Gang Wei
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Endocrinology and Metabolism, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Huai-dong Song
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
BAO WEI, HAN QIANGUANG, GUAN XIAO, WANG ZIJIE, GU MIN. Solute carrier-related signature for assessing prognosis and immunity in patients with clear-cell renal cell carcinoma. Oncol Res 2023; 31:181-192. [PMID: 37304236 PMCID: PMC10208045 DOI: 10.32604/or.2023.028051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/07/2023] [Indexed: 06/13/2023] Open
Abstract
Background Clear-cell renal cell carcinoma (ccRCC) is the most common malignant kidney cancer. However, the tumor microenvironment and crosstalk involved in metabolic reprogramming in ccRCC are not well-understood. Methods We used The Cancer Genome Atlas to obtain ccRCC transcriptome data and clinical information. The E-MTAB-1980 cohort was used for external validation. The GENECARDS database contains the first 100 solute carrier (SLC)-related genes. The predictive value of SLC-related genes for ccRCC prognosis and treatment was assessed using univariate Cox regression analysis. An SLC-related predictive signature was developed through Lasso regression analysis and used to determine the risk profiles of patients with ccRCC. Patients in each cohort were separated into high- and low-risk groups based on their risk scores. The clinical importance of the signature was assessed through survival, immune microenvironment, drug sensitivity, and nomogram analyses using R software. Results SLC25A23, SLC25A42, SLC5A1, SLC3A1, SLC25A37, SLC5A6, SLCO5A1, and SCP2 comprised the signatures of the eight SLC-related genes. Patients with ccRCC were separated into high- and low-risk groups based on the risk value in the training and validation cohorts; the high-risk group had a significantly worse prognosis (p < 0.001). The risk score was an independent predictive indicator of ccRCC in the two cohorts according to univariate and multivariate Cox regression (p < 0.05). Analysis of the immune microenvironment showed that immune cell infiltration and immune checkpoint gene expression differed between the two groups (p < 0.05). Drug sensitivity analysis showed that compared to the low-risk group, the high-risk group was more sensitive to sunitinib, nilotinib, JNK-inhibitor-VIII, dasatinib, bosutinib, and bortezomib (p < 0.001). Survival analysis and receiver operating characteristic curves were validated using the E-MTAB-1980 cohort. Conclusions SLC-related genes have predictive relevance in ccRCC and play roles in the immunological milieu. Our results provide insight into metabolic reprogramming in ccRCC and identify promising treatment targets for ccRCC.
Collapse
Affiliation(s)
- WEI BAO
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - QIANGUANG HAN
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - XIAO GUAN
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - ZIJIE WANG
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - MIN GU
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Zhang ZH, Wang Y, Zhang Y, Zheng SF, Feng T, Tian X, Abudurexiti M, Wang ZD, Zhu WK, Su JQ, Zhang HL, Shi GH, Wang ZL, Cao DL, Ye DW. The function and mechanisms of action of circular RNAs in Urologic Cancer. Mol Cancer 2023; 22:61. [PMID: 36966306 PMCID: PMC10039696 DOI: 10.1186/s12943-023-01766-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Kidney, bladder, and prostate cancer are the three major tumor types of the urologic system that seriously threaten human health. Circular RNAs (CircRNAs), special non-coding RNAs with a stabile structure and a unique back-splicing loop-forming ability, have received recent scientific attention. CircRNAs are widely distributed within the body, with important biologic functions such as sponges for microRNAs, as RNA binding proteins, and as templates for regulation of transcription and protein translation. The abnormal expression of circRNAs in vivo is significantly associated with the development of urologic tumors. CircRNAs have now emerged as potential biomarkers for the diagnosis and prognosis of urologic tumors, as well as targets for the development of new therapies. Although we have gained a better understanding of circRNA, there are still many questions to be answered. In this review, we summarize the properties of circRNAs and detail their function, focusing on the effects of circRNA on proliferation, metastasis, apoptosis, metabolism, and drug resistance in kidney, bladder, and prostate cancers.
Collapse
Affiliation(s)
- Zi-Hao Zhang
- Qingdao Institute, School of Life Medicine, Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Qingdao, 266500, China
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Yue Wang
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Ya Zhang
- Department of Nephrology, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Sheng-Feng Zheng
- Qingdao Institute, School of Life Medicine, Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Qingdao, 266500, China
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Tao Feng
- Qingdao Institute, School of Life Medicine, Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Qingdao, 266500, China
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Xi Tian
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Mierxiati Abudurexiti
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Zhen-Da Wang
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Wen-Kai Zhu
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Jia-Qi Su
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Hai-Liang Zhang
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Guo-Hai Shi
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Zi-Liang Wang
- Institute of Cancer Research, Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Da-Long Cao
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Ding-Wei Ye
- Department of Urology, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200433, China.
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
41
|
Dong K, Wei G, Sun H, Gu D, Liu J, Wang L. Metabolic crosstalk between thermogenic adipocyte and cancer cell: Dysfunction and therapeutics. Curr Opin Pharmacol 2023; 68:102322. [PMID: 36502545 DOI: 10.1016/j.coph.2022.102322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 12/13/2022]
Abstract
As one of the largest endocrine organs with a wide distribution in organisms, adipose tissue secretes multiple adipokines, cytokines, metabolites, and exosomes to promote tumour development. Elaborating the crosstalk between cancer cells and adipocytes provides a tissue-level perspective of cancer progression, which reflects the heterogeneity and complexity of human tumours. Three main types of adipose tissues, white, brown, and beige adipose tissue, have been described. Thermogenic capacity is a prominent characteristic of brown and beige adipocytes. Most studies so far mainly focus on the contribution of white adipocytes to the tumour microenvironment. However, the role of thermogenic adipose tissue in malignant cancer behaviour has been largely overlooked. Recently, emerging evidence suggests that beige/brown adipocytes play a key role in the development and progression of various cancers. This review focuses on the bidirectional communication between tumour cells and thermogenic adipocytes and the therapeutic strategies to disrupt this interaction.
Collapse
Affiliation(s)
- Kai Dong
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Gang Wei
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Honglin Sun
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Di Gu
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Junli Liu
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
42
|
Abstract
Rather than serving as a mere onlooker, adipose tissue is a complex endocrine organ and active participant in disease initiation and progression. Disruptions of biological processes operating within adipose can disturb healthy systemic physiology, the sequelae of which include metabolic disorders such as obesity and type 2 diabetes. A burgeoning interest in the field of adipose research has allowed for the elucidation of regulatory networks underlying both adipose tissue function and dysfunction. Despite this progress, few diseases are treated by targeting maladaptation in the adipose, an oft-overlooked organ. In this review, we elaborate on the distinct subtypes of adipocytes, their developmental origins and secretory roles, and the dynamic interplay at work within the tissue itself. Central to this discussion is the relationship between adipose and disease states, including obesity, cachexia, and infectious diseases, as we aim to leverage our wealth of knowledge for the development of novel and targeted therapeutics.
Collapse
Affiliation(s)
- Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; .,Howard Hughes Medical Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
43
|
Li B, Liu S, Yang Q, Li Z, Li J, Wu J, Sun S, Xu Z, Sun S, Wu Q. Macrophages in Tumor-Associated Adipose Microenvironment Accelerate Tumor Progression. Adv Biol (Weinh) 2023; 7:e2200161. [PMID: 36266968 DOI: 10.1002/adbi.202200161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/02/2022] [Indexed: 11/06/2022]
Abstract
Adipose-tissue macrophages (ATMs), a complex ensemble of diverse macrophage subtypes, are prevalent in the tumor-adipose microenvironment (TAME) and facilitate tumor growth. However, the mechanisms in which the tumor-adipocyte crosstalk may enable the properties and plasticity of macrophages remain unclear. The single-cell RNA-sequence profiling reveals that a subset of macrophages expressed CD163, CCL2, and CCL5 in TAME, exhibiting an immunosuppressive subtype. It is demonstrated that CD163+ macrophages aggregate to surround adipocytes in breast cancer tissues. The expressions of CCL2 and CCL5 are also elevated in TAME and enable the recruitment and polarize macrophages. Mechanically, the level of exosomal miRNA-155 increased in the coculture of tumor cells and adipocytes, and then it promoted the generation and release of CCL2 and CCL5 from adipocytes by targeting the SOCS6/STAT3 pathway. Inhibition of exosomal miRNA-155 in tumor cells reduced the CCL2 and CCL5 levels in tumor-adipocytes coculture and further retarded tumor growth. Finally, the deletion of macrophages partially inhibited adipocyte-induced tumor proliferation. Likewise, inhibiting chemokines and their receptors or suppressing the phosphorylation of STAT3 decreased tumor burden in preclinical models. These results demonstrate that the niche factors in TAME, such as exosomal miRNA-155, regulate the function and polarity of macrophages to facilitate tumor progression.
Collapse
Affiliation(s)
- Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Siqing Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Qian Yang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Zhiyu Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Juan Wu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Zhiliang Xu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China.,Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200092, P. R. China
| |
Collapse
|
44
|
Yang J, Wang K, Yang Z. Treatment strategies for clear cell renal cell carcinoma: Past, present and future. Front Oncol 2023; 13:1133832. [PMID: 37025584 PMCID: PMC10070676 DOI: 10.3389/fonc.2023.1133832] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent histological subtype of kidney cancer, which is prone to metastasis, recurrence, and resistance to radiotherapy and chemotherapy. The burden it places on human health due to its refractory nature and rising incidence rate is substantial. Researchers have recently determined the ccRCC risk factors and optimized the clinical therapy based on the disease's underlying molecular mechanisms. In this paper, we review the established clinical therapies and novel potential therapeutic approaches for ccRCC, and we support the importance of investigating novel therapeutic options in the context of combining established therapies as a research hotspot, with the goal of providing diversified therapeutic options that promise to address the issue of drug resistance, with a view to the early realization of precision medicine and individualized treatment.
Collapse
Affiliation(s)
- Junwei Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhichun Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- *Correspondence: Zhichun Yang,
| |
Collapse
|
45
|
Jiang A, Luo P, Chen M, Fang Y, Liu B, Wu Z, Qu L, Wang A, Wang L, Cai C. A new thinking: deciphering the aberrance and clinical implication of copper-death signatures in clear cell renal cell carcinoma. Cell Biosci 2022; 12:209. [PMID: 36581992 PMCID: PMC9801655 DOI: 10.1186/s13578-022-00948-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
RATIONALE Recent research has indicated that cuprotosis, or copper induced cell death, is a novel type of cell death that could be utilized as a new weapon for cancer management. However, the characteristics and implications of such signatures in cancers, especially in clear cell renal cell cancer (ccRCC), remain elusive. METHODS Expression, methylation, mutation, clinical information, copy number variation, functional implication, and drug sensitivity data at the pan-cancer level were collected from The Cancer Genome Atlas. An unsupervised clustering algorithm was applied to decipher ccRCC heterogeneity. Immune microenvironment construction, immune therapy response, metabolic pattern, and cancer progression signature between subgroups were also investigated. RESULTS Cuprotosis related genes were specifically downregulated in various cancer tissues compared with normal tissues and were correlated with hypermethylation and copy number variation. Cuprotosis scores were also dysregulated in tumor tissues, and we found that such a signature could positively regulate oxidative phosphorylation and Myc and negatively regulate epithelial mesenchymal translation and myogenesis pathways. CPCS1 (cuprotosis scores high) and CPCS2 (cuprotosis scores low) in ccRCC displayed distinctive clinical profiles and biological characteristics; the CPCS2 subtype had a higher clinical stage and a worse prognosis and might positively regulate cornification and epidermal cell differentiation to fuel cancer progression. CPCS2 also displayed a higher tumor mutation burden and low tumor stemness index, while it led to a low ICI therapy response and dysfunctional tumor immunity state. The genome-copy numbers of CPCS2, including arm- gain and arm- loss, were higher than those of CPCS1. The prognostic model constructed based on subgroup biomarkers exerted satisfactory performance in both the training and validation cohorts. In addition, overexpression of the copper death activator DLAT suppressed the malignant ability, including cell migration and proliferation, of renal cell lines in vitro and in vivo. Finally, activation of cuprotosis in tumors could enhance antitumor immunity through dsDNA-cGAS-STING signaling in ccRCC. CONCLUSION The activation of cuprotosis might function as a promising approach among multiple cancers. The cuprotosis related signatures could reshape tumor immunity in the ccRCC microenvironment via cGAS-STING signal, thus activating tumor antigen-presenting process. Upregulation of DLAT expression in ccRCC cell lines could reactivate the copper death pattern and be treated as a suitable target for ccRCC.
Collapse
Affiliation(s)
- Aimin Jiang
- grid.73113.370000 0004 0369 1660Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433 China
| | - Peng Luo
- grid.284723.80000 0000 8877 7471Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280 China
| | - Ming Chen
- grid.73113.370000 0004 0369 1660Department of Urology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003 China
| | - Yu Fang
- grid.73113.370000 0004 0369 1660Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433 China
| | - Bing Liu
- grid.73113.370000 0004 0369 1660Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, 201805 China
| | - Zhenjie Wu
- grid.73113.370000 0004 0369 1660Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433 China
| | - Le Qu
- grid.41156.370000 0001 2314 964XDepartment of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210046 China
| | - Anbang Wang
- grid.73113.370000 0004 0369 1660Department of Urology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003 China
| | - Linhui Wang
- grid.73113.370000 0004 0369 1660Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433 China
| | - Chen Cai
- grid.73113.370000 0004 0369 1660Department of Special Clinic, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433 China
| |
Collapse
|
46
|
Zeng X, Teng Y, Zhu C, Li Z, Liu T, Sun Y, Han S. Combined Ibuprofen-Nanoconjugate Micelles with E-Selectin for Effective Sunitinib Anticancer Therapy. Int J Nanomedicine 2022; 17:6031-6046. [PMID: 36510619 PMCID: PMC9740013 DOI: 10.2147/ijn.s388234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Sunitinib, a first-line therapy with a certain effect, was utilized in the early stages of renal cell carcinoma treatment. However, its clinical toxicity, side effects, and its limited bioavailability, resulted in inadequate clinical therapeutic efficacy. Building neoteric, simple, and safe drug delivery systems with existing drugs offers new options. Therefore, we aimed to construct a micelle to improve the clinical efficacy of sunitinib by reusing ibuprofen. Methods We synthesized the sialic acid-poly (ethylene glycol)-ibuprofen (SA-PEG-IBU) amphipathic conjugate in two-step reaction. The SA-PEG-IBU amphiphilic conjugates can form into stable SPI nanomicelles in aqueous solution, which can be further loaded sunitinib (SU) to obtain the SPI/SU system. Following nanomicelle creation, sialic acid exposed to the nanomicelle surface can recognize the overexpressed E-selectin receptor on the membrane of cancer cells to enhance cellular uptake. The properties of morphology, stability, and drug release about the SPI/SU nanomicelles were investigated. Confocal microscopy and flow cytometry were used to assess the cellular uptake efficiency of nanomicelles in vitro. Finally, a xenograft tumor model in nude mice was constructed to investigate the body distribution and tumor suppression of SPI/SU in vivo. Results The result showed that SPI nanomicelles exhibited excellent tumor targeting performance and inhibited the migration and invasion of tumor cell in vitro. The SPI nanomicelles can improve the accumulation of drugs in the tumor site that showed effective tumor inhibition in vivo. In addition, H&E staining and immunohistochemical analysis demonstrated that the SPI/SU nanomicelles had a superior therapeutic effect and lower biotoxicity. Conclusion The SPI/SU nanomicelles displayed excellent anti-tumor ability, and can suppress the metastasis of tumor cell by decreasing the expression of Cyclooxygenase-2 due to the ibuprofen, providing an optimistic clinical application potential by developing a simple but safe drug delivery system.
Collapse
Affiliation(s)
- Xianhu Zeng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China
| | - Yi Teng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China
| | - Chunrong Zhu
- Department of Pharmacy Intravenous Admixture Service, Weifang Maternal and Child Health Hospital, Weifang, People’s Republic of China
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China
| | - Tian Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China,Correspondence: Shangcong Han; Yong Sun, Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China, Tel/Fax +86 532 82991508, Email ;
| |
Collapse
|
47
|
Guo H, Zhang Y, Ma H, Gong P, Shi Y, Zhao W, Wang A, Liu M, Sun Z, Wang F, Wang Q, Ba X. T-stage-specific abdominal visceral fat, haematological nutrition indicators and inflammation as prognostic factors in patients with clear renal cell carcinoma. Adipocyte 2022; 11:133-142. [PMID: 35285399 PMCID: PMC8920171 DOI: 10.1080/21623945.2022.2048546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Clear cell renal carcinoma (ccRCC) is the most common histological type of renal cancer and has the highest mortality. Several studies have been conducted on the relationship between adipose tissue and ccRCC prognosis, however, the results have been inconsistent to date. The current study aimed at establishing a link between abdominal fat composition and short-term prognosis in patients with ccRCC after T-stage stratification. We retrospectively analysed 250 patients with pathologically confirmed ccRCC (173 low T-stage and 77 high T-stage) in our hospital. The computed tomography (CT) images were evaluated using ImageJ. Then, subcutaneous and visceral fat areas (SFA and VFA), total fat areas (TFA) and the relative VFA (rVFA) were measured and computed. Meanwhile, biochemical indices of blood serum were analysed. The results showed that rVFA in low T-stage cohort who had a history of short-term postoperative complications were significantly lower than those who did not. No such association was observed in the high T-stage cohort. Further investigation revealed that the correlations between biochemical indexes and fat area-related variables varied across T-stage groups. As a result, rVFA is a reliable independent predictor of short-term prognosis in patients with low T-stage ccRCC but not in patients with high T-stage ccRCC.
Collapse
Affiliation(s)
- Hao Guo
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong province, Jinan, China
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Yumei Zhang
- Department of Radiology, Lanshan Branch of Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Peiyou Gong
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Yinghong Shi
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Wenlei Zhao
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Aijie Wang
- Department of Radiology, Yaitai Shan Hospital, Shandong province, Yantai, China
| | - Ming Liu
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Zehua Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Fang Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong province, Jinan, China
| | - Qing Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong province, Jinan, China
| | - Xinru Ba
- Department of Radiology, Yaitai Shan Hospital, Shandong province, Yantai, China
| |
Collapse
|
48
|
Wei G, Zhang CX, Jing Y, Chen X, Song HD, Yang L. The influence of sunitinib and sorafenib, two tyrosine kinase inhibitors, on development and thyroid system in zebrafish larvae. CHEMOSPHERE 2022; 308:136354. [PMID: 36087734 DOI: 10.1016/j.chemosphere.2022.136354] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
Recently, the potential toxic effects of various pharmaceuticals on the thyroid endocrine system have raised considerable concerns. In this study, we evaluated the adverse effects of sorafenib and sunitinib, two widely used anti-tumor drugs, on the developmental toxicities and thyroid endocrine disruption by using zebrafish (Danio rerio) model. Zebrafish embryos/larvae were exposed to different contentions (0, 10, 50 and 100 nM) of sorafenib and sunitinib for 96 hpf. The results revealed that waterborne exposure to sorafenib and sunitinib exhibited remarkable toxic effects on the survival and development in zebrafish embryos/larvae, which was accompanied by obvious disturbances of thyroid endocrine system (e.g., decreased T3 and T4 content, increased TSH content) and genes' transcription changes within the hypothalamus-pituitary-thyroid (HPT) axis. In addition, we verified a strikingly abnormal thyroid gland organogenesis in zebrafish larvae in response to sorafenib and sunitinib, by assessing the development of thyroid follicles using the WISH staining of tg, the Tg (tg:GFP) zebrafish transgenic line, and histopathological analysis. Taken together, our results indicated sorafenib and sunitinib exposure could induce obvious developmental toxicities and thyroid function disruption in zebrafish embryos/larvae, which might involve a regulatory mechanism, at least in part, by destroying the thyroid follicle structure, and by disturbing the balance of the HPT axis.
Collapse
Affiliation(s)
- Gang Wei
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Hangzhou, 310015, China; Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Cao-Xu Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yu Jing
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xia Chen
- Department of Endocrinology, Shanghai Gongli Hospital, Shanghai, 200135, China
| | - Huai-Dong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Liu Yang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
49
|
Li J, Qiu J, Han J, Li X, Jiang Y. Tumor Microenvironment Characterization in Breast Cancer Identifies Prognostic Pathway Signatures. Genes (Basel) 2022; 13:1976. [PMID: 36360212 PMCID: PMC9690299 DOI: 10.3390/genes13111976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 01/07/2024] Open
Abstract
Breast cancer is one of the most common female malignancies worldwide. Due to its early metastases formation and a high degree of malignancy, the 10 year-survival rate of metastatic breast cancer does not exceed 30%. Thus, more precise biomarkers are urgently needed. In our study, we first estimated the tumor microenvironment (TME) infiltration using the xCell algorithm. Based on TME infiltration, the three main TME clusters were identified using consensus clustering. Our results showed that the three main TME clusters cause significant differences in survival rates and TME infiltration patterns (log-rank test, p = 0.006). Then, multiple machine learning algorithms were used to develop a nine-pathway-based TME-related risk model to predict the prognosis of breast cancer (BRCA) patients (the immune-related pathway-based risk score, defined as IPRS). Based on the IPRS, BRCA patients were divided into two subgroups, and patients in the IPRS-low group presented significantly better overall survival (OS) rates than the IPRS-high group (log-rank test, p < 0.0001). Correlation analysis revealed that the IPRS-low group was characterized by increases in immune-related scores (cytolytic activity (CYT), major histocompatibility complex (MHC), T cell-inflamed immune gene expression profile (GEP), ESTIMATE, immune, and stromal scores) while exhibiting decreases in tumor purity, suggesting IPRS-low patients may have a strong immune response. Additionally, the gene-set enrichment analysis (GSEA) result confirmed that the IPRS-low patients were significantly enriched in several immune-associated signaling pathways. Furthermore, multivariate Cox analysis revealed that the IPRS was an independent prognostic biomarker after adjustment by clinicopathologic characteristics. The prognostic value of the IPRS model was further validated in three external validation cohorts. Altogether, our findings demonstrated that the IPRS was a powerful predictor to screen out certain populations with better prognosis in breast cancer and may serve as a potential biomarker guiding clinical treatment decisions.
Collapse
Affiliation(s)
- Ji Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Jiayue Qiu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xiangmei Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Ying Jiang
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| |
Collapse
|
50
|
Yin X, Chen Y, Ruze R, Xu R, Song J, Wang C, Xu Q. The evolving view of thermogenic fat and its implications in cancer and metabolic diseases. Signal Transduct Target Ther 2022; 7:324. [PMID: 36114195 PMCID: PMC9481605 DOI: 10.1038/s41392-022-01178-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractThe incidence of metabolism-related diseases like obesity and type 2 diabetes mellitus has reached pandemic levels worldwide and increased gradually. Most of them are listed on the table of high-risk factors for malignancy, and metabolic disorders systematically or locally contribute to cancer progression and poor prognosis of patients. Importantly, adipose tissue is fundamental to the occurrence and development of these metabolic disorders. White adipose tissue stores excessive energy, while thermogenic fat including brown and beige adipose tissue dissipates energy to generate heat. In addition to thermogenesis, beige and brown adipocytes also function as dynamic secretory cells and a metabolic sink of nutrients, like glucose, fatty acids, and amino acids. Accordingly, strategies that activate and expand thermogenic adipose tissue offer therapeutic promise to combat overweight, diabetes, and other metabolic disorders through increasing energy expenditure and enhancing glucose tolerance. With a better understanding of its origins and biological functions and the advances in imaging techniques detecting thermogenesis, the roles of thermogenic adipose tissue in tumors have been revealed gradually. On the one hand, enhanced browning of subcutaneous fatty tissue results in weight loss and cancer-associated cachexia. On the other hand, locally activated thermogenic adipocytes in the tumor microenvironment accelerate cancer progression by offering fuel sources and is likely to develop resistance to chemotherapy. Here, we enumerate current knowledge about the significant advances made in the origin and physiological functions of thermogenic fat. In addition, we discuss the multiple roles of thermogenic adipocytes in different tumors. Ultimately, we summarize imaging technologies for identifying thermogenic adipose tissue and pharmacologic agents via modulating thermogenesis in preclinical experiments and clinical trials.
Collapse
|