1
|
Blanco R, Quezada-Romegialli C, Muñoz JP. Bovine Leukemia Virus and Human Breast Cancer: A Review of Clinical and Molecular Evidence. Viruses 2025; 17:324. [PMID: 40143252 PMCID: PMC11946124 DOI: 10.3390/v17030324] [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: 12/28/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Despite significant advancements in early diagnosis and treatment, breast cancer (BC) remains a major global health challenge. Ongoing research is essential to identify novel risk factors, implement innovative screening programs, and develop personalized treatment approaches. Among the various risk factors, infection with certain oncogenic viruses has emerged as a potential contributor to BC development. Increasing evidence suggests that bovine leukemia virus (BLV) may contribute to zoonotic infections in humans, with a potential role in BC initiation and progression. This review evaluates clinical and experimental data on BLV presence in both malignant and non-malignant breast tissues, exploring potential mechanisms through which BLV may access human breast tissue and contribute to carcinogenesis. Current data reveal a higher prevalence of BLV infection in BC tissues compared to non-tumor tissues, correlating with an increased risk of BC development. In this context, dairy and meat products from BLV-infected animals have been proposed as potential transmission sources. BLV-encoded proteins disrupt key oncogenic pathways, which support their possible role in breast carcinogenesis. However, the interpretation of these findings is limited by potential confounding factors such as genetic predisposition, environmental exposures, and dietary influences. Further research, including well-controlled epidemiological studies, longitudinal cohorts, and mechanistic investigations into BLV proteins in human breast cells, is necessary to determine its role in BC development.
Collapse
Affiliation(s)
- Rancés Blanco
- Independent Researcher, Av. Vicuña Mackenna Poniente 6315, La Florida 8240000, Chile;
| | - Claudio Quezada-Romegialli
- Plataforma de Monitoreo Genómico y Ambiental, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile;
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
2
|
Liao Y, Chen J, Yao H, Zheng T, Tu J, Chen W, Guo Z, Zou Y, Wen L, Xie X. Single-cell profiling of SLC family transporters: uncovering the role of SLC7A1 in osteosarcoma. J Transl Med 2025; 23:103. [PMID: 39844299 PMCID: PMC11752724 DOI: 10.1186/s12967-025-06086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most common malignant bone tumor in children and adolescents, characterized by high disability and mortality rates. Over the past three decades, therapeutic outcomes have plateaued, underscoring the critical need for innovative therapeutic targets. Solute carrier (SLC) family transporters have been implicated in the malignant progression of a variety of tumors, however, their specific role in osteosarcoma remains poorly understood. METHODS The single-cell sequencing data from GSE152048 and GSE162454, along with RNA-seq from the TARGET and GSE21257 cohorts, were utilized for the analysis in this study. LASSO regression analysis was conducted to identify prognostic genes and construct an SLC-related prognostic signature. Survival analysis and ROC analysis evaluated the validity of the prognostic signature. The ESTIMATE and CIBERSORT Packages were utilized to assess the immune infiltration status. Pseudotime and CellChat analyses were performed to investigate the relationship between SLC7A1, malignant phenotypes, and the immune microenvironment. CCK8 assays, EdU staining, colony formation assays, Transwell assays, and co-culture systems were used to assess the effects of SLC7A1 on cell proliferation, metastasis, and macrophage polarization. Finally, virtual docking identified potential drugs targeting SLC7A1. RESULTS SLCs displayed distinct expression patterns across various cell types within the osteosarcoma microenvironment, with myeloid cells exhibiting a preference for amino acid uptake. A prognostic model comprising nine genes was constructed via LASSO regression, with SLC7A1 showing the highest hazard ratio. Multiple analytical algorithms indicated that SLCs were associated with immune cell infiltration and immune checkpoint gene expression. Single-cell analysis indicated that SLC7A1 was predominantly expressed in osteosarcoma cells and correlated with various malignant tumor characteristics. SLC7A1 also regulate interactions between tumor cells and macrophages, as well as modulate macrophage function through multiple pathways. In vitro assays and survival analysis demonstrated that inhibition of SLC7A1 suppressed the malignant phenotype of osteosarcoma cells, with SLC7A1 expression correlating with poor prognosis. Co-culture models confirmed the involvement of SLC7A1 in macrophage polarization. Finally, virtual screening and CETSA identified Cepharanthine as potential inhibitors of SLC7A1. CONCLUSION SLC-related prognostic signatures can be utilized for the prognostic evaluation of osteosarcoma. Pharmacological inhibition of SLC7A1 may be a feasible therapeutic approach for osteosarcoma.
Collapse
Affiliation(s)
- Yan Liao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Hao Yao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Ting Zheng
- The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, 510620, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Weidong Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - ZeHao Guo
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Yutong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| | - Lili Wen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
3
|
Bai M, Liu H, Yan Y, Duan S, Szeto IMY, He J, Hu J, Fu Y, Xu K, Xiong X. Hydrolyzed protein formula improves the nutritional tolerance by increasing intestinal development and altering cecal microbiota in low-birth-weight piglets. Front Nutr 2024; 11:1439110. [PMID: 39555191 PMCID: PMC11565607 DOI: 10.3389/fnut.2024.1439110] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Background Prematurity or low birth weight (LBW), poses a significant challenge in global health. Exploring appropriate and effective nutritional interventions is crucial for the growth and development of LBW infants. Hydrolyzed protein formula has been suggested as a potential solution to prevent intestinal dysfunction and improve digestion and absorption in infants. Objectives This study aimed to investigate the benefits of hydrolyzed protein formula on feeding intolerance, intestinal morphological development, and microbiota in a LBW piglet model. Methods A total of 24 male piglets (3 d of age, 0.95-1.25 kg average BW) were assigned (8 pens/diet; 1 pigs/pen) into three dietary treatments and fed with a basic formula (BF), standard premature infant formula (SF) and hydrolyzed protein formula (HF) respectively, for 7 d. After the piglets sacrifice, growth performance, amino acid metabolism and intestinal morphology were assessed. 16S rRNA sequencing and microbial metabolic phenotypes analyzed the effects of different formula treatments on intestinal flora structure of LBW piglets. Results The HF diet reduced the rates of diarrhea and milk vomiting were reduced by 20.44% (p > 0.05) and 58.44% (p > 0.05), and decreased the crypt depth in the ileum while increasing the ratio of villus height/crypt depth and the mRNA expressions of y+LAT1 and b0,+AT in the ileum (p < 0.05). HF increased the final body weight, serum Thr and essential amino acid contents, and CAT2 and b0,+AT mRNA expressions in ileal mucosa compared with the SF diet (p < 0.05). Microbiota sequencing results showed that the colonic microbial richness indices (Chao1, ACE, and observed species), the diversity indices (Shannon and Simpson), and the phyla Actinobacteriota, unidentified_Bacteria, Acidobacteriota and Actinobacteria, the genus Rubrobacter and RB41 were reduced (p < 0.05) in SF and HF groups. Microbial metabolic phenotypes analysis showed a reduction in the richness of biofilm-forming bacteria (p < 0.05). Conclusion In summary, hydrolyzed protein formula had better nutrition and tolerance in LBW suckling piglets by improving amino acid transport and intestinal development, and regulating gut microbial communities.
Collapse
Affiliation(s)
- Miaomiao Bai
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Hongnan Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Yalu Yan
- Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
| | - Sufang Duan
- Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Jian He
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Jinjie Hu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yawei Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Kang Xu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Hunan, Changsha, China
| | - Xia Xiong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| |
Collapse
|
4
|
You S, Han X, Xu Y, Sui L, Song K, Yao Q. High expression of SLC7A1 in high-grade serous ovarian cancer promotes tumor progression and is involved in MAPK/ERK pathway and EMT. Cancer Med 2024; 13:e7217. [PMID: 38752472 PMCID: PMC11097251 DOI: 10.1002/cam4.7217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024] Open
Abstract
Our previous studies have shown that upregulation of SLC7A1 in epithelial ovarian cancer (EOC) tumor cells significantly increases cancer cell proliferation, migration, and cisplatin resistance; however, the molecular mechanism by which SLC7A1 functions in EOC remains unknown. In later studies, we found that SLC7A1 is also highly expressed in the interstitial portion of high-grade serous ovarian cancer (HGSOC), but the significance of this high expression in the interstitial remains unclear. Here, we showed the Interstitial high expression of SLC7A1 in HGSOC by immunohistochemistry. SLC7A1 enriched in cancer-associated fibroblasts (CAFs) was upregulated by TGF-β1. Transwell assay, scratch assay, cck8 assay and cell adhesion assay showed that SLC7A1 highly expressed in CAFs promoted tumor cells invasion, migration and metastasis in vitro. The effect of SLC7A1 on MAPK and EMT pathway proteins in ovarian cancer (OC) was verified by RNA sequencing and western blotting. Overexpression of SLC7A1 in OC is involved in MAPK/ ERK pathway and EMT. In general, in HGSOC, CAFs overexpressing SLC7A1 supported the migration and invasion of tumor cells; SLC7A1 is highly expressed in ovarian cancer and is involved in ERK phosphorylation and EMT signaling in MAPK signaling pathway. This suggests that SLC7A1 may be a potential therapeutic target for OC metastasis.
Collapse
Affiliation(s)
- Shijing You
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Xiahui Han
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Yuance Xu
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Lei Sui
- Department of Gynecological OncologyAffiliated Qingdao Central Hospital of Qingdao UniversityQingdaoShandongChina
| | - Kejuan Song
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Qin Yao
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
5
|
Jakobsen S, Nielsen CU. Exploring Amino Acid Transporters as Therapeutic Targets for Cancer: An Examination of Inhibitor Structures, Selectivity Issues, and Discovery Approaches. Pharmaceutics 2024; 16:197. [PMID: 38399253 PMCID: PMC10893028 DOI: 10.3390/pharmaceutics16020197] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Amino acid transporters are abundant amongst the solute carrier family and have an important role in facilitating the transfer of amino acids across cell membranes. Because of their impact on cell nutrient distribution, they also appear to have an important role in the growth and development of cancer. Naturally, this has made amino acid transporters a novel target of interest for the development of new anticancer drugs. Many attempts have been made to develop inhibitors of amino acid transporters to slow down cancer cell growth, and some have even reached clinical trials. The purpose of this review is to help organize the available information on the efforts to discover amino acid transporter inhibitors by focusing on the amino acid transporters ASCT2 (SLC1A5), LAT1 (SLC7A5), xCT (SLC7A11), SNAT1 (SLC38A1), SNAT2 (SLC38A2), and PAT1 (SLC36A1). We discuss the function of the transporters, their implication in cancer, their known inhibitors, issues regarding selective inhibitors, and the efforts and strategies of discovering inhibitors. The goal is to encourage researchers to continue the search and development within the field of cancer treatment research targeting amino acid transporters.
Collapse
Affiliation(s)
- Sebastian Jakobsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| |
Collapse
|
6
|
Shen X, Wang G, He H, Shang P, Yan B, Wang X, Shen W. SLC38A5 promotes glutamine metabolism and inhibits cisplatin chemosensitivity in breast cancer. Breast Cancer 2024; 31:96-104. [PMID: 37914960 DOI: 10.1007/s12282-023-01516-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/15/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Solute carrier family 38 member 5 (SLC38A5), as an amino acid transporter, play a vital role in cellular biological processes. In this study, we analyzed the function of SLC38A5 and its potential mechanism in breast cancer (BC) progression. METHODS The expression of SLC38A5 in cancer and adjacent-normal tissues was analyzed by qRT-PCR and Western blot, and its correlation with patient prognosis was analyzed. The immunohistochemical staining of cancer tissues and adjacent-normal tissues was performed on SLC38A5-positive specimens. BC mice were successfully applied to examine the role of SLC38A5 on tumor proliferation using the CCK-8 assay. In BC cells and mouse tumor tissues, SLC38A5 and PCNA expression were determined by Western blotting. RESULTS The study found that SLC38A5 was highly expressed in BC patients and associated with a poor survival. SLC38A5 silencing inhibited BC cell viability and glutamine uptake. In addition, SLC38A5 overexpression promoted BC cell viability via the glutamine metabolism. SLC38A5 inhibited cisplatin chemosensitivity in BC cells. Importantly, SLC38A5 silencing inhibited tumor growth in vivo. CONCLUSION Our findings suggest that SLC38A5 enhances BC cell viability by glutamine metabolism, inhibits the chemical sensitivity of cisplatin in BC cells, and promotes tumor growth, emphasizing the clinical relevance of SLC38A5 in BC management as a novel potential therapeutic target.
Collapse
Affiliation(s)
- Xiaowei Shen
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, 201700, China.
| | - Ganggang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hua He
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, 201700, China
| | - Ping Shang
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, 201700, China
| | - Bin Yan
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, 201700, China
| | - Xiaoliang Wang
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, 201700, China
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Weixing Shen
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, 201700, China
| |
Collapse
|