1
|
Ren X, Wang G, Chen J, Liu L. A Population-based Analysis of Gender Differences in Survival Outcomes for Gastric Gastrointestinal Stromal Tumors. J Gastrointest Surg 2025:102055. [PMID: 40210083 DOI: 10.1016/j.gassur.2025.102055] [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: 12/18/2024] [Revised: 03/03/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND While gender-based differences have been demonstrated to impact the prognosis of multiple tumors, their specific influence on the survival of gastric gastrointestinal stromal tumors (gGISTs) at the population level is still uncertain. Consequently, we aimed to investigate gender disparities in the prognosis of gGISTs by utilizing the Surveillance, Epidemiology, and End Results (SEER) database. METHODS Patients with gGISTs from the years 2000 to 2019 were retrieved from the SEER database. To minimize selection bias, propensity score matching (PSM) was employed. Kaplan-Meier analyses and Cox proportional hazard models were utilized to assess the influence of clinical characteristics on overall survival (OS) and cancer-specific survival (CSS). RESULTS A total of 3006 patients with gGISTs were analyzed, including 1459 males and 1547 females. When compared to female patients, male patients exhibited a higher proportion of Caucasians, more advanced T stage, larger tumor sizes, and elevated mitotic index. Before PSM, male patients experienced significantly worse OS (hazard ratio [HR] 1.55, 95% confidence interval [CI] 1.33-1.81, P < 0.001) and CSS (HR 1.61, 95% CI 1.28-2.02, P < 0.001) outcomes compared to female patients. Furthermore, they had lower mean OS and CSS rates at the 1-, 3-, and 5-year follow-up intervals (P < 0.05). Even after PSM, male patients continued to show poorer OS (HR 1.30, 95% CI 1.10-1.54, P = 0.002) and CSS (HR 1.34, 95% CI 1.05-1.70, P = 0.016) outcomes compared to female patients, with persistently lower mean OS and CSS rates across the 1-, 3-, and 5-year follow-up intervals (P < 0.05). For males without distant metastasis, 5-year OS was 78.8% and CSS was 90.0%, both lower than females' 85.5% and 93.7% (P < 0.05). For males with distant metastasis, 5-year OS was 48.7% and CSS was 58.8%, similar to females' 55.4% and 65.3% (P > 0.05). Multivariate Cox regression analysis identified age, race, sex, M stage, surgical intervention, tumor size, and mitotic index as independent risk factors for both OS and CSS. CONCLUSIONS Patients with gGISTs exhibit distinct clinical characteristics between males and females, with female patients showing a tendency towards improved OS and CSS. Surgical treatment has the potential to enhance the prognosis of patients with gGISTs.
Collapse
Affiliation(s)
- Xia Ren
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou, China
| | - Ganhong Wang
- Department of Gastroenterology, Changshu Traditional Chinese Medical Hospital, Suzhou, China
| | - Jian Chen
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou, China.
| | - Luojie Liu
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Gao X, Liu J, Jia B, Guo J. USP30 Aggravating the Malignant Progression of Breast Cancer and Its Resistance to Tamoxifen by Inhibiting the Ubiquitination of TOMM40. J Biochem Mol Toxicol 2025; 39:e70258. [PMID: 40227042 DOI: 10.1002/jbt.70258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Breast cancer (BC) is the most common malignancy among women, with high incidence and mortality rates globally. Translocase of outer mitochondrial membrane 40 (TOMM40) has also been identified as an important prognostic biomarker for BC. Meanwhile, the ubiquitin-specific protease 30 (USP30) has also been shown to promote BC progression. However, the specific mechanisms underlying the role of USP30/TOMM40 in BC development remain unclear. Therefore, this study aims to delve into the potential mechanisms of USP30/TOMM40 in the progression of BC. The expression of TOMM40 and USP30 in BC tumors and cells was verified by bioinformatics analysis and western blot (WB). The effects of USP30/TOMM40 on BC cell proliferation, angiogenesis, glycolysis, and ferroptosis were determined by colony formation, tube formation assays and commercial kits. The co-immunoprecipitation (Co-IP) experiment was applied to verify the interaction between USP30 and TOMM40. The ubiquitination level of TOMM40 was detected by ubiquitinated antibodies. The effect of tamoxifen (TAM) on BC cell viability was measured by MTT assay. TOMM40 and USP30 were highly expressed in BC tumors and cells. Silencing TOMM40 blocked the proliferation, angiogenesis, glycolytic, and induced ferroptosis of BC cells. USP30 bound to TOMM40 and reduced its ubiquitination level. TOMM40 overexpressed abolished the tumor suppressive effect of USP30 knockdown and enhanced the resistance of BC to TAM. In conclusion, USP30 deubiquitinating TOMM40 promoted BC development and TAM resistance.
Collapse
Affiliation(s)
- Xinran Gao
- Department of Breast Tumor Surgery, Cancer Center, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China
| | - Junbiao Liu
- Department of Breast Tumor Surgery, Cancer Center, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China
| | - Baoqing Jia
- Department of Breast Tumor Surgery, Cancer Center, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China
| | - Jiaxin Guo
- Graduate School of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Cheng B, Wu J, Chen K, Li W, Yang J, Shangguan W, Yang L, Huang W, Ma C, Li Z, Sun B, Wang Q, Huang H, Wu P. Association of 5α-reductase inhibitor prescription with immunotherapy efficacy in metastatic renal cell carcinoma: a multicenter retrospective analysis. J Immunother Cancer 2025; 13:e011154. [PMID: 40010773 DOI: 10.1136/jitc-2024-011154] [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] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic renal cell carcinoma (mRCC), but response rates remain heterogeneous, and reliable predictive biomarkers are lacking. Recent studies suggest that androgen receptor (AR) signaling plays a role in regulating CD8+ T-cell function, implying that 5α-reductase inhibitors (5-ARIs), which lower androgen activity, could enhance antitumor immunity and improve clinical outcomes in patients receiving immunotherapy. This study retrospectively investigates the impact of a history of 5-ARI use (≥12 months) on the efficacy of ICIs in mRCC. METHODS We conducted a multicenter retrospective cohort study of 185 patients with mRCC who received ICIs. Patients were stratified based on their history of 5-ARI use. Baseline characteristics included age, body mass index, International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk group, programmed death-ligand 1 (PD-L1) expression levels, tumor stage, and metastasis sites. The primary endpoints were progression-free survival (PFS) and overall survival (OS), analyzed using Cox proportional hazards models. Secondary endpoints included objective response rate (ORR) and disease control rate (DCR). Key immunological insights were gained through single-cell RNA sequencing analysis of tumor samples. RESULTS Patients with a history of 5-ARI use demonstrated improved ORR (59.8% vs 39.8%, p=0.0075) and DCR (87.0% vs 78.7%, p=0.1747) compared with those without. The median PFS and OS were significantly longer in the 5-ARI group, with HRs of 0.64 (95% CI: 0.47 to 0.86, p=0.0085) for PFS and 0.65 (95% CI: 0.47 to 0.90, p=0.0271) for OS. Subgroup analysis further indicated enhanced ICI efficacy with 5-ARI use across age, IMDC risk scores, and PD-L1 expression levels. Single-cell RNA sequencing analysis revealed that 5-ARI treated patients exhibited a reduced presence of regulatory T cells and CD8 T-cell exhaustion (CD8 Tex), and lower programmed cell death protein-1 expression in CD8 Tex cells, suggesting an immunologically favorable modification of the tumor. CONCLUSION A history of 5-ARI use is associated with improved responses to ICI therapy in mRCC, potentially through AR-related modulation of CD8+ T-cell activity and favorable alterations in the immune microenvironment. These findings support further investigation into androgen-targeted approaches as adjunctive strategies in immunotherapy for RCC.
Collapse
Affiliation(s)
- Bisheng Cheng
- Department of Urology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Jilin Wu
- Department of Urology, Peking University People's Hospital, Beijing, Beijing, China
| | - Ke Chen
- Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Weijia Li
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianghua Yang
- Department of Urology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - WenTai Shangguan
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Lin Yang
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenxue Huang
- Department of Urology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cunzhen Ma
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhuohang Li
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Boyuan Sun
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiong Wang
- Department of Urology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Peng Wu
- Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Agnoletto A, Brisken C. Hormone Signaling in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:279-307. [PMID: 39821031 DOI: 10.1007/978-3-031-70875-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Hormones control normal breast development and function. They also impinge on breast cancer (BC) development and disease progression in direct and indirect ways. The major ovarian hormones, estrogens and progesterone, have long been established as key regulators of mammary gland development in rodents and linked to human disease. However, their roles have been difficult to disentangle because they act on multiple tissues and can act directly and indirectly on different cell types in the breast, and their receptors interact at different levels within the target cell. Estrogens are well-recognized drivers of estrogen receptor-positive (ER+) breast cancers, and the ER is successfully targeted in ER+ disease. The role of progesterone receptor (PR) as a potential target to be activated or inhibited is debated, and androgen receptor (AR) signaling has emerged as a potentially interesting pathway to target on the stage.In this chapter, we discuss hormone signaling in normal breast development and in cancer, with a specific focus on the key sex hormones: estrogen, progesterone, and testosterone. We will highlight the complexities of endocrine control mechanisms at the organismal, tissue, cellular, and molecular levels. As we delve into the mechanisms of action of hormone receptors, their interplay and their context-dependent roles in breast cancer will be discussed. Drawing insights from new preclinical models, we will describe the lessons learned and the current challenges in understanding hormone action in breast cancer.
Collapse
Affiliation(s)
- Andrea Agnoletto
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Hargrove-Wiley E, Obodo D, Bindeman W, Fingleton B. Elucidating Sex-Specific Immune Profiles in a Breast Cancer Model. Int J Mol Sci 2024; 25:13113. [PMID: 39684829 DOI: 10.3390/ijms252313113] [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: 10/30/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer is commonly thought of as a "women's disease". However, men are increasingly diagnosed with the disease, and their mortality rates are disparately higher than those of female patients. The abundance and composition of the immune microenvironment are determinants of breast cancer progression and survival. It is well documented that there are sex-specific differences in the immune response to several diseases, including various cancers. However, the effects of these differences in the context of breast cancer remain to be explored. This study demonstrates sex differences in the hormonal and immune landscape of the MMTV-PyMT transgenic murine model of female and male ER+ breast cancer using single-cell RNA sequencing (scRNA-Seq), whole-slide immunohistochemistry, and flow cytometry. Mammary tumors of transgenic male mice had increased estrogen receptor alpha expression and enriched nuclear binding signatures compared to female tumors. In the tumor immune compartment, male mice had lower intratumoral leukocyte infiltration. Yet, scRNA-Seq analysis reveals a more immunostimulatory microenvironment and increased antitumor immune populations in the primary and metastatic lungs as compared to transgenic females. Despite a more favorable innate immune profile, the metastatic burden was increased in male mice. Our data support a sex-dependent immune response in mammary carcinoma associated with the tumor, and likely host, hormonal environment. With emerging therapeutics targeting the tumor immune microenvironment, characterizing immune profiles is critical for optimizing their use in all breast cancer patients.
Collapse
Affiliation(s)
- Ebony Hargrove-Wiley
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Dora Obodo
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wendy Bindeman
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Barbara Fingleton
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Hu J, Luo Z, Song J, Kong D, Li Z, Chen C, Sun S. High C-reactive protein is associated with the efficacy of neoadjuvant chemotherapy for hormone receptor-positive breast cancer. Medicine (Baltimore) 2024; 103:e40775. [PMID: 39612416 DOI: 10.1097/md.0000000000040775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
C-reactive protein (CRP) is a nonspecific biomarker for systemic inflammatory response and is linked to the prognosis of breast cancer (BC); however, few studies have investigated the correlation between CRP and the effectiveness of neoadjuvant chemotherapy treatment for BC. We recruited 177 patients with BC who underwent neoadjuvant chemotherapy in our clinical trial. the median CRP level (0.24 mg/L), patients were categorized into high and low groups. We examined the relationship between CRP levels and various clinicopathological factors, including pathological complete response (pCR), using the chi-square test or Fisher exact test. Furthermore, we evaluated the predictive capacity of CRP for different molecular subtypes by constructing receiver operating characteristic curves. To identify the independent variables associated with pCR, we conducted logistic regression multivariate analysis. No association was found between C-reactive levels at baseline and pCR rates. CRP level was significantly associated with higher body mass index, and the high CRP group had more overweight patients (47.06% vs. 16.30%, P < .001). In hormone receptor-positive patients, the high CRP group demonstrated a significantly higher pCR rate (OR = 4.115, 95% CI: 1.481-11.36, P = .009). The areas under the curve was 0.670 (95% CI: 0.550-0.792, P < .001). Multivariate logistic analysis showed that the CRP level was a significant independent predictor of pCR (OR = 5.882, 95% CI: 1.470-28.57, P = .017). High CRP levels were found to be associated with a higher pCR rate, indicating their independent predictive value in determining the efficacy of neoadjuvant chemotherapy in hormone receptor-positive BC patients.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, PR China
| | | | | | | | | | | | | |
Collapse
|
7
|
Li S, Jin J, Zhang W, Cao Y, Qin H, Wang J, Yu J, Wang W. Association of weight-adjusted waist index with all-cause and cause-specific mortality among cancer survivors: a cohort study of the NHANES 1999-2018. Front Endocrinol (Lausanne) 2024; 15:1422071. [PMID: 39574952 PMCID: PMC11578743 DOI: 10.3389/fendo.2024.1422071] [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: 04/23/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024] Open
Abstract
Background Obesity is becoming more widely acknowledged as a chronic illness that raises the risk of oncogenesis. This inquiry aimed to look into the correlation between cancer patient mortality and obesity, as measured by the weight-adjusted waist index (WWI). Methods We used continuous data from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2018 as the benchmark, with a follow-up validity period of December 31, 2019. First, we assessed the correlation between WWI and the all-cause and cause-specific execution of cancer sufferers using multivariable Cox proportional hazards models. Second, a smoothed curve fit was utilized to examine the relationship between WWI and both cause-specific and all-cause mortality in cancer patients. Lastly, we analyzed the relationship between WWI and both cause-specific and all-cause mortality in cancer patients, to find out if this link held across the population subgroup evaluation and impact analyses were used as well during the last step. Results With a median follow-up of 87.8 months, 1,547 (34.7%) of the 4,463 cancer patients had deceased. Among them, 508 (11.4%) succumbed to cancer, while 322 (7.2%) passed away due to cardiovascular disease. Multivariate Cox proportional hazards model of mortality among cancer patients revealed an all-cause mortality hazard ratio [HR=1.13; 95% CI (1.04, 1.23)] and cardiovascular mortality [HR=1.39; 95% CI (1.16, 1.67)]. Furthermore, for each unit increase in WWI, all-cause mortality was significantly higher in male cancer survivors than in female cancer survivors. Conclusions Our study reveals substantial correlations between WWI and all-cause and cardiovascular mortality in US cancer survivors, helping to identify cancer survivors at higher risk of death and thus potentially guiding targeted interventions.
Collapse
Affiliation(s)
- Shi Li
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jing Jin
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wenshun Zhang
- The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Ying Cao
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Haiyun Qin
- The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jianguang Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiaxiang Yu
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wenping Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
8
|
Carleton N, Lee S, Li R, Zou J, Brown DD, Hooda J, Chang A, Kumar R, Klei LR, Rigatti LH, Newsome J, John Mary DJS, Atkinson JM, West RE, Nolin TD, Oberly PJ, Huang Z, Poirier D, Diego EJ, Lucas PC, Tseng G, Lotze MT, McAuliffe PF, Zervantonakis IK, Oesterreich S, Lee AV. Systemic and local chronic inflammation and hormone disposition promote a tumor-permissive environment for breast cancer in older women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.616978. [PMID: 39484485 PMCID: PMC11526964 DOI: 10.1101/2024.10.18.616978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Estrogen receptor positive (ER+) breast cancer is the most common subtype of breast cancer and is an age-related disease. The peak incidence of diagnosis occurs around age 70, even though these post-menopausal patients have low circulating levels of estradiol (E2). Despite the hormone sensitivity of age-related tumors, we have a limited understanding of the interplay between systemic and local hormones, chronic inflammation, and immune changes that contribute to the growth and development of these tumors. Here, we show that aged F344 rats treated with the dimethylbenz(a)anthracene / medroxyprogestrone acetate (DMBA/MPA) carcinogen develop more tumors at faster rates than their younger counterparts, suggesting that the aged environment promotes tumor initiation and impacts growth. Single-nuclei RNA-seq (snRNA-seq) of the tumors showed broad local immune dysfunction that was associated with circulating chronic inflammation. Across a broad cohort of specimens from patients with ER+ breast cancer and age-matched donors of normal breast tissue, we observe that even with an estrone (E1)-predominant estrogen disposition in the systemic circulation, tumors in older patients increase HSD17B7 expression to convert E1 to E2 in the tumor microenvironment (TME) and have local E2 levels similar to pre-menopausal patients. Concurrently, trackable increases in several chemokines, defined most notably by CCL2, promote a chronically inflamed but immune dysfunctional TME. This unique milieu in the aged TME, characterized by high local E2 and chemokine-enriched chronic inflammation, promotes both accumulation of tumor-associated macrophages (TAMs), which serve as signaling hubs, as well as polarization of TAMs towards a CD206+/PD-L1+, immunosuppressive phenotype. Pharmacologic targeting of estrogen signaling (either by HSD17B7 inhibition or with fulvestrant) and chemokine inflammation both decrease local E2 and prevent macrophage polarization. Overall, these findings suggest that chronic inflammation and hormonal disposition are critical contributors to the age-related nature of ER+ breast cancer development and growth and offer potential therapeutic insight to treat these patients. Translational Summary We uncover the unique underpinnings establishing how the systemic host environment contributes to the aged breast tumor microenvironment, characterized by high local estradiol and chronic inflammation with immune dysregulation, and show that targeting avenues of estrogen conversion and chronic inflammation work to restore anti-tumor immunity.
Collapse
|
9
|
Duan X, Nie Y, Xie X, Zhang Q, Zhu C, Zhu H, Chen R, Xu J, Zhang J, Yang C, Yu Q, Cai K, Wang Y, Tian W. Sex differences and testosterone interfere with the structure of the gut microbiota through the bile acid signaling pathway. Front Microbiol 2024; 15:1421608. [PMID: 39493843 PMCID: PMC11527610 DOI: 10.3389/fmicb.2024.1421608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Background The gut microbiome has a significant impact on human wellness, contributing to the emergence and progression of a range of health issues including inflammatory and autoimmune conditions, metabolic disorders, cardiovascular problems, and psychiatric disorders. Notably, clinical observations have revealed that these illnesses can display differences in incidence and presentation between genders. The present study aimed to evaluate whether the composition of gut microbiota is associated with sex-specific differences and to elucidate the mechanism. Methods 16S-rRNA-sequencing technology, hormone analysis, gut microbiota transplantation, gonadectomy, and hormone treatment were employed to investigate the correlation between the gut microbiome and sex or sex hormones. Meanwhile, genes and proteins involved bile acid signaling pathway were analyzed both in the liver and ileum tissues. Results The composition and diversity of the microbiota from the jejunum and feces and the level of sex hormones in the serum differed between the sexes in young and middle-aged Sprague Dawley (SD) rats. However, no similar phenomenon was found in geriatric rats. Interestingly, whether in young, middle-aged, or old rats, the composition of the microbiota and bacterial diversity differed between the jejunum and feces in rats. Gut microbiota transplantation, gonadectomy, and hormone replacement also suggested that hormones, particularly testosterone (T), influenced the composition of the gut microbiota in rats. Meanwhile, the mRNA and protein level of genes involved bile acid signaling pathway (specifically SHP, FXR, CYP7A1, and ASBT) exhibited gender-specific differences, and T may play a significant role in mediating the expression of this pathway. Conclusion Sex-specific differences in the structure of the gut microbiota are mediated by T through the bile acid signaling pathway, pointing to potential targets for disease prevention and management techniques by indicating that sex differences and T levels may alter the composition of the gut microbiota via the bile acid signaling pathway.
Collapse
Affiliation(s)
- Xueqing Duan
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Yinli Nie
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Xin Xie
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Qi Zhang
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Chen Zhu
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Han Zhu
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Rui Chen
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Jun Xu
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Jinqiang Zhang
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Changfu Yang
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Qi Yu
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Kun Cai
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Yong Wang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Weiyi Tian
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| |
Collapse
|
10
|
Coradini D. Impact of De Novo Cholesterol Biosynthesis on the Initiation and Progression of Breast Cancer. Biomolecules 2024; 14:64. [PMID: 38254664 PMCID: PMC10813427 DOI: 10.3390/biom14010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024] Open
Abstract
Cholesterol (CHOL) is a multifaceted lipid molecule. It is an essential structural component of cell membranes, where it cooperates in regulating the intracellular trafficking and signaling pathways. Additionally, it serves as a precursor for vital biomolecules, including steroid hormones, isoprenoids, vitamin D, and bile acids. Although CHOL is normally uptaken from the bloodstream, cells can synthesize it de novo in response to an increased requirement due to physiological tissue remodeling or abnormal proliferation, such as in cancer. Cumulating evidence indicated that increased CHOL biosynthesis is a common feature of breast cancer and is associated with the neoplastic transformation of normal mammary epithelial cells. After an overview of the multiple biological activities of CHOL and its derivatives, this review will address the impact of de novo CHOL production on the promotion of breast cancer with a focus on mammary stem cells. The review will also discuss the effect of de novo CHOL production on in situ and invasive carcinoma and its impact on the response to adjuvant treatment. Finally, the review will discuss the present and future therapeutic strategies to normalize CHOL biosynthesis.
Collapse
Affiliation(s)
- Danila Coradini
- Laboratory of Medical Statistics and Biometry, "Giulio A. Maccacaro", Department of Clinical Sciences and Community Health, University of Milan, Campus Cascina Rosa, 20133 Milan, Italy
| |
Collapse
|