1
|
Zhu T, Liu Y, Du J, Lei C, Wang C, Li S, Song H. Effects of short-term salt exposure on gill damage, serum components and gene expression patterns in juvenile Largemouth bass (Micropterus salmoides). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 53:101365. [PMID: 39603077 DOI: 10.1016/j.cbd.2024.101365] [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: 07/21/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
The Largemouth bass (Micropterus salmoides; LMB) is a freshwater fish that plays a significant role in aquaculture, and its cultural base is expanding into inland saline water areas. To study the effect of short-term salt exposure on LMB, fish with an average body weight of 11.69 (±1.82) g were cultured for 14 days at three different salt concentrations (0 ‰, 6 ‰, and 12 ‰). After 14 days, the second gill arch was collected for tissue sectioning and transcriptome sequencing, while serum samples were collected to analyze serum components. The results showed that the mortality rate in the 0 ‰ and 6 ‰ groups was 0 %, whereas the mortality rate in the 12 ‰ group was 62 %. In the gill tissue sections, no apparent damage was observed in the 0 ‰ and 6 ‰ groups. However, in the 12 ‰ group, the secondary lamellae became shorter, thicker, and exhibited a disordered arrangement. The serum component test results showed that osmolality and K+ significantly increased in the 12 ‰ group, while Na+, K+, and Cl- concentrations showed slight increases, but the differences were not significant. Comparative transcriptome analysis revealed that, along the salinity gradient, gene expression exhibited five profiles. Genes related to ion transport and immunity were highly expressed in the 6 ‰ and 12 ‰ groups, while genes associated with biosynthesis and ATP production showed decreased expression levels as salinity increased. Notably, seven solute carrier genes, two Na+/K+-ATPase genes, and two insulin-like growth factor genes were significantly highly expressed in the 12 ‰ salinity group, playing important roles in the transmembrane transport of ions. Based on the results, the LMB can acclimatize to a salt concentration of at least 6 ‰. However, exposure to 12 ‰ salinity can lead to a series of adverse effects, including organ damage, reduced energy metabolism efficiency, and disruption of ion homeostasis.
Collapse
Affiliation(s)
- Tao Zhu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Yang Liu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Jinxing Du
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China
| | - Caixia Lei
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China
| | - Chenghui Wang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Shengjie Li
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China
| | - Hongmei Song
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China
| |
Collapse
|
2
|
Abrahams-October Z, Kippie Y, Pearce K, Johnson R, Benjeddou M. Effects of Xhosa specific solute carrier family 22-member 2 haplotypes on the cellular uptake of metformin and cimetidine. Gene 2025; 937:149157. [PMID: 39653092 DOI: 10.1016/j.gene.2024.149157] [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: 07/18/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Studies have shown that solute carrier transporters play an important role in the transport and distribution of metformin, and that genetic variation(s) in solute carrier genes have play a role in the variation of metformin efficacy and disposition observed in populations. This study aimed to determine the cellular uptake efficiency of metformin in SLC22A2 coding haplotypes of an indigenous South African population. METHODS AND RESULTS To determine metformin and cimetidine cellular uptake in transfected HEK-293 cells, ultra high-performance liquid chromatography was used to quantitate substrate concentration(s). Haplotypes 3 and 4 showed decreased metformin uptake, and haplotypes 2 and 5 displayed increased metformin uptake in comparison to haplotype 1 (i.e. wildtype haplotype). Haplotypes 2-5 showed decreased uptake of cimetidine in comparison to haplotype 1, implying a reduced sensitivity to the inhibition of cimetidine. In all haplotypes, no significant transport was observed for metformin and cimetidine. Passive permeability of metformin was favoured in haplotypes 3 and 5, whilst the remaining haplotypes demonstrate higher passive permeability ratios in favour of cimetidine. CONCLUSION Haplotype 4, which is characterised by the non-synonymous single nucleotide polymorphisms rs316019 and rs8177517, demonstrates potential impaired metformin transport.
Collapse
Affiliation(s)
- Zainonesa Abrahams-October
- Precision Medicine Unit, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville 7535, South Africa
| | - Yunus Kippie
- School of Pharmacy, University of the Western Cape, Robert Sobukwe Road, Bellville 7535, South Africa
| | - Keenau Pearce
- Precision Medicine Unit, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville 7535, South Africa.
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Francie Van Zijl Drive, Parow Valley, 7501 Cape Town, South Africa; Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Mongi Benjeddou
- Precision Medicine Unit, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville 7535, South Africa
| |
Collapse
|
3
|
Kędzia K, Szmajda-Krygier D, Krygier A, Jabłoński S, Balcerczak E, Wcisło S. Altered carnitine transporter genes ( SLC22A5, SLC22A16, SLC6A14) expression pattern among lung cancer patients. Transl Lung Cancer Res 2024; 13:2903-2917. [PMID: 39670016 PMCID: PMC11632432 DOI: 10.21037/tlcr-24-448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/20/2024] [Indexed: 12/14/2024]
Abstract
Background Despite the decrease of morbidity rate of non-small cell lung cancer (NSCLC) in recent years, it is still a cancer with poor prognosis. Lung cancers (LCs) are usually diagnosed at a late stage of the disease due to non-specific clinical symptoms. Proper regulation of carnitine levels is important in the context of development and increased risk of cancer cells proliferation. The expression profiles and clinical value of SLC family members in LC remain largely unexplored. The aim of the study was the assessment of SLC22A16, SLC22A5 and SLC6A14 mRNA expression level among patients suffering from NSCLC. The obtained results were compared with the clinical and the pathological features of NSCLC patients. Methods Through reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and bioinformatics studies, the evaluation of carnitine transporting genes (SLC22A16, SLC22A5 and SLC6A14) mRNA levels was performed in order to elucidate their connection to clinical features of patients and influence on overall survival (OS). Results The analysis showed a significant difference for the SLC22A5 gene of NSCLC patients and for SLC6A14 and SLC22A5 genes in LUSC patients in terms of sex (P=0.002, P=0.02 and P=0.001, respectively) and in terms of tobacco smoking (P=0.04). Analysis also revealed a significant negative correlation for SLC22A5 and SLC22A16 genes expression level in the lung adenocarcinoma (LUAD) subtype with standardized uptake value (SUV) (r=-0.40, P=0.02 and r=-0.43, P=0.04). The significant downregulation of gene expression compared to normal adjacent tissue was observed for SLC22A5 in lung squamous cell carcinoma (LUSC) and for SLC6A14 in both LUAD and LUSC subtypes. The effect of the SLC22A5, SLC22A16 and SLC6A14 gene expression at the time of diagnosis on the OS time of LC patients revealed that lower expression correlated with a shorter 5 years OS (all P values <0.01). The effects were distinct after division for LUAD and LUSC subtypes. Conclusions The expression levels of genes encoding carnitine transporters are diverse, hinting at a potentially altered carnitine metabolism in LC patients. Notably, this variance is not uniform and exhibits specificity across LC subtypes, with marked distinctions between LUAD and LUSC. The correlation between gene expression levels and OS of patients underlines the prognostic significance of SLC genes within these cancer subtypes.
Collapse
Affiliation(s)
- Konrad Kędzia
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz and Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz-Central Veteran Hospital, Lodz, Poland
| | - Dagmara Szmajda-Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Lodz, Poland
| | - Adrian Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Lodz, Poland
| | - Sławomir Jabłoński
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz and Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz-Central Veteran Hospital, Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Lodz, Poland
| | - Szymon Wcisło
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz and Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz-Central Veteran Hospital, Lodz, Poland
| |
Collapse
|
4
|
Ajmeriya S, Kashyap N, Gul A, Ahirwar A, Singh S, Tripathi S, Dhar R, Nayak NR, Karmakar S. Aberrant expression of solute carrier family transporters in placentas associated with pregnancy complications. Placenta 2024; 159:9-19. [PMID: 39602836 DOI: 10.1016/j.placenta.2024.11.011] [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: 06/26/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Solute carrier family transporters (SLCs), crucial for nutrient and trace element uptake in the placenta, play a significant role in fetal growth and development. Their dysregulation is associated with various pregnancy disorders. However, a comprehensive understanding of their role and regulation in placental function and pregnancy complications is still a largely unexplored area, making this study novel and significant. METHODS We performed a rigorous meta-analysis of publicly available NCBI GEO microarray and RNA-Seq datasets followed by bioinformatics analysis of differentially expressed SLCs in PE and IUGR. The identified SLCs were then validated using qPCR on PE placental samples, ensuring the reliability and validity of the findings. RESULTS Bioinformatics analysis of preeclampsia (PE) and Intrauterine Growth restriction (IUGR) datasets revealed significant associations between specific SLC transporters with disease pathology, identified by studying differentially expressed SLCs. Subsequent validation using qPCR on placental samples confirmed considerable downregulation of SLC6A8, SLC16A10, SLC25A3, and SLC29A3, highlighting their dysregulation in the pathogenesis of PE and IUGR. DISCUSSION The significant downregulation of SLC6A8, SLC16A10, SLC25A3, and SLC29A3 observed by bioinformatics analyses and validated by qPCR indicates atypical expression of these SLCs in gestational disorders. Our findings underscore the potential contribution of multiple SLC gene families to the development of placental pathologies associated with diverse pregnancy complications.
Collapse
Affiliation(s)
- Swati Ajmeriya
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Neha Kashyap
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Anamta Gul
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Ashok Ahirwar
- Department of Laboratory Medicine, AIl India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Smita Tripathi
- Department of Biochemistry, Lady Harding Medical College, New Delhi, 110029, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Nihar R Nayak
- Department of Obstetrics and Gynecology, University of Missouri, Kansas City, USA
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
5
|
Li L, Fan B, Zhang Y, Zhao M, Kong Z, Wang F, Li M. Cannabidiol exposure during embryonic period caused serious malformation in embryos and inhibited the development of reproductive system in adult zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175315. [PMID: 39111451 DOI: 10.1016/j.scitotenv.2024.175315] [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: 04/08/2024] [Revised: 07/17/2024] [Accepted: 08/04/2024] [Indexed: 08/11/2024]
Abstract
Cannabidiol (CBD) is a non-psychoactive component of cannabis with potential applications in biomedicine, food, and cosmetics due to its analgesic, anti-inflammatory, and anticonvulsant properties. However, increasing reports of adverse CBD exposure events underscore the necessity of evaluating its toxicity. In this study, we investigated the developmental toxicity of CBD in zebrafish during the embryonic (0-4 dpf, days post fertilization) and early larval stages (5-7 dpf). The median lethal concentration of CBD in embryos/larvae is 793.28 μg/L. CBD exhibited concentration-dependent manner (ranging from 250 to 1500 μg/L) in inducing serious malformed somatotypes, like shorter body length, pericardial cysts, vitelline cysts, spinal curvature, and smaller eyes. However, no singular deformity predominates. The 5-month-old zebrafish treated with 100 and 200 μg/L of CBD during the embryonic and early larval stages produced fewer offspring with higher natural mortality and malformation rate. Gonadal growth and gamete development were inhibited. Transcriptomic and metabolomic analyses conducted with 400 μg/L CBD on embryos/larvae from 0 to 5 dpf suggested that CBD promoted the formation and transportation of extracellular matrix components on 1 dpf, promoting abnormal cell division and migration, probably resulting in random malformed somatotypes. It inhibited optical vesicle development and photoreceptors formation on 2 and 3 dpf, resulting in damaged sight and smaller eye size. CBD also induced an integrated stress response on 4 and 5 dpf, disrupting redox, protein, and cholesterol homeostasis, contributing to cellular damage, physiological dysfunction, embryonic death, and inhibited reproductive system and ability in adult zebrafish. At the tested concentrations, CBD exhibited developmental toxicity, lethal toxicity, and reproductive inhibition in zebrafish. These findings demonstrate that CBD threatens the model aquatic animal, highlighting the need for additional toxicological evaluations of CBD before its inclusion in dietary supplements, edible food, and other products.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Agro-products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs/Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Bei Fan
- Key Laboratory of Agro-products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs/Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China; Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, PR China
| | - Yifan Zhang
- Key Laboratory of Agro-products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs/Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Mengying Zhao
- Key Laboratory of Agro-products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs/Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Zhiqiang Kong
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Fengzhong Wang
- Key Laboratory of Agro-products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs/Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| | - Minmin Li
- Key Laboratory of Agro-products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs/Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China; Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, PR China.
| |
Collapse
|
6
|
Kamynina M, Rozenberg JM, Kushchenko AS, Dmitriev SE, Modestov A, Kamashev D, Gaifullin N, Shaban N, Suntsova M, Emelianova A, Buzdin AA. Forced Overexpression and Knockout Analysis of SLC30A and SLC39A Family Genes Suggests Their Involvement in Establishing Resistance to Cisplatin in Human Cancer Cells. Int J Mol Sci 2024; 25:12049. [PMID: 39596116 PMCID: PMC11594112 DOI: 10.3390/ijms252212049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The metabolism of zinc and manganese plays a pivotal role in cancer progression by mediating cancer cell growth and metastasis. The SLC30A family proteins SLC30A3 and SLC30A10 mediate the efflux of zinc, manganese, and probably other transition element ions outside the cytoplasm to the extracellular space or into intracellular membrane compartments. The SLC39A family members SLC39A8 and SLC39A14 are their functional antagonists that transfer these ions into the cytoplasm. Recently, the SLC30A10 gene was suggested as a promising methylation biomarker of colorectal cancer. Here, we investigated whether forced overexpression or inactivation of SLC30A and SLC39A family genes has an impact on the phenotype of cancer cells and their sensitivity to cancer therapeutics. In the human colon adenocarcinoma HCT-15 and duodenal adenocarcinoma HuTu80 cell lines, we generated clones with knockouts of the SLC39A8 and SLC39A14 genes and forced overexpression of the SLC30A3, SLC30A10, and SLC39A8 genes. Gene expression in the mutant and control cells was assessed by RNA sequencing. The cell growth rate, mitochondrial activity, zinc accumulation, and sensitivity to the drugs cetuximab and cisplatin were investigated in functional tests. Overexpression or depletion of SLC30A or SLC39A family genes resulted in the deep reshaping of intracellular signaling and provoked hyperactivation of mitochondrial respiration. Variation in the expression of the SLC30A/SLC39A genes did not increase the sensitivity to cetuximab but significantly altered the sensitivity to cisplatin: overexpression of SLC30A10 resulted in an ~2.7-4 times increased IC50 of cisplatin, and overexpression of SLC30A3 resulted in an ~3.3 times decreased IC50 of cisplatin. The SLC30A/SLC39A genes should be considered as potential cancer drug resistance biomarkers and putative therapeutic targets.
Collapse
Affiliation(s)
- Margarita Kamynina
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
| | | | - Artem S. Kushchenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.S.K.); (S.E.D.)
| | - Sergey E. Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.S.K.); (S.E.D.)
| | - Aleksander Modestov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
| | - Dmitry Kamashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Nurshat Gaifullin
- Faculty of Fundamental Medicine, Moscow State University, 119992 Moscow, Russia;
| | - Nina Shaban
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
| | - Maria Suntsova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
| | - Anna Emelianova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
| | - Anton A. Buzdin
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.M.); (D.K.); (N.S.); (M.S.); (A.E.); (A.A.B.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
7
|
Parida KK, Lahiri M, Ghosh M, Dalal A, Kalia NP. P-glycoprotein inhibitors as an adjunct therapy for TB. Drug Discov Today 2024; 29:104108. [PMID: 39032811 DOI: 10.1016/j.drudis.2024.104108] [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: 03/21/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
The primary challenge in TB treatment is the emergence of multidrug-resistant TB (MDR-TB). One of the major factors responsible for MDR is the upregulation of efflux pumps. Permeation-glycoprotein (P-gp), an efflux pump, hinders the bioavailability of the administered drugs inside the infected cells. Simultaneously, angiogenesis, the formation of new blood vessels, contributes to drug delivery complexities. TB infection triggers a cascade of events that upregulates the expression of angiogenic factors and P-gp. The combined action of P-gp and angiogenesis foster the emergence of MDR-TB. Understanding these mechanisms is pivotal for developing targeted interventions to overcome MDR in TB. P-gp inhibitors, such as verapamil, and anti-angiogenic drugs, including bevacizumab, have shown improvement in TB drug delivery to granuloma. In this review, we discuss the potential of P-gp inhibitors as an adjunct therapy to shorten TB treatment.
Collapse
Affiliation(s)
- Kishan Kumar Parida
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Monali Lahiri
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Mainak Ghosh
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Aman Dalal
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Nitin Pal Kalia
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
8
|
Simon-Molas H, Montironi C, Kabanova A, Eldering E. Metabolic reprogramming in the CLL TME; potential for new therapeutic targets. Semin Hematol 2024; 61:155-162. [PMID: 38493076 DOI: 10.1053/j.seminhematol.2024.02.001] [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: 11/11/2023] [Revised: 01/28/2024] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
Chronic lymphocytic leukemia (CLL) cells circulate between peripheral (PB) blood and lymph node (LN) compartments, and strictly depend on microenvironmental factors for proliferation, survival and drug resistance. All cancer cells display metabolic reprogramming and CLL is no exception - though the inert status of the PB CLL cells has hampered detailed insight into these processes. We summarize previous work on reactive oxygen species (ROS), oxidative stress, and hypoxia, as well as the important roles of Myc, and PI3K/Akt/mTor pathways. In vitro co-culture systems and gene expression analyses have provided a partial picture of CLL LN metabolism. New broad omics techniques allow to obtain molecular and also single-cell level understanding of CLL plasticity and metabolic reprogramming. We summarize recent developments and describe the new concept of glutamine addiction for CLL, which may hold therapeutic promise.
Collapse
Affiliation(s)
- Helga Simon-Molas
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Chiara Montironi
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Anna Kabanova
- Tumour Immunology Unit, Toscana Life Sciences Foundation, Siena, Italy
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Montjean D, Beaumont M, Natiq A, Louanjli N, Hazout A, Miron P, Liehr T, Cabry R, Ratbi I, Benkhalifa M. Genome and Epigenome Disorders and Male Infertility: Feedback from 15 Years of Clinical and Research Experience. Genes (Basel) 2024; 15:377. [PMID: 38540436 PMCID: PMC10970370 DOI: 10.3390/genes15030377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 06/14/2024] Open
Abstract
Infertility affects around 20% of couples of reproductive age; however, in some societies, as many as one-third of couples are unable to conceive. Different factors contribute to the decline of male fertility, such us environmental and professional exposure to endocrine disruptors, oxidative stress, and life habits with the risk of de novo epigenetics dysregulation. Since the fantastic development of new "omes and omics" technologies, the contribution of inherited or de novo genomes and epigenome disorders to male infertility have been further elucidated. Many other techniques have become available to andrology laboratories for the investigation of genome and epigenome integrity and the maturation and the competency of spermatozoa. All these new methods of assessment are highlighting the importance of genetics and epigenetics investigation for assisted reproduction pathology and for supporting professionals in counselling patients and proposing different management strategies for male infertility. This aims to improve clinical outcomes while minimizing the risk of genetics or health problems at birth.
Collapse
Affiliation(s)
- Debbie Montjean
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
| | - Marion Beaumont
- Genetics Department, Eylau/Unilabs Laboratory, 92110 Clichy, France;
| | - Abdelhafid Natiq
- Center for Genomics of Human Pathologies (GENOPATH), Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco (I.R.)
- National Laboratory Mohammed VI, Mohammed VI Foundation of Casablanca, Casablanca, Morocco
| | | | - Andre Hazout
- Andro-Genetics Unit, Labomac, Casablanca, Morocco (A.H.)
| | - Pierre Miron
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
| | - Thomas Liehr
- Institute für Humangenetik, Universitätsklinikum Jena, Friedrich Schiller Universität, 07743 Jena, Germany
| | - Rosalie Cabry
- Reproductive Medicine, Reproductive Biology & Genetics, CECOS Picardie, University Hospital & School of Medicine, Picardie University Jules Verne, 80000 Amiens, France
- PeriTox Laboratory, Perinatality & Toxic Risks, UMR-I 01 INERIS, Picardie University Jules Verne, 80000 Amiens, France
| | - Ilham Ratbi
- Center for Genomics of Human Pathologies (GENOPATH), Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco (I.R.)
- Medical Genetics Unit, Ibn Sina University Hospital Center, Rabat, Morocco
| | - Moncef Benkhalifa
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
- Reproductive Medicine, Reproductive Biology & Genetics, CECOS Picardie, University Hospital & School of Medicine, Picardie University Jules Verne, 80000 Amiens, France
- PeriTox Laboratory, Perinatality & Toxic Risks, UMR-I 01 INERIS, Picardie University Jules Verne, 80000 Amiens, France
| |
Collapse
|
10
|
Cheng F, Dennis AB, Baumann O, Kirschbaum F, Abdelilah-Seyfried S, Tiedemann R. Gene and Allele-Specific Expression Underlying the Electric Signal Divergence in African Weakly Electric Fish. Mol Biol Evol 2024; 41:msae021. [PMID: 38410843 PMCID: PMC10897887 DOI: 10.1093/molbev/msae021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
In the African weakly electric fish genus Campylomormyrus, electric organ discharge signals are strikingly different in shape and duration among closely related species, contribute to prezygotic isolation, and may have triggered an adaptive radiation. We performed mRNA sequencing on electric organs and skeletal muscles (from which the electric organs derive) from 3 species with short (0.4 ms), medium (5 ms), and long (40 ms) electric organ discharges and 2 different cross-species hybrids. We identified 1,444 upregulated genes in electric organ shared by all 5 species/hybrid cohorts, rendering them candidate genes for electric organ-specific properties in Campylomormyrus. We further identified several candidate genes, including KCNJ2 and KLF5, and their upregulation may contribute to increased electric organ discharge duration. Hybrids between a short (Campylomormyrus compressirostris) and a long (Campylomormyrus rhynchophorus) discharging species exhibit electric organ discharges of intermediate duration and showed imbalanced expression of KCNJ2 alleles, pointing toward a cis-regulatory difference at this locus, relative to electric organ discharge duration. KLF5 is a transcription factor potentially balancing potassium channel gene expression, a crucial process for the formation of an electric organ discharge. Unraveling the genetic basis of the species-specific modulation of the electric organ discharge in Campylomormyrus is crucial for understanding the adaptive radiation of this emerging model taxon of ecological (perhaps even sympatric) speciation.
Collapse
Affiliation(s)
- Feng Cheng
- Unit of Evolutionary Biology/Systematic Zoology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Alice B Dennis
- Unit of Evolutionary Biology/Systematic Zoology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
- Laboratory of Adaptive Evolution and Genomics, Research Unit of Environmental and Evolutionary Biology, Institute of Life, Earth & Environment, University of Namur, Namur, Belgium
| | - Otto Baumann
- Department of Animal Physiology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Frank Kirschbaum
- Unit of Evolutionary Biology/Systematic Zoology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
- Department of Crop and Animal Science, Faculty of Life Sciences, Humboldt University, Berlin, Germany
| | - Salim Abdelilah-Seyfried
- Department of Animal Physiology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Ralph Tiedemann
- Unit of Evolutionary Biology/Systematic Zoology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
11
|
Zheng Q, Li M, Qiu Y, Yang J, Cao Y. Overexpression of SLC35F2 is a potential prognostic biomarker for lung adenocarcinoma. Heliyon 2024; 10:e23828. [PMID: 38187235 PMCID: PMC10767229 DOI: 10.1016/j.heliyon.2023.e23828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024] Open
Abstract
Objective To explore the potential clinical and prognostic significance of Homo sapiens solute carrier family 35 member F2 (SLC35F2) in the context of lung adenocarcinoma (LUAD). Methods The expression pattern of SLC35F2 in LUAD tissues and normal tissues was analyzed in The Cancer Genome Atlas (TCGA) datasets and validated in 12 pairs of fresh clinical LUAD tissues and their corresponding adjacent normal tissues using quantitative real-time PCR (qRT-PCR) and western blotting. Immunohistochemistry (IHC) was used to assess the protein expression of SLC35F2 in 60 paraffin-embedded LUAD tissues, and its associations with clinicopathological parameters were further examined. The prognostic significance of SLC35F2 mRNA expression was also evaluated using the Kaplan-Meier method, and Cox regression models in LUAD patients from the TCGA database. The potential utility of SLC35F2 as an indicator of recurrence or metastasis was explored through the follow-up of selected clinical LUAD cases. Lastly, gene set enrichment analysis (GSEA) was conducted to investigate the underlying biological mechanisms and signaling pathways. Results Bioinformatics analysis utilizing the TCGA database indicated that SLC35F2 mRNA exhibited heightened expression in LUAD tissues when compared to normal tissues. These findings were further substantiated through the examination of 12 pairs of clinical LUAD tissues and their corresponding adjacent normal tissues, employing qRT-PCR and western blotting techniques. IHC results from a cohort of 60 LUAD patients demonstrated an up-regulation of SLC35F2 in 38 out of 60 individuals (63.3 %), which exhibited a significant correlation with tumor size, lymph node metastasis, and clinical stage (all P < 0.05). Both the Kaplan-Meier curve and the Cox proportional hazard analyses indicated a strong association between the up-regulation of SLC35F2 mRNA expression and unfavorable overall survival (OS) in patients with LUAD, as observed in the TCGA datasets (P < 0.05). The follow-up findings from select clinical LUAD cases provided evidence that the expression of SLC35F2 could serve as a dependable biomarker for monitoring the recurrence or metastasis. Additionally, the GSEA highlighted the enrichment of apoptosis, adhesion, small cell lung cancer (SCLC), and p53 signaling pathways in the subgroup of LUAD patients with elevated SLC35F2 expression. Conclusion SLC35F2 exhibited an up-regulated in both mRNA and protein expression, rendering it a valuable independent prognostic indicator for patients diagnosed with LUAD.
Collapse
Affiliation(s)
- Qingzhu Zheng
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mingjie Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yingkun Qiu
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jiahao Yang
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, 350004, China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| |
Collapse
|
12
|
Lu S, Sun X, Tang H, Yu J, Wang B, Xiao R, Qu J, Sun F, Deng Z, Li C, Yang P, Yang Z, Rao B. Colorectal cancer with low SLC35A3 is associated with immune infiltrates and poor prognosis. Sci Rep 2024; 14:329. [PMID: 38172565 PMCID: PMC10764849 DOI: 10.1038/s41598-023-51028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
The expression level of SLC35A3 is associated with the prognosis of many cancers, but its role in colorectal cancer (CRC) is unclear. The purpose of our study was to elucidate the role of SLC35A3 in CRC. The expression levels of SLC35A3 in CRC were evaluated through tumor immune resource assessment (TIMER), The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), International Cancer Genome Consortium (ICGC), Human Protein Atlas (HPA), qRT-PCR, and immunohistochemical evaluation. TCGA, GEO, and ICGC databases were used to analyze the diagnostic and prognostic value of SLC35A3 in CRC. A overall survival (OS) model was constructed and validated based on the expression level of SLC35A3 and multivariable analysis results. The cBioPortal tool was used to analyze SLC35A3 mutation in CRC. The UALCAN tool was used to analyze the promoter methylation level of SLC35A3 in colorectal cancer. In addition, the role of SLC35A3 in CRC was determined through GO analysis, KEGG analysis, gene set enrichment analysis (GSEA), immune infiltration analysis, and immune checkpoint correlation analysis. In vitro experiments validated the function of SLC35A3 in colorectal cancer cells. Compared with adjacent normal tissues and colonic epithelial cells, the expression of SLC35A3 was decreased in CRC tissues and CRC cell lines. Low expression of SLC35A3 was associated with N stage, pathological stage, and lymphatic infiltration, and it was unfavorable for OS, disease-specific survival (DSS), recurrence-free survival (RFS), and post-progression survival (PPS). According to the Receiver Operating Characteristic (ROC) analysis, SLC35A3 is a potential important diagnostic biomarker for CRC patients. The nomograph based on the expression level of SLC35A3 showed a better predictive model for OS than single prognostic factors and TNM staging. SLC35A3 has multiple types of mutations in CRC, and its promoter methylation level is significantly decreased. GO and KEGG analysis indicated that SLC35A3 may be involved in transmembrane transport protein activity, cell communication, and interaction with neurotransmitter receptors. GSEA revealed that SLC35A3 may be involved in energy metabolism, DNA repair, and cancer pathways. In addition, SLC35A3 was closely related to immune cell infiltration and immune checkpoint expression. Immunohistochemistry confirmed the positive correlation between SLC35A3 and helper T cell infiltration. In vitro experiments showed that overexpression of SLC35A3 inhibited the proliferation and invasion capability of colorectal cancer cells and promoted apoptosis. The results of this study indicate that decreased expression of SLC35A3 is closely associated with poor prognosis and immune cell infiltration in colorectal cancer, and it can serve as a promising independent prognostic biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Shuai Lu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Beijing, 100038, China
| | - Xibo Sun
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Shandong, 271000, China
| | - Huazhen Tang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Beijing, 100038, China
| | - Jinxuan Yu
- Zibo Central Hospital Affiliated to Binzhou Medical College, Zibo, 255020, China
| | - Bing Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Beijing, 100038, China
| | - Ruixue Xiao
- Inner Mongolia Medical University, Hohhot, 010100, China
| | - Jinxiu Qu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Beijing, 100038, China
| | - Fang Sun
- The Fifth Medical Center of the General Hospital of the People's Liberation Army of China, Beijing, 100000, China
| | - Zhuoya Deng
- The First Medical Center of Chinese, PLA General Hospital, Beijing, 100000, China
| | - Cong Li
- The First Medical Center of Chinese, PLA General Hospital, Beijing, 100000, China
| | - Penghui Yang
- The First Medical Center of Chinese, PLA General Hospital, Beijing, 100000, China.
| | - Zhenpeng Yang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Benqiang Rao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Beijing, 100038, China.
| |
Collapse
|
13
|
Rios ACH, Nasner SLC, Londoño-Gil M, Gonzalez-Herrera LG, Lopez-Herrera A, Flórez JCR. Genome-wide association study for reproduction traits in Colombian Creole Blanco Orejinegro cattle. Trop Anim Health Prod 2023; 55:429. [PMID: 38044379 DOI: 10.1007/s11250-023-03847-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
The profitability of the beef cattle production system relies heavily on reproductive traits. Unfortunately, certain traits, such as age at first calving (AFC), calving interval (CI), and gestation length (GL), can pose challenges in traditional breeding programs because of their low heritability (0.01-0.12) and sex-limited characteristics. Another important aspect is the conservation of the genetic resources of animals adapted to the Colombian regions, which implies the preservation and rational use of the creole breeds in the country market. Therefore, this study aimed to identify genomic regions in the creole cattle breed Blanco Orejinegro (BON) that influence the reproductive traits in females. The dataset comprised 439 animals and 118,116 single-nucleotide polymorphisms' (SNPs) markers. The GS3 program was used to identify the SNP effects employing the BAYES Cπ methodology. The number of SNPs with effect for AFC was 25, 1527 for CI, and 23 for GL. Some of the genes found associated with reproductive and growth traits as well as immune response and environmental adaptation ECE1, EPH, EPHB2, SMARCAL1, IGFBP5, IGFBP2, FCGRT, EGFR, MUL1, PINK1, STPG1, CNGB1, TGFB1, OXTR, IL22RA1, MYOM3, OXTR, CNR2, HIVEP3, CTPS1, CXCL8, FCGRT, MREG, TMEM169, PECR, and MC1R. Our results evidenced a high contribution of the genetic architecture of the Colombian creole cattle breed Blanco Orejinegro that may impact should be included in implementing genetic improvement and conservation programs.
Collapse
Affiliation(s)
- Ana Cristina Herrera Rios
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia.
- Grupo de Investigación Nutri-Solla, SOLLA S.A., Cra 42 #33-80, Itagüí, Antioquia, Colombia.
| | - Sindy Liliana Caivio Nasner
- Grupo de Investigación Biomolecular y Pecuaria BIOPEC, Universidad Tecnológica de Pereira, Cra. 27 N10-02, 660003, Pereira, Risaralda, Colombia
| | - Marisol Londoño-Gil
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
| | - Luis Gabriel Gonzalez-Herrera
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
| | - Albeiro Lopez-Herrera
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
| | - Juan Carlos Rincón Flórez
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Palmira, Carrera 32 N 12 - 00, PC 763352, Palmira, Colombia
| |
Collapse
|
14
|
Colaco JC, Chandrasekaran AP, Karapurkar JK, Birappa G, Rajkumar S, Gowda DAA, Suresh B, Lee J, Singh V, Hong SH, Kim KS, Ramakrishna S. βTrCP1 promotes SLC35F2 protein ubiquitination and inhibits cancer progression in HeLa cells. Biochem Biophys Res Commun 2023; 682:27-38. [PMID: 37801987 DOI: 10.1016/j.bbrc.2023.09.095] [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: 07/13/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/08/2023]
Abstract
The solute carrier family 35 F2 (SLC35F2) belongs to membrane-bound carrier proteins that are associated with multiple cancers. The main factor that determines cancer progression is the expression level of SLC35F2. Thus, identifying the E3 ligase that controls SLC35F2 protein abundance in cancer cells is critical. Here, we identified βTrCP1 interacting with and reducing the SLC35F2 protein level. βTrCP1 signals SLC35F2 protein ubiquitination and reduces SLC35F2 protein half-life. The mRNA expression pattern between βTrCP1 and SLC35F2 across a panel of cancer cell lines showed a negative correlation. Additionally, the depletion of βTrCP1 accumulated SLC35F2 protein and promoted SLC35F2-mediated cell growth, migration, invasion, and colony formation ability in HeLa cells. Overall, we demonstrate that βTrCP1 acts as a tumor suppressor by controlling SLC35F2 protein abundance in cancer cells. The depletion of βTrCP1 promotes SLC35F2-mediated carcinogenesis. Thus, we envision that βTrCP1 may be a potential target for cancer therapeutics.
Collapse
Affiliation(s)
- Jencia Carminha Colaco
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | | | | | - Girish Birappa
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Sripriya Rajkumar
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - D A Ayush Gowda
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Junwon Lee
- Institute of Vision Research, Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Eonjuro 211, Gangnam-Gu, Seoul, 06273, South Korea
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, 382715, Gujarat, India
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea.
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
15
|
Colaco JC, Chandrasekaran AP, Karapurkar JK, Gowda DAA, Birappa G, Rajkumar S, Suresh B, Ko N, Hong SH, Oh SJ, Kim KS, Ramakrishna S. E3 ubiquitin ligase APC/C Cdh1 regulates SLC35F2 protein turnover and inhibits cancer progression in HeLa cells. Biochim Biophys Acta Gen Subj 2023; 1867:130454. [PMID: 37689217 DOI: 10.1016/j.bbagen.2023.130454] [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: 06/15/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND The solute carrier family 35 F2 (SLC35F2), belongs to membrane-bound carrier proteins that control various physiological functions and are activated in several cancers. However, the molecular mechanism regulating SLC35F2 protein turnover and its implication in cancer progression remains unexplored. Therefore, screening for E3 ligases that promote SLC35F2 protein degradation is essential during cancer progression. METHODS The immunoprecipitation and Duolink proximity ligation assays (PLA) were used to determine the interaction between APC/CCdh1 and SLC35F2 proteins. A CRISPR/Cas9-mediated knockdown and rescue experiment were used to validate the functional significance of APC/CCdh1 on SLC35F2 protein stabilization. The ubiquitination function of APC/CCdh1 on SLC35F2 protein was validated using in vitro ubiquitination assay and half-life analysis. The role of APC/CCdh1 regulating SLC35F2-mediated tumorigenesis was confirmed by in vitro oncogenic experiments in HeLa cells. RESULTS Based on the E3 ligase screen and in vitro biochemical experiments, we identified that APC/CCdh1 interacts with and reduces SLC35F2 protein level. APC/CCdh1 promotes SLC35F2 ubiquitination and decreases the half-life of SLC35F2 protein. On the other hand, the CRISPR/Cas9-mediated depletion of APC/CCdh1 increased SLC35F2 protein levels. The mRNA expression analysis revealed a negative correlation between APC/CCdh1 and SLC35F2 across a panel of cancer cell lines tested. Additionally, we demonstrated that depletion in APC/CCdh1 promotes SLC35F2-mediated cell proliferation, colony formation, migration, and invasion in HeLa cells. CONCLUSION Our study highlights that APC/CCdh1 is a critical regulator of SLC35F2 protein turnover and depletion of APC/CCdh1 promotes SLC35F2-mediated tumorigenesis. Thus, we envision that APC/CCdh1-SLC35F2 axis might be a therapeutic target in cancer.
Collapse
Affiliation(s)
- Jencia Carminha Colaco
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | | | | | - D A Ayush Gowda
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | - Girish Birappa
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | - Sripriya Rajkumar
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | - Nare Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul 05505, Republic of Korea; Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
16
|
Sharma S, Chepurna O, Sun T. Drug resistance in glioblastoma: from chemo- to immunotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:688-708. [PMID: 38239396 PMCID: PMC10792484 DOI: 10.20517/cdr.2023.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 01/22/2024]
Abstract
As the most common and aggressive type of primary brain tumor in adults, glioblastoma is estimated to end over 10,000 lives each year in the United States alone. Stand treatment for glioblastoma, including surgery followed by radiotherapy and chemotherapy (i.e., Temozolomide), has been largely unchanged since early 2000. Cancer immunotherapy has significantly shifted the paradigm of cancer management in the past decade with various degrees of success in treating many hematopoietic cancers and some solid tumors, such as melanoma and non-small cell lung cancer (NSCLC). However, little progress has been made in the field of neuro-oncology, especially in the application of immunotherapy to glioblastoma treatment. In this review, we attempted to summarize the common drug resistance mechanisms in glioblastoma from Temozolomide to immunotherapy. Our intent is not to repeat the well-known difficulty in the area of neuro-oncology, such as the blood-brain barrier, but to provide some fresh insights into the molecular mechanisms responsible for resistance by summarizing some of the most recent literature. Through this review, we also hope to share some new ideas for improving the immunotherapy outcome of glioblastoma treatment.
Collapse
Affiliation(s)
| | | | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
17
|
Ghaderi S, Levkau B. An erythrocyte-centric view on the MFSD2B sphingosine-1-phosphate transporter. Pharmacol Ther 2023; 249:108483. [PMID: 37390971 DOI: 10.1016/j.pharmthera.2023.108483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
MFSD2B has been identified as the exclusive sphingosine-1-phosphate (S1P) transporter in red blood cells (RBC) and platelets. MFSD2B-mediated S1P export from platelets is required for aggregation and thrombus formation, whereas RBC MFSD2B maintains plasma S1P levels in concert with SPNS2, the vascular and lymphatic endothelial cell S1P exporter, to control endothelial permeability and ensure normal vascular development. However, the physiological function of MFSD2B in RBC remains rather elusive despite mounting evidence that the intracellular S1P pool plays important roles in RBC glycolysis, adaptation to hypoxia and the regulation of cell shape, hydration, and cytoskeletal organisation. The large accumulation of S1P and sphingosine in MFSD2B-deficient RBC coincides with stomatocytosis and membrane abnormalities, the reasons for which have remained obscure. MFS family members transport substrates in a cation-dependent manner along electrochemical gradients, and disturbances in cation permeability are known to alter cell hydration and shape in RBC. Furthermore, the mfsd2 gene is a transcriptional target of GATA together with mylk3, the gene encoding myosin light chain kinase (MYLK). S1P is known to activate MYLK and thereby impact on myosin phosphorylation and cytoskeletal architecture. This suggests that metabolic, transcriptional and functional interactions may exist between MFSD2B-mediated S1P transport and RBC deformability. Here, we review the evidence for such interactions and the implications for RBC homeostasis.
Collapse
Affiliation(s)
- Shahrooz Ghaderi
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany.
| |
Collapse
|
18
|
Chen Y, Li H, Wang K, Wang Y. Recent Advances in Synthetic Drugs and Natural Actives Interacting with OAT3. Molecules 2023; 28:4740. [PMID: 37375294 DOI: 10.3390/molecules28124740] [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: 05/18/2023] [Revised: 06/03/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Organic anion transporter 3 (OAT3) is predominantly expressed in the kidney and plays a vital role in drug clearance. Consequently, co-ingestion of two OAT3 substrates may alter the pharmacokinetics of the substrate. This review summarizes drug-drug interactions (DDIs) and herbal-drug interactions (HDIs) mediated by OAT3, and inhibitors of OAT3 in natural active compounds in the past decade. This provides a valuable reference for the combined use of substrate drugs/herbs for OAT3 in clinical practice in the future and for the screening of OAT3 inhibitors to avoid harmful interactions.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Hongyan Li
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Ke Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Yousheng Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| |
Collapse
|
19
|
Huang X, Li H, Shenkar N, Zhan A. Multidimensional plasticity jointly contributes to rapid acclimation to environmental challenges during biological invasions. RNA (NEW YORK, N.Y.) 2023; 29:675-690. [PMID: 36810233 PMCID: PMC10159005 DOI: 10.1261/rna.079319.122] [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: 06/16/2022] [Accepted: 02/01/2023] [Indexed: 05/06/2023]
Abstract
Rapid plastic response to environmental changes, which involves extremely complex underlying mechanisms, is crucial for organismal survival during many ecological and evolutionary processes such as those in global change and biological invasions. Gene expression is among the most studied molecular plasticity, while co- or posttranscriptional mechanisms are still largely unexplored. Using a model invasive ascidian Ciona savignyi, we studied multidimensional short-term plasticity in response to hyper- and hyposalinity stresses, covering the physiological adjustment, gene expression, alternative splicing (AS), and alternative polyadenylation (APA) regulations. Our results demonstrated that rapid plastic response varied with environmental context, timescales, and molecular regulatory levels. Gene expression, AS, and APA regulations independently acted on different gene sets and corresponding biological functions, highlighting their nonredundant roles in rapid environmental adaptation. Stress-induced gene expression changes illustrated the use of a strategy of accumulating free amino acids under high salinity and losing/reducing them during low salinity to maintain the osmotic homoeostasis. Genes with more exons were inclined to use AS regulations, and isoform switches in functional genes such as SLC2a5 and Cyb5r3 resulted in enhanced transporting activities by up-regulating the isoforms with more transmembrane regions. The extensive 3'-untranslated region (3'UTR) shortening through APA was induced by both salinity stresses, and APA regulation predominated transcriptomic changes at some stages of stress response. The findings here provide evidence for complex plastic mechanisms to environmental changes, and thereby highlight the importance of systemically integrating different levels of regulatory mechanisms in studying initial plasticity in evolutionary trajectories.
Collapse
Affiliation(s)
- Xuena Huang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Haidian District, Beijing 100085, China
| | - Hanxi Li
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Haidian District, Beijing 100085, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shijingshan District, Beijing 100049, China
| | - Noa Shenkar
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, 6997801 Tel-Aviv, Israel
- The Steinhardt Museum of Natural History, Israel National Center for Biodiversity Studies, Tel Aviv University, Tel-Aviv, Israel
| | - Aibin Zhan
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Haidian District, Beijing 100085, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shijingshan District, Beijing 100049, China
| |
Collapse
|
20
|
Feng W, Zuo M, Li W, Chen S, Wang Z, Yuan Y, Yang Y, Liu Y. A novel score system based on arginine metabolism-related genes to predict prognosis, characterize immune microenvironment, and forecast response to immunotherapy in IDH-wildtype glioblastoma. Front Pharmacol 2023; 14:1145828. [PMID: 37214463 PMCID: PMC10196947 DOI: 10.3389/fphar.2023.1145828] [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: 01/16/2023] [Accepted: 04/06/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction: Glioblastoma is one of the most lethal cancers and leads to more than 200,000 deaths annually. However, despite lots of researchers devoted to exploring novel treatment regime, most of these attempts eventually failed to improve the overall survival of glioblastoma patients in near 20 years. Immunotherapy is an emerging therapy for cancers and have succeeded in many cancers. But most of its application in glioblastoma have been proved with no improvement in overall survival, which may result from the unique immune microenvironment of glioblastoma. Arginine is amino acid and is involved in many physiological processes. Many studies have suggested that arginine and its metabolism can regulate malignancy of multiple cancers and influence the formation of tumor immune microenvironment. However, there is hardly study focusing on the role of arginine metabolism in glioblastoma. Methods: In this research, based on mRNA sequencing data of 560 IDH-wildtype glioblastoma patients from three public cohorts and one our own cohort, we aimed to construct an arginine metabolism-related genes signature (ArMRS) based on four essential arginine metabolism-related genes (ArMGs) that we filtered from all genes with potential relation with arginine metabolism. Subsequently, the glioblastoma patients were classified into ArMRS high-risk and low-risk groups according to calculated optimal cut-off values of ArMRS in these four cohorts. Results: Further validation demonstrated that the ArMRS was an independent prognostic factor and displayed fine efficacy in prediction of glioblastoma patients' prognosis. Moreover, analyses of tumor immune microenvironment revealed that higher ArMRS was correlated with more immune infiltration and relatively "hot" immunological phenotype. We also demonstrated that ArMRS was positively correlated with the expression of multiple immunotherapy targets, including PD1 and B7-H3. Additionally, the glioblastomas in the ArMRS high-risk group would present with more cytotoxic T cells (CTLs) infiltration and better predicted response to immune checkpoint inhibitors (ICIs). Discussion: In conclusion, our study constructed a novel score system based on arginine metabolism, ArMRS, which presented with good efficacy in prognosis prediction and strong potential to predict unique immunological features, resistance to immunotherapy, and guide the application of immunotherapy in IDH-wild type glioblastoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuan Yang
- *Correspondence: Yuan Yang, ; Yanhui Liu,
| | - Yanhui Liu
- *Correspondence: Yuan Yang, ; Yanhui Liu,
| |
Collapse
|
21
|
Huang C, Deng W, Xu HZ, Zhou C, Zhang F, Chen J, Bao Q, Zhou X, Liu M, Li J, Liu C. Short-chain fatty acids reprogram metabolic profiles with the induction of reactive oxygen species production in human colorectal adenocarcinoma cells. Comput Struct Biotechnol J 2023; 21:1606-1620. [PMID: 36874158 PMCID: PMC9975252 DOI: 10.1016/j.csbj.2023.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023] Open
Abstract
Short-chain fatty acids (SCFAs) exhibit anticancer activity in cellular and animal models of colon cancer. Acetate, propionate, and butyrate are the three major SCFAs produced from dietary fiber by gut microbiota fermentation and have beneficial effects on human health. Most previous studies on the antitumor mechanisms of SCFAs have focused on specific metabolites or genes involved in antitumor pathways, such as reactive oxygen species (ROS) biosynthesis. In this study, we performed a systematic and unbiased analysis of the effects of acetate, propionate, and butyrate on ROS levels and metabolic and transcriptomic signatures at physiological concentrations in human colorectal adenocarcinoma cells. We observed significantly elevated levels of ROS in the treated cells. Furthermore, significantly regulated signatures were involved in overlapping pathways at metabolic and transcriptomic levels, including ROS response and metabolism, fatty acid transport and metabolism, glucose response and metabolism, mitochondrial transport and respiratory chain complex, one-carbon metabolism, amino acid transport and metabolism, and glutaminolysis, which are directly or indirectly linked to ROS production. Additionally, metabolic and transcriptomic regulation occurred in a SCFAs types-dependent manner, with an increasing degree from acetate to propionate and then to butyrate. This study provides a comprehensive analysis of how SCFAs induce ROS production and modulate metabolic and transcriptomic levels in colon cancer cells, which is vital for understanding the mechanisms of the effects of SCFAs on antitumor activity in colon cancer.
Collapse
Key Words
- 1H–13C HMBC, 1H–13C Heteronuclear Multiple Bond Correlation Spectroscopy
- 1H–13C HSQC, 1H–13C Heteronuclear Single Quantum Coherence Spectroscopy
- 1H–1H COSY, 1H–1H Correlation Spectroscopy
- 1H–1H TOCSY, 1H–1H Total Correlation Spectroscopy
- ADP, Adenosine diphosphate
- AMP, Adenosine monophosphate
- ATP, Adenosine triphosphate
- Ace, Acetate
- Ach, Acetylcholine
- Ala, Alanine
- CRC, Colorectal Cancer
- Caco-2, Human Colon Adenocarcinoma
- Cho, Choline
- CoA, Coenzyme A
- Cre, Creatine
- DCFH-DA, Dichloro-Dihydro-Fluorescein Diacetate
- DEGs, Differentially Expressed Genes
- DMEM, Dulbecco's Modified Eagle Medium
- DMG, Dimethylglycine
- DNA, Deoxyribonucleic Acid
- EP, Eppendorf
- FA, Formate
- FDR, False Discovery Rate
- Fru, Fructose
- Fum, Fumaric acid
- GLS, Glutaminase
- GSEA, Gene Set Enrichment Analysis
- GSH, Glutathione
- Gal-1-P, Galactose-1-phosphate
- Glc, Glucose
- Gln, Glutamine
- Glu, Glutamate
- Gly, Glycine
- HCT116, Human Colorectal Carcinoma Cell Line
- HEK, Human Embryonic Kidney cells
- HT29, Human Colorectal Adenocarcinoma Cell Line with Epithelial Morphology
- His, Histidine
- Ile, Isoleucine
- J-Res, J-resolved Spectroscopy
- LDH, Lactate Dehydrogenase
- Lac, Lactate
- Leu, Leucine
- Lys, Lysine
- MCF-7, Human Breast Cancer Cell Line with Estrogen
- MCT, Monocarboxylate Transporters
- Met, Methionine
- MetS, Metabolic Syndrome
- Mitochondrial function
- NAD+, Nicotinamide adenine dinucleotide
- NAG, N-Acetyl-L-Glutamine
- NMR, Nuclear Magnetic Resonance
- NMR-based Metabolomics
- NOESY, Nuclear Overhauser Effect Spectroscopy
- O-PLS-DA, Orthogonal Projection to the Latent Structures Discriminant Analysis
- PA, Pantothenate
- PC, Phosphocholine
- PCA, Principal Component Analysis
- PDC, Pyruvate Decarboxylase
- PDK, Pyruvate Dehydrogenase Kinase
- PKC, Protein Kinase C
- PPP, Pentose Phosphate Pathway
- Phe, Phenylalanine
- Pyr, Pyruvate
- RNA, Ribonucleic Acid
- ROS, Reactive Oxygen Species
- RPKM, Reads per Kilobase of Transcript per Million Reads Mapped
- Reactive oxygen species
- SCFAs, Short Chain Fatty Acids
- SLC, Solute-Carrier Genes
- Short-chain fatty acids
- Suc, Succinate
- T2DM, Type 2 Diabetes
- TCA, Tricarboxylic Acid
- Tau, Taurine
- Thr, Threonine
- Transcriptomics
- Tyr, Tyrosine
- UDP, Uridine 5′-diphosphate
- UDP-GLC, UDP Glucose
- UDPG, UDP Glucuronate
- UDPGs, UDP Glucose and UDP Glucuronate
- UMP, Uridine 5′-monophosphate
- Val, Valine
- WST-1, Water-Soluble Tetrazolium salts
- dDNP, dissolution Dynamic Nuclear Polarization
- qRT-PCR, Real-Time Quantitative Reverse Transcription Polymerase Chain Reaction
- α-KIV, α-Keto-isovalerate
- α-KMV, α-keto-β-methyl-valerate
Collapse
Affiliation(s)
- Chongyang Huang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Wenjun Deng
- Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Huan-zhou Xu
- Department of Pediatrics, Division of Infectious Diseases, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Chen Zhou
- Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Fan Zhang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Junfei Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Qinjia Bao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Optics Valley Laboratory, Hubei 430074, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Optics Valley Laboratory, Hubei 430074, China
| | - Jing Li
- Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chaoyang Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Optics Valley Laboratory, Hubei 430074, China
| |
Collapse
|
22
|
Van Goor A, Pasternak A, Walugembe M, Chehab N, Hamonic G, Dekkers JCM, Harding JCS, Lunney JK. Genome wide association study of thyroid hormone levels following challenge with porcine reproductive and respiratory syndrome virus. Front Genet 2023; 14:1110463. [PMID: 36845393 PMCID: PMC9947478 DOI: 10.3389/fgene.2023.1110463] [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: 11/28/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction: Porcine reproductive and respiratory syndrome virus (PRRSV) causes respiratory disease in piglets and reproductive disease in sows. Piglet and fetal serum thyroid hormone (i.e., T3 and T4) levels decrease rapidly in response to Porcine reproductive and respiratory syndrome virus infection. However, the genetic control of T3 and T4 levels during infection is not completely understood. Our objective was to estimate genetic parameters and identify quantitative trait loci (QTL) for absolute T3 and/or T4 levels of piglets and fetuses challenged with Porcine reproductive and respiratory syndrome virus. Methods: Sera from 5-week-old pigs (N = 1792) at 11 days post inoculation (DPI) with Porcine reproductive and respiratory syndrome virus were assayed for T3 levels (piglet_T3). Sera from fetuses (N = 1,267) at 12 or 21 days post maternal inoculation (DPMI) with Porcine reproductive and respiratory syndrome virus of sows (N = 145) in late gestation were assayed for T3 (fetal_T3) and T4 (fetal_T4) levels. Animals were genotyped using 60 K Illumina or 650 K Affymetrix single nucleotide polymorphism (SNP) panels. Heritabilities, phenotypic correlations, and genetic correlations were estimated using ASREML; genome wide association studies were performed for each trait separately using Julia for Whole-genome Analysis Software (JWAS). Results: All three traits were low to moderately heritable (10%-16%). Phenotypic and genetic correlations of piglet_T3 levels with weight gain (0-42 DPI) were 0.26 ± 0.03 and 0.67 ± 0.14, respectively. Nine significant quantitative trait loci were identified for piglet_T3, on Sus scrofa chromosomes (SSC) 3, 4, 5, 6, 7, 14, 15, and 17, and collectively explaining 30% of the genetic variation (GV), with the largest quantitative trait loci identified on SSC5, explaining 15% of the genetic variation. Three significant quantitative trait loci were identified for fetal_T3 on SSC1 and SSC4, which collectively explained 10% of the genetic variation. Five significant quantitative trait loci were identified for fetal_T4 on SSC1, 6, 10, 13, and 15, which collectively explained 14% of the genetic variation. Several putative immune-related candidate genes were identified, including CD247, IRF8, and MAPK8. Discussion: Thyroid hormone levels following Porcine reproductive and respiratory syndrome virus infection were heritable and had positive genetic correlations with growth rate. Multiple quantitative trait loci with moderate effects were identified for T3 and T4 levels during challenge with Porcine reproductive and respiratory syndrome virus and candidate genes were identified, including several immune-related genes. These results advance our understanding of growth effects of both piglet and fetal response to Porcine reproductive and respiratory syndrome virus infection, revealing factors associated with genomic control of host resilience.
Collapse
Affiliation(s)
- Angelica Van Goor
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Services, Beltsville Agricultural Research Center, Beltsville, MD, United States
| | - Alex Pasternak
- Department of Animal Science, Purdue University, West Lafayette, IN, United States
| | - Muhammed Walugembe
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Nadya Chehab
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Services, Beltsville Agricultural Research Center, Beltsville, MD, United States
| | - Glenn Hamonic
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jack C. M. Dekkers
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - John C. S. Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joan K. Lunney
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Services, Beltsville Agricultural Research Center, Beltsville, MD, United States,*Correspondence: Joan K. Lunney,
| |
Collapse
|
23
|
Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, Miquelajáuregui Graf A, Estrada-Sánchez AM. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci 2023; 16:1037641. [PMID: 36744061 PMCID: PMC9893894 DOI: 10.3389/fncel.2022.1037641] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte's role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Collapse
Affiliation(s)
- Ares Orlando Cuellar-Santoyo
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Teresa Belem Mares-Barbosa
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Araceli Patrón-Soberano
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Andrew G. Howe
- Intelligent Systems Laboratory, HRL Laboratories, LLC, Malibu, CA, United States
| | - Diana Patricia Portales-Pérez
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | | | - Ana María Estrada-Sánchez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| |
Collapse
|
24
|
Global analysis of the association between pig muscle fatty acid composition and gene expression using RNA-Seq. Sci Rep 2023; 13:535. [PMID: 36631502 PMCID: PMC9834388 DOI: 10.1038/s41598-022-27016-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
Fatty acids (FAs) play an essential role as mediators of cell signaling and signal transduction, affecting metabolic homeostasis and determining meat quality in pigs. However, FAs are transformed by the action of several genes, such as those encoding desaturases and elongases of FAs in lipogenic tissues. The aim of the current work was to identify candidate genes, biological processes, and pathways involved in the modulation of intramuscular FA profile from longissimus dorsi muscle. FA profile by gas chromatography of methyl esters and gene expression by RNA-Seq were determined in 129 Iberian × Duroc backcrossed pigs. An association analysis between the muscle transcriptome and its FA profile was performed, followed by a concordance and functional analysis. Overall, a list of well-known (e.g., PLIN1, LEP, ELOVL6, SC5D, NCOA2, ACSL1, MDH1, LPL, LGALS12, TFRC, GOT1, and FBP1) and novel (e.g., TRARG1, TANK, ENSSSCG00000011196, and ENSSSCG00000038429) candidate genes was identified, either in association with specific or several FA traits. Likewise, several of these genes belong to biological processes and pathways linked to energy, lipid, and carbohydrate metabolism, which seem determinants in the modulation of FA compositions. This study can contribute to elucidate the complex relationship between gene expression and FA profile in pig muscle.
Collapse
|
25
|
Aryankalayil MJ, Bylicky MA, Martello S, Chopra S, Sproull M, May JM, Shankardass A, MacMillan L, Vanpouille-Box C, Dalo J, Scott KMK, Norman Coleman C. Microarray analysis identifies coding and non-coding RNA markers of liver injury in whole body irradiated mice. Sci Rep 2023; 13:200. [PMID: 36604457 PMCID: PMC9814510 DOI: 10.1038/s41598-022-26784-w] [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: 05/02/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Radiation injury from medical, accidental, or intentional sources can induce acute and long-term hepatic dysregulation, fibrosis, and cancer. This long-term hepatic dysregulation decreases quality of life and may lead to death. Our goal in this study is to determine acute changes in biological pathways and discover potential RNA biomarkers predictive of radiation injury. We performed whole transcriptome microarray analysis of mouse liver tissue (C57BL/6 J) 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray to identify significant expression changes in mRNAs, lncRNAs, and miRNAs, We also validated changes in specific RNAs through qRT-PCR. We used Ingenuity Pathway Analysis (IPA) to identify pathways associated with gene expression changes. We observed significant dysregulation of multiple mRNAs across all doses. In contrast, miRNA dysregulation was observed upwards of 2 Gray. The most significantly upregulated mRNAs function as tumor suppressors: Cdkn1a, Phlda3, and Eda2r. The most significantly downregulated mRNAs were involved in hemoglobin synthesis, inflammation, and mitochondrial function including multiple members of Hbb and Hba. The most significantly upregulated miRNA included: miR-34a-5p, miR-3102-5p, and miR-3960, while miR-342-3p, miR-142a-3p, and miR-223-3p were most significantly downregulated. IPA predicted activation of cell cycle checkpoint control pathways and inhibition of pathways relevant to inflammation and erythropoietin. Clarifying expression of mRNA, miRNA and lncRNA at a short time point (48 h) offers insight into potential biomarkers, including radiation markers shared across organs and animal models. This information, once validated in human models, can aid in development of bio-dosimetry biomarkers, and furthers our understanding of acute pathway dysregulation.
Collapse
Affiliation(s)
- Molykutty J. Aryankalayil
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Michelle A. Bylicky
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Shannon Martello
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Sunita Chopra
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Mary Sproull
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Jared M. May
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Aman Shankardass
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Laurel MacMillan
- grid.420517.50000 0004 0490 0428Gryphon Scientific, Takoma Park, MD 20912 USA
| | - Claire Vanpouille-Box
- grid.5386.8000000041936877XDepartment of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065 USA
| | - Juan Dalo
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Kevin M. K. Scott
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - C. Norman Coleman
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA ,grid.48336.3a0000 0004 1936 8075Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850 USA
| |
Collapse
|
26
|
Zhang X, Liu J, Wang H. The cGAS-STING-autophagy pathway: Novel perspectives in neurotoxicity induced by manganese exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120412. [PMID: 36240967 DOI: 10.1016/j.envpol.2022.120412] [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: 07/26/2022] [Revised: 08/28/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Chronic high-level heavy metal exposure increases the risk of developing different neurodegenerative diseases. Chronic excessive manganese (Mn) exposure is known to lead to neurodegenerative diseases. In addition, some evidence suggests that autophagy dysfunction plays an important role in the pathogenesis of various neurodegenerative diseases. Over the past decade, the DNA-sensing receptor cyclic GMP-AMP synthase (cGAS) and its downstream signal-efficient interferon gene stimulator (STING), as well as the molecular composition and regulatory mechanisms of this pathway have been well understood. The cGAS-STING pathway has emerged as a crucial mechanism to induce effective innate immune responses by inducing type I interferons in mammalian cells. Moreover, recent studies have found that Mn2+ is the second activator of the cGAS-STING pathway besides dsDNA, and inducing autophagy is a primitive function for the activation of the cGAS-STING pathway. However, overactivation of the immune response can lead to tissue damage. This review discusses the mechanism of neurotoxicity induced by Mn exposure from the cGAS-STING-autophagy pathway. Future work exploiting the cGAS-STING-autophagy pathway may provide a novel perspective for manganese neurotoxicity.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
27
|
Sheikh A, Tumala B, Vickers TJ, Martin JC, Rosa BA, Sabui S, Basu S, Simoes RD, Mitreva M, Storer C, Tyksen E, Head RD, Beatty W, Said HM, Fleckenstein JM. Enterotoxigenic Escherichia coli heat-labile toxin drives enteropathic changes in small intestinal epithelia. Nat Commun 2022; 13:6886. [PMID: 36371425 PMCID: PMC9653437 DOI: 10.1038/s41467-022-34687-7] [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: 06/07/2022] [Accepted: 11/01/2022] [Indexed: 11/14/2022] Open
Abstract
Enterotoxigenic E. coli (ETEC) produce heat-labile (LT) and/or heat-stable (ST) enterotoxins, and commonly cause diarrhea in resource-poor regions. ETEC have been linked repeatedly to sequelae in children including enteropathy, malnutrition, and growth impairment. Although cellular actions of ETEC enterotoxins leading to diarrhea are well-established, their contributions to sequelae remain unclear. LT increases cellular cAMP to activate protein kinase A (PKA) that phosphorylates ion channels driving intestinal export of salt and water resulting in diarrhea. As PKA also modulates transcription of many genes, we interrogated transcriptional profiles of LT-treated intestinal epithelia. Here we show that LT significantly alters intestinal epithelial gene expression directing biogenesis of the brush border, the major site for nutrient absorption, suppresses transcription factors HNF4 and SMAD4 critical to enterocyte differentiation, and profoundly disrupts microvillus architecture and essential nutrient transport. In addition, ETEC-challenged neonatal mice exhibit substantial brush border derangement that is prevented by maternal vaccination with LT. Finally, mice repeatedly challenged with toxigenic ETEC exhibit impaired growth recapitulating the multiplicative impact of recurring ETEC infections in children. These findings highlight impacts of ETEC enterotoxins beyond acute diarrheal illness and may inform approaches to prevent major sequelae of these common infections including malnutrition that impact millions of children.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brunda Tumala
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tim J Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John C Martin
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Subrata Sabui
- Departments of Medicine and Physiology/Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, 92697, USA
- Department of Research, VA Medical Center, Long Beach, CA, 90822, USA
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rita D Simoes
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chad Storer
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Erik Tyksen
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Richard D Head
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Hamid M Said
- Departments of Medicine and Physiology/Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, 92697, USA
- Department of Research, VA Medical Center, Long Beach, CA, 90822, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Infectious Diseases, Medicine Service, Veterans Affairs Saint Louis Health Care System, Saint Louis, MO, 63106, USA.
| |
Collapse
|
28
|
Abrahams-October Z, Johnson R, Benjeddou M, Cloete R. The determination of the effect(s) of solute carrier family 22-member 2 (SLC22A2) haplotype variants on drug binding via molecular dynamic simulation systems. Sci Rep 2022; 12:16936. [PMID: 36209293 PMCID: PMC9547889 DOI: 10.1038/s41598-022-21291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/26/2022] [Indexed: 12/29/2022] Open
Abstract
Single nucleotide polymorphisms detected in the solute carrier member family-22 has been shown to result in a variable response in the treatment of type 2 diabetes mellitus with Metformin. This study predicted a three-dimensional protein structure for the SLC22A2 protein sequence using AlphaFold 2 and modelled five haplotypes within SLC22A2 protein structure observed in the Xhosa population of South Africa. The protein models were used to determine the effect(s) of haplotype variations on the transport function of Metformin and 10 other drugs by the SLC22A2 protein. Molecular dynamic simulation studies, molecular docking and interaction analysis of the five SLC22A2 haplotypes were performed in complex with the ligand 5RE in a POPC lipid bilayer to understand the mechanism of drug binding. Weakest binding free energy was found between 5RE and haplotype 1. Molecular docking studies indicated the top binding ligands as well as Metformin to bind inside the transport channel in all haplotypes increasing the probability of Metformin inhibition during co-administration of drugs. Metformin showed reduced binding affinity and number of interactions compared to the top four binding molecules. Molecular dynamic simulation analysis indicated that haplotypes 1, 3 and 4 were less stable than 2 and 5. The findings suggest haplotypes 4 and 5 having stronger preference for large inhibitor molecule binding in the active site and this could result in haplotypes 4 and 5 demonstrating reduced Metformin clearance via the SLC22A2 transporter during co-administration of drugs. The current study is the first to investigate the potential effect(s) of haplotype variation on the protein structure of SLC22A2 to assess its ability to transport Metformin in an indigenous South African population.
Collapse
Affiliation(s)
- Zainonesa Abrahams-October
- grid.8974.20000 0001 2156 8226Precision Medicine Unit, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville, 7535 South Africa
| | - Rabia Johnson
- grid.415021.30000 0000 9155 0024Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505 South Africa ,grid.11956.3a0000 0001 2214 904XDivision of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505 South Africa
| | - Mongi Benjeddou
- grid.8974.20000 0001 2156 8226Precision Medicine Unit, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Robert Sobukwe Road, Bellville, 7535 South Africa
| | - Ruben Cloete
- grid.8974.20000 0001 2156 8226South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535 South Africa
| |
Collapse
|
29
|
Xiao Y, Zhang H, Li Z, Huang T, Akihiro T, Xu J, Xu H, Lin F. An amino acid transporter-like protein (OsATL15) facilitates the systematic distribution of thiamethoxam in rice for controlling the brown planthopper. PLANT BIOTECHNOLOGY JOURNAL 2022; 20:1888-1901. [PMID: 35678495 PMCID: PMC9491460 DOI: 10.1111/pbi.13869] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Characterization and genetic engineering of plant transporters involved in the pesticide uptake and translocation facilitate pesticide relocation to the tissue where the pests feed, thus improving the bioavailability of the agrichemicals. We aimed to identify thiamethoxam (THX) transporters in rice and modify their expression for better brown planthopper (BPH) control with less pesticide application. A yeast library expressing 1385 rice transporters was screened, leading to the identification of an amino acid transporter-like (ATL) gene, namely OsATL15, which facilitates THX uptake in both yeast cells and rice seedlings. In contrast to a decrease in THX content in osatl15 knockout mutants, ectopic expression of OsATL15 under the control of the CaMV 35S promoter or a vascular-bundle-specific promoter gdcsPpro significantly increased THX accumulation in rice plants, thus further enhancing the THX efficacy against BPH. OsATL15 was localized in rice cell membrane and abundant in the root transverse sections, vascular bundles of leaf blade, and stem longitudinal sections, but not in hull and brown rice at filling stages. Our study shows that OsATL15 plays an essential role in THX uptake and its systemic distribution in rice. OsATL15 could be valuable in achieving precise pest control by biotechnology approaches.
Collapse
Affiliation(s)
- Yuyan Xiao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources/Key Laboratory of Natural Pesticide and Chemical BiologyMinistry of Education, South China Agricultural UniversityGuangzhouChina
| | - Hanlin Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources/Key Laboratory of Natural Pesticide and Chemical BiologyMinistry of Education, South China Agricultural UniversityGuangzhouChina
| | - Zhiwei Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources/Key Laboratory of Natural Pesticide and Chemical BiologyMinistry of Education, South China Agricultural UniversityGuangzhouChina
| | - Tinghong Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources/Key Laboratory of Natural Pesticide and Chemical BiologyMinistry of Education, South China Agricultural UniversityGuangzhouChina
| | - Takashi Akihiro
- Faculty of Life and Environmental ScienceShimane UniversityShimaneJapan
| | - Jian Xu
- College of Life SciencesSouth China Agricultural UniversityGuangzhouChina
| | - Hanhong Xu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources/Key Laboratory of Natural Pesticide and Chemical BiologyMinistry of Education, South China Agricultural UniversityGuangzhouChina
| | - Fei Lin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources/Key Laboratory of Natural Pesticide and Chemical BiologyMinistry of Education, South China Agricultural UniversityGuangzhouChina
| |
Collapse
|
30
|
Wu Z, Han T, Su H, Xuan J, Wang X. Comprehensive analysis of fatty acid and lactate metabolism–related genes for prognosis value, immune infiltration, and therapy in osteosarcoma patients. Front Oncol 2022; 12:934080. [PMID: 36119478 PMCID: PMC9478861 DOI: 10.3389/fonc.2022.934080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma is the most frequent bone tumor. Notwithstanding that significant medical progress has been achieved in recent years, the 5-year overall survival of osteosarcoma patients is inferior. Regulation of fatty acids and lactate plays an essential role in cancer metabolism. Therefore, our study aimed to comprehensively assess the fatty acid and lactate metabolism pattern and construct a fatty acid and lactate metabolism–related risk score system to predict prognosis in osteosarcoma patients. Clinical data and RNA expression data were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Gene Expression Omnibus (GEO) databases. We used the least absolute shrinkage and selection operator (LASSO) and Cox regression analyses to construct a prognostic risk score model. Relationships between the risk score model and age, gender, tumor microenvironment characteristics, and drug sensitivity were also explored by correlation analysis. We determined the expression levels of prognostic genes in osteosarcoma cells via Western blotting. We developed an unknown fatty acid and lactate metabolism–related risk score system based on three fatty acid and lactate metabolism–related genes (SLC7A7, MYC, and ACSS2). Survival analysis showed that osteosarcoma patients in the low-risk group were likely to have a better survival time than those in the high-risk group. The area under the curve (AUC) value shows that our risk score model performs well in predicting prognosis. Elevated fatty acids and lactate risk scores weaken immune function and the environment of the body, which causes osteosarcoma patients’ poor survival outcomes. In general, the constructed fatty acid and lactate metabolism–related risk score model can offer essential insights into subsequent mechanisms in available research. In addition, our study may provide rational treatment strategies for clinicians based on immune correlation analysis and drug sensitivity in the future.
Collapse
Affiliation(s)
- Zhouwei Wu
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Han
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haohan Su
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiangwei Xuan
- Department of Orthopaedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, China
- *Correspondence: Xinwei Wang, ; Jiangwei Xuan,
| | - Xinwei Wang
- Department of Orthopaedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, China
- *Correspondence: Xinwei Wang, ; Jiangwei Xuan,
| |
Collapse
|
31
|
Anabtawi N, Drabison T, Hu S, Sparreboom A, Talebi Z. The role of OATP1B1 and OATP1B3 transporter polymorphisms in drug disposition and response to anticancer drugs: a review of the recent literature. Expert Opin Drug Metab Toxicol 2022; 18:459-468. [PMID: 35983889 DOI: 10.1080/17425255.2022.2113380] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Members of the solute carrier family of organic anion transporting polypeptides are responsible for the cellular uptake of a broad range of endogenous compounds and xenobiotics in multiple tissues. In particular, the polymorphic transporters OATP1B1 and OATP1B3 are highly expressed in the liver and have been identified as critical regulators of hepatic eliminaton. As these transporters are also expressed in cancer cells, the function alteration of these proteins have important consequences for an individual's susceptibility to certain drug-induced side effects, drug-drug interactions, and treatment efficacy. AREAS COVERED In this mini-review, we provide an update of this rapidly emerging field, with specific emphasis on the direct contribution of genetic variants in OATP1B1 and OATP1B3 to the transport of anticancer drugs, the role of these carriers in regulation of their disposition and toxicity profiles, and recent advances in attempts to integrate information on transport function in patients to derive individualized treatment strategies. EXPERT OPINION Based on currently available data, it appears imperative that different aspects of disease, physiology, and drugs of relevance should be evaluated along with an individual's genetic signature, and that tools such as biomarker levels can be implemented to achieve the most reliable prediction of clinically relevant pharmacodynamic endpoints.
Collapse
Affiliation(s)
- Nadeen Anabtawi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Thomas Drabison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio.,Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Zahra Talebi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| |
Collapse
|
32
|
Yu KH, Park J, Mittal A, Abou-Alfa GK, Dika IE, Epstein AS, Ilson DH, Kelsen DP, Ku GY, Li J, Park W, Varghese AM, Chou JFL, Capanu M, Cooper B, Bartlett A, McCarthy D, Sangar V, McCarthy B, O’Reilly EM. Circulating tumor and invasive cell expression profiling predicts effective therapy in pancreatic cancer. Cancer 2022; 128:2958-2966. [PMID: 35647938 PMCID: PMC10131181 DOI: 10.1002/cncr.34269] [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: 03/02/2022] [Revised: 04/06/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic adenocarcinoma (PDAC) remains a refractory disease; however, modern cytotoxic chemotherapeutics can induce tumor regression and extend life. A blood-based, pharmacogenomic, chemosensitivity assay using gene expression profiling of circulating tumor and invasive cells (CTICs) to predict treatment response was previously developed. The combination regimen of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel (G/nab-P) are established frontline approaches for treating advanced PDAC; however, there are no validated biomarkers for treatment selection. A similar unmet need exists for choosing second-line therapy. METHODS The chemosensitivity assay was evaluated in metastatic PDAC patients presenting for frontline treatment. A prospective study enrolled patients (n = 70) before receiving either FOLFIRINOX or G/nab-P at a 1:1 ratio. Six milliliters of peripheral blood was collected at baseline and at time of disease progression. CTICs were isolated, gene-expression profiling was performed, and the assay was used to predict effective and ineffective chemotherapeutic agents. Treating physicians were blinded to the assay prediction results. RESULTS Patients receiving an effective regimen as predicted by the chemosensitivity assay experienced significantly longer median progression-free survival (mPFS; 7.8 months vs. 4.2 months; hazard ratio [HR], 0.35; p = .0002) and median overall survival (mOS; 21.0 months vs. 9.7 months; HR, 0.40; p = .005), compared with an ineffective regimen. Assay prediction for effective second-line therapy was explored. The entire study cohort experienced favorable outcomes compared with historical controls, 7.1-month mPFS and 12.3-month mOS. CONCLUSIONS Chemosensitivity assay profiling is a promising tool for guiding therapy in advanced PDAC. Further prospective validation is under way (clinicaltrials.gov NCT03033927).
Collapse
Affiliation(s)
- Kenneth H. Yu
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Jennifer Park
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Avni Mittal
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ghassan K. Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Imane El Dika
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Andrew S. Epstein
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - David H. Ilson
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - David P. Kelsen
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Geoffrey Y. Ku
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Jia Li
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Wungki Park
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Anna M. Varghese
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | | | | | | | | | | | | | | | - Eileen M. O’Reilly
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
33
|
Adhikari B, Lee CN, Khadka VS, Deng Y, Fukumoto G, Thorne M, Caires K, Odani J, Mishra B. RNA-Sequencing based analysis of bovine endometrium during the maternal recognition of pregnancy. BMC Genomics 2022; 23:494. [PMID: 35799127 PMCID: PMC9264496 DOI: 10.1186/s12864-022-08720-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/24/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Maternal recognition is the crucial step for establishing pregnancy in cattle. This study aims to identify endometrial genes and biological pathways involved in the maternal recognition of pregnancy. Caruncular endometrial tissues were collected from Day 15-17 of gestation (pregnant), non-pregnant (absence of conceptus), and cyclic (non-bred) heifers. RESULTS Total RNAs were isolated from the caruncular endometrial tissues of pregnant, non-pregnant, and cyclic heifers, and were subjected to high-throughput RNA-sequencing. The genes with at least two-fold change and Benjamini and Hochberg p-value ≤ 0.05 were considered differentially expressed genes and further confirmed with quantitative real-time PCR. A total of 107 genes (pregnant vs cyclic) and 98 genes (pregnant vs non-pregnant) were differentially expressed in the pregnant endometrium. The most highly up-regulated genes in the pregnant endometrium were MRS2, CST6, FOS, VLDLR, ISG15, IFI6, MX2, C15H11ORF34, EIF3M, PRSS22, MS4A8, and TINAGL1. Interferon signaling, immune response, nutrient transporter, synthesis, and secretion of proteins are crucial pathways during the maternal recognition of pregnancy. CONCLUSIONS The study demonstrated that the presence of conceptus at Day 15-17 of gestation affects the endometrial gene expression related to endometrial remodeling, immune response, nutrients and ion transporters, and relevant signaling pathways in the caruncular region of bovine endometrium during the maternal recognition of pregnancy.
Collapse
Affiliation(s)
- Bindu Adhikari
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Chin N Lee
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Vedbar S Khadka
- Department of Quantitative Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Glen Fukumoto
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Mark Thorne
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Kyle Caires
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Jenee Odani
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Birendra Mishra
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
34
|
ChemoSensitivity Assay Guided Metronomic Chemotherapy Is Safe and Effective for Treating Advanced Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14122906. [PMID: 35740571 PMCID: PMC9220997 DOI: 10.3390/cancers14122906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Innovative chemotherapy regimens and tools to guide therapy in advanced pancreatic cancer are greatly needed. We present results of a study combining an innovative, metronomic chemotherapy strategy together with a blood-based pharmacogenomic tool to guide effective drug therapy. This study provides proof of principle that guided, metronomic chemotherapy for treatment of pancreatic cancer is a promising approach. Abstract Cytotoxic chemotherapy remains the mainstay of treatment for advanced pancreatic adenocarcinoma (PDAC). Emerging studies support metronomic chemotherapy (MCT) as effective, challenging established paradigms of dosing and schedules. The blood-based ChemoSensitivity Assay has been shown to predict response and survival in advanced PDAC patients treated with standard chemotherapy. The current study combines these concepts for a highly personalized treatment approach. This was a retrospective analysis; a pilot (n = 50) and validation cohort (n = 45) were studied. The ChemoSensitivity Assay was performed at baseline and during therapy; results were correlated to drugs administered and patient outcomes. MCT was administered based on the assay results at the treating physician′s discretion. Patients in the pilot cohort experienced favorable survival compared with historical controls (median overall survival (mOS) 16.8 mo). Patients whose treatment closely matched the ChemoSensitivity Assay predictions experienced longer median time on lines of therapy (5.3 vs. 3.3 mo, p = 0.02) and showed a trend for longer mOS (20.9 vs. 12.5 mo, p = 0.055) compared with those not closely matched. These findings were confirmed in the validation cohort. Overall, patients treated with MCT closely matching Assay results experienced a remarkable mOS of 27.7 mo. ChemoSensitivity profiling-guided MCT is a promising approach for personalized therapy in advanced PDAC.
Collapse
|
35
|
Dorostgou Z, Yadegar N, Dorostgou Z, Khorvash F, Vakili O. Novel insights into the role of circular RNAs in Parkinson disease: An emerging renaissance in the management of neurodegenerative diseases. J Neurosci Res 2022; 100:1775-1790. [PMID: 35642104 DOI: 10.1002/jnr.25094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 05/11/2022] [Accepted: 05/15/2022] [Indexed: 11/06/2022]
Abstract
Parkinson's disease (PD), as a debilitating neurodegenerative disease, particularly affects the elderly population, and is clinically identified by resting tremor, rigidity, and bradykinesia. Pathophysiologically, PD is characterized by an early loss of dopaminergic neurons in the Substantia nigra pars compacta, accompanied by the extensive aggregation of alpha-synuclein (α-Syn) in the form of Lewy bodies. The onset of PD has been reported to be influenced by multiple biological molecules. In this context, circular RNAs (circRNAs), as tissue-specific noncoding RNAs with closed structures, have been recently demonstrated to involve in a set of PD's pathogenic processes. These RNA molecules can either up- or downregulate the expression of α-Syn, as well as moderating its accumulation through different regulatory mechanisms, in which targeting microRNAs (miRNAs) is considered the most common pathway. Since circRNAs have prominent structural and biological characteristics, they could also be considered as promising candidates for PD diagnosis and treatment. Unfortunately, PD has become a global health concern, and a large number of its pathogenic processes are still unclear; thus, it is crucial to elucidate the ambiguous aspects of PD pathophysiology to improve the efficiency of diagnostic and therapeutic strategies. In line with this fact, the current review aims to highlight the interplay between circRNAs and PD pathogenesis, and then discusses the diagnostic and therapeutic potential of circRNAs in PD progression. This study will thus be the first of its kind reviewing the relationship between circRNAs and PD.
Collapse
Affiliation(s)
- Zahra Dorostgou
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Negar Yadegar
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zeynab Dorostgou
- Department of Biology, Kavian Institute of Higher Education, Mashhad, Iran
| | - Fariborz Khorvash
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Isfahan Neurosciences Research Center, Al-zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
36
|
Influence of a Polyherbal Choline Source in Dogs: Body Weight Changes, Blood Metabolites, and Gene Expression. Animals (Basel) 2022; 12:ani12101313. [PMID: 35625159 PMCID: PMC9137459 DOI: 10.3390/ani12101313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/03/2022] [Accepted: 05/14/2022] [Indexed: 11/16/2022] Open
Abstract
Choline chloride is used to provide choline in dog foods; however, in other domestic species, it has been replaced with a polyherbal containing phosphatidylcholine. A polyherbal containing Achyrantes aspera, Trachyspermum ammi, Citrullus colocynthis, Andrographis paniculata, and Azadirachta indica was evaluated in adult dogs through body weight changes, subcutaneous fat thickness, blood metabolites, and gene expression. Forty dogs (4.6 ± 1.6 years old) who were individually housed in concrete kennels were randomly assigned to the following treatments: unsupplemented diet (377 mg choline/kg), choline chloride (3850 mg/kg equivalent to 2000 mg choline/kg diet), and polyherbal (200, 400, and 800 mg/kg) for 60 days. Blood samples were collected on day 59 for biochemistry, biometry, and gene expression analysis through microarray assays. Intake, final body weight, and weight changes were similar for the two choline sources. Feed intake variation among dogs (p = 0.01) and dorsal fat (p = 0.03) showed a quadratic response to herbal choline. Dogs that received the polyherbal diet had reduced blood cholesterol levels (Quadratic, p = 0.02). The gene ontology analysis indicated that 15 biological processes were modified (p ≤ 0.05) with implications for preventing cardiovascular and metabolic diseases, cancer prevention, inflammatory and immune response, and behavior and cognitive process. According to these results that were observed in a 60 day trial, the polyherbal form could replace choline chloride in dog diets at a concentration of 400 mg/kg.
Collapse
|
37
|
Kim TW, Pyo DH, Ko E, Yun NH, Song SJ, Choi SM, Hong HK, Kim SH, Choi YL, Lee J, Lee WY, Cho YB. Expression of SLC22A18 regulates oxaliplatin resistance by modulating the ERK pathway in colorectal cancer. Am J Cancer Res 2022; 12:1393-1408. [PMID: 35411243 PMCID: PMC8984883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/24/2021] [Indexed: 06/14/2023] Open
Abstract
Although oxaliplatin-based chemotherapy is the current standard adjuvant therapy for colorectal cancer (CRC), the molecular mechanisms underlying oxaliplatin resistance remain unclear. Here, we examined the molecular mechanisms underlying SLC22A18-associated oxaliplatin resistance and strategies for overcoming oxaliplatin resistance. We evaluated the association between SLC22A18 and prognosis in 337 patients with CRC and its functional significance and studied the mechanisms through which SLC22A18 affects oxaliplatin resistance development in CRC cells, using CRC cell lines and patient-derived cells (PDCs). SLC22A18 downregulation was positively correlated with worse survival in patients with CRC. Low SLC22A18-expressing cells showed relatively lower sensitivity to oxaliplatin than high SLC22A18-expressing cells. In addition, ERK activation was found to be involved in the mechanisms underlying SLC22A18-related oxaliplatin resistance. To confirm ERK pathway dependence, we used an ERK inhibitor and found that combined treatment with oxaliplatin and the ERK inhibitor overcame oxaliplatin resistance in the low SLC22A18-expressing cells. Ex vivo approaches using PDC confirmed the correlation between SLC22A18 expression and oxaliplatin resistance. Results of the in vivo study showed that SLC22A18 expression regulated oxaliplatin efficacy, and that combined treatment with an ERK inhibitor could be a useful therapeutic strategy when SLC22A18 is downregulated. Together, our findings indicate that SLC22A18 could serve as a biomarker for the prediction of oxaliplatin resistance. In cases of oxaliplatin resistance due to low SLC22A18 expression, resistance can be overcome by combined treatment with an ERK inhibitor.
Collapse
Affiliation(s)
- Tae Won Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan UniversitySeoul, Republic of Korea
| | - Dae Hee Pyo
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, Republic of Korea
| | - Eunbyeol Ko
- Institute for Future Medicine Samsung Medical CenterSeoul, Republic of Korea
| | - Nak Hyeon Yun
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan UniversitySeoul, Republic of Korea
| | - Su Jeong Song
- Institute for Future Medicine Samsung Medical CenterSeoul, Republic of Korea
| | - Soo Min Choi
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan UniversitySeoul, Republic of Korea
| | - Hye Kyung Hong
- Institute for Future Medicine Samsung Medical CenterSeoul, Republic of Korea
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, Republic of Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, Republic of Korea
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, Republic of Korea
| | - Yong Beom Cho
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan UniversitySeoul, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, Republic of Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan UniversitySeoul, Republic of Korea
| |
Collapse
|
38
|
Weyrich A, Frericks M, Eichenlaub M, Schneider S, Hofmann T, Van Cruchten S, van Ravenzwaay B. Ontogeny of renal, hepatic, and placental expression of ATP-binding cassette and solute carrier transporters in the rat and the rabbit. Reprod Toxicol 2022; 107:1-9. [PMID: 34757165 DOI: 10.1016/j.reprotox.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 02/08/2023]
Abstract
Species differences in developmental toxicity can be due to varying expression of xenobiotic transporters. Hence, knowledge on the ontogeny of these transporters, especially in human, rat and rabbit, is pivotal. Two superfamilies of transporters, the ATP-binding cassette (ABC) and the solute carrier (SLC) transporters, are well known for their role in the absorption, distribution and/or elimination of xenobiotics and endogenous substances. The aim of this study was to compare the expression levels of these xenobiotic transporters in liver, kidney and placenta of man, Wistar rat and New Zealand White rabbit during pre- and postnatal development. For this purpose, qPCR experiments were performed for rat and rabbit tissues and the gene expression profiles were compared with literature data from man, rat and rabbit. Data analysis showed large differences in transporter expression in development and between species. These results can be used to better understand developmental toxicity findings in non-clinical species and their relevance for man.
Collapse
Affiliation(s)
- Anastasia Weyrich
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany.
| | - Markus Frericks
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Michael Eichenlaub
- Bioscience Research, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Steffen Schneider
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Thomas Hofmann
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Steven Van Cruchten
- Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Bennard van Ravenzwaay
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| |
Collapse
|
39
|
Benítez R, Núñez Y, Ayuso M, Isabel B, Fernández-Barroso MA, De Mercado E, Gómez-Izquierdo E, García-Casco JM, López-Bote C, Óvilo C. Changes in Biceps femoris Transcriptome along Growth in Iberian Pigs Fed Different Energy Sources and Comparative Analysis with Duroc Breed. Animals (Basel) 2021; 11:ani11123505. [PMID: 34944282 PMCID: PMC8697974 DOI: 10.3390/ani11123505] [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: 10/28/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The genetic mechanisms that regulate biological processes, such as skeletal muscle development and growth, or intramuscular fat deposition, have attracted great interest, given their impact on production traits and meat quality. In this sense, a comparison of the transcriptome of skeletal muscle between phenotypically different pig breeds, or along growth, could be useful to improve the understanding of the molecular processes underlying the differences in muscle metabolism and phenotypic traits, potentially driving the identification of causal genes, regulators and metabolic pathways involved in their variability. Abstract This experiment was conducted to investigate the effects of developmental stage, breed, and diet energy source on the genome-wide expression, meat quality traits, and tissue composition of biceps femoris muscle in growing pure Iberian and Duroc pigs. The study comprised 59 Iberian (IB) and 19 Duroc (DU) animals, who started the treatment at an average live weight (LW) of 19.9 kg. The animals were kept under identical management conditions and fed two diets with different energy sources (6% high oleic sunflower oil or carbohydrates). Twenty-nine IB animals were slaughtered after seven days of treatment at an average LW of 24.1 kg, and 30 IB animals plus all the DU animals were slaughtered after 47 days at an average LW of 50.7 kg. The main factors affecting the muscle transcriptome were age, with 1832 differentially expressed genes (DEGs), and breed (1055 DEGs), while the effect of diet on the transcriptome was very small. The results indicated transcriptome changes along time in Iberian animals, being especially related to growth and tissue development, extracellular matrix (ECM) composition, and cytoskeleton organization, with DEGs affecting relevant functions and biological pathways, such as myogenesis. The breed also affected functions related to muscle development and cytoskeleton organization, as well as functions related to solute transport and lipid and carbohydrate metabolism. Taking into account the results of the two main comparisons (age and breed effects), we can postulate that the Iberian breed is more precocious than the Duroc breed, regarding myogenesis and muscle development, in the studied growing stage.
Collapse
Affiliation(s)
- Rita Benítez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28040 Madrid, Spain; (R.B.); (Y.N.); (M.A.F.-B.); (J.M.G.-C.)
| | - Yolanda Núñez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28040 Madrid, Spain; (R.B.); (Y.N.); (M.A.F.-B.); (J.M.G.-C.)
| | - Miriam Ayuso
- Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, B-2610 Wilrijk, Belgium;
| | - Beatriz Isabel
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (B.I.); (C.L.-B.)
| | - Miguel A. Fernández-Barroso
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28040 Madrid, Spain; (R.B.); (Y.N.); (M.A.F.-B.); (J.M.G.-C.)
| | - Eduardo De Mercado
- Centro de Pruebas de Porcino ITACYL, Hontalbilla, 40353 Segovia, Spain; (E.D.M.); (E.G.-I.)
| | - Emilio Gómez-Izquierdo
- Centro de Pruebas de Porcino ITACYL, Hontalbilla, 40353 Segovia, Spain; (E.D.M.); (E.G.-I.)
| | - Juan M. García-Casco
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28040 Madrid, Spain; (R.B.); (Y.N.); (M.A.F.-B.); (J.M.G.-C.)
| | - Clemente López-Bote
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (B.I.); (C.L.-B.)
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28040 Madrid, Spain; (R.B.); (Y.N.); (M.A.F.-B.); (J.M.G.-C.)
- Correspondence: ; Tel.: +34-91-3471492
| |
Collapse
|
40
|
Ta HDK, Minh Xuan DT, Tang WC, Anuraga G, Ni YC, Pan SR, Wu YF, Fitriani F, Putri Hermanto EM, Athoillah M, Andriani V, Ajiningrum PS, Wang CY, Lee KH. Novel Insights into the Prognosis and Immunological Value of the SLC35A (Solute Carrier 35A) Family Genes in Human Breast Cancer. Biomedicines 2021; 9:1804. [PMID: 34944621 PMCID: PMC8698499 DOI: 10.3390/biomedicines9121804] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
According to statistics 2020, female breast cancer (BRCA) became the most commonly diagnosed malignancy worldwide. Prognosis of BRCA patients is still poor, especially in population with advanced or metastatic. Particular functions of each members of the solute carrier 35A (SLC35A) gene family in human BRCA are still unknown regardless of awareness that they play critical roles in tumorigenesis and progression. Using integrated bioinformatics analyses to identify therapeutic targets for specific cancers based on transcriptomics, proteomics, and high-throughput sequencing, we obtained new information and a better understanding of potential underlying molecular mechanisms. Leveraging BRCA dataset that belongs to The Cancer Genome Atlas (TCGA), which were employed to clarify SLC35A gene expression levels. Then we used a bioinformatics approach to investigate biological processes connected to SLC35A family genes in BRCA development. Beside that, the Kaplan-Meier estimator was leveraged to explore predictive values of SLC35A family genes in BCRA patients. Among individuals of this family gene, expression levels of SLC35A2 were substantially related to poor prognostic values, result from a hazard ratio of 1.3 (with 95 percent confidence interval (95% CI: 1.18-1.44), the p for trend (ptrend) is 3.1 × 10-7). Furthermore, a functional enrichment analysis showed that SLC35A2 was correlated with hypoxia-inducible factor 1A (HIF1A), heat shock protein (HSP), E2 transcription factor (E2F), DNA damage, and cell cycle-related signaling. Infiltration levels observed in specific types of immune cell, especially the cluster of differentiation found on macrophages and neutrophils, were positively linked with SLC35A2 expression in multiple BRCA subclasses (luminal A, luminal B, basal, and human epidermal growth factor receptor 2). Collectively, SLC35A2 expression was associated with a lower recurrence-free survival rate, suggesting that it could be used as a biomarker in treating BRCA.
Collapse
Affiliation(s)
- Hoang Dang Khoa Ta
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.D.K.T.); (G.A.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
| | - Do Thi Minh Xuan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
| | - Wan-Chun Tang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Gangga Anuraga
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.D.K.T.); (G.A.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia; (F.F.); (E.M.P.H.); (M.A.)
| | - Yi-Chun Ni
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
| | - Syu-Ruei Pan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
| | - Yung-Fu Wu
- National Defense Medical Center, School of Medicine, Department of Medical Research, Tri-Service General Hospital, Taipei 11490, Taiwan;
| | - Fenny Fitriani
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia; (F.F.); (E.M.P.H.); (M.A.)
| | - Elvira Mustikawati Putri Hermanto
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia; (F.F.); (E.M.P.H.); (M.A.)
| | - Muhammad Athoillah
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia; (F.F.); (E.M.P.H.); (M.A.)
| | - Vivin Andriani
- Department of Biological Science, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia; (V.A.); (P.S.A.)
| | - Purity Sabila Ajiningrum
- Department of Biological Science, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia; (V.A.); (P.S.A.)
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.D.K.T.); (G.A.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
| | - Kuen-Haur Lee
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.D.K.T.); (G.A.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (D.T.M.X.); (Y.-C.N.); (S.-R.P.)
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei 11031, Taiwan
| |
Collapse
|
41
|
Kim M, Park SC, Lee DY. Glycyrrhizin as a Nitric Oxide Regulator in Cancer Chemotherapy. Cancers (Basel) 2021; 13:cancers13225762. [PMID: 34830916 PMCID: PMC8616433 DOI: 10.3390/cancers13225762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Glycyrrhizin (GL) has anti-cancer, anti-inflammatory, anti-viral, and anti-oxidant activity. In particular, GL reduces multidrug resistance (MDR) in cancer cells, which is a major obstacle to chemotherapy. Nitric oxide (NO) also plays an important role in MDR, and GL affects NO concentration in the tumor microenvironment. However, the effects of GL and NO interaction on MDR have not been reviewed. Here, we review the role of GL as an NO regulator in cancer cells and its subsequent anti-MDR effect and posit that GL is a promising MDR inhibitor for cancer chemotherapy. Abstract Chemotherapy is used widely for cancer treatment; however, the evolution of multidrug resistance (MDR) in many patients limits the therapeutic benefits of chemotherapy. It is important to overcome MDR for enhanced chemotherapy. ATP-dependent efflux of drugs out of cells is the main mechanism of MDR. Recent studies have suggested that nitric oxide (NO) can be used to overcome MDR by inhibiting the ATPase function of ATP-dependent pumps. Several attempts have been made to deliver NO to the tumor microenvironment (TME), however there are limitations in delivery. Glycyrrhizin (GL), an active compound of licorice, has been reported to both reduce the MDR effect by inhibiting ATP-dependent pumps and function as a regulator of NO production in the TME. In this review, we describe the potential role of GL as an NO regulator and MDR inhibitor that efficiently reduces the MDR effect in cancer chemotherapy.
Collapse
Affiliation(s)
- Minsu Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea; (M.K.); (S.C.P.)
| | - Seok Chan Park
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea; (M.K.); (S.C.P.)
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea; (M.K.); (S.C.P.)
- Institute of Nano Science & Technology (INST), Hanyang University, Seoul 04763, Korea
- Elixir Pharmatech Inc., Seoul 04763, Korea
- Correspondence:
| |
Collapse
|
42
|
Auranofin: Past to Present, and repurposing. Int Immunopharmacol 2021; 101:108272. [PMID: 34731781 DOI: 10.1016/j.intimp.2021.108272] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 01/15/2023]
Abstract
Auranofin (AF), a gold compound, has been used to treat rheumatoid arthritis (RA) for more than 40 years; however, its mechanism of action remains unknown. We revealed that AF inhibited the induction of proinflammatory proteins and their mRNAs by the inflammatory stimulants, cyclooxygenase-2 and inducible nitric oxide synthase, and their upstream regulator, NF-κB. AF also activated the proteins peroxyredoxin-1, Kelch-like ECH-associated protein 1, and NF-E2-related factor 2, and inhibited thioredoxin reductase, all of which are involved in oxidative or electrophilic stress under physiological conditions. Although the cell membrane was previously considered to be permeable to AF because of its hydrophobicity, the mechanisms responsible for transporting AF into and out of cells as well as its effects on the uptake and excretion of other drugs have not yet been elucidated. Antibodies for cytokines have recently been employed in the treatment of RA, which has had an impact on the use of AF. Trials to repurpose AF as a risk-controlled agent to treat cancers or infectious diseases, including severe acute respiratory syndrome coronavirus 2/coronavirus disease 2019, are ongoing. Novel gold compounds are also under development as anti-cancer and anti-infection agents.
Collapse
|
43
|
Nazempour N, Taleqani MH, Taheri N, Haji Ali Asgary Najafabadi AH, Shokrollahi A, Zamani A, Fattahi Dolatabadi N, Peymani M, Mahdevar M. The role of cell surface proteins gene expression in diagnosis, prognosis, and drug resistance of colorectal cancer: In silico analysis and validation. Exp Mol Pathol 2021; 123:104688. [PMID: 34592197 DOI: 10.1016/j.yexmp.2021.104688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022]
Abstract
Cell surface proteins (CSPs) are an important type of protein in different essential cell functions. This study aimed to distinguish overexpressed CSPs in colorectal cancer to investigate their biomarker, prognosis, and drug resistance potential. Raw data of three datasets including 1187 samples was downloaded then normalization and differential expression were performed. By the combination of the cancer genome atlas (TCGA) clinical data, survival analysis was carried out. Information of all CSPs was collected from cell surface protein atlas. The role of each candidate gene expression was investigated in drug resistance by CCEL and GDSC data from PharmacoGX. CRC samples including 30 tumor samples and adjacent normal were used to confirm data by RT-qPCR. Outcomes showed that 66 CSPs overexpressed in three datasets, and 146 CSPs expression associated with poor prognosis features in TCGA data that TIMP1 and QSOX2 can associate with poor patient survival independently. High-risk patients illustrated more fatality than low-risk patients based on the risk score calculated by the expression level of these genes. Receiver operating characteristic curve analysis showed that 39 CSPs as perfect biomarkers for diagnosis in CRC. Furthermore, QSOX2 and TIMP1 expression levels increased in tumor samples compared to adjacent normal samples. The Drug resistance analysis demonstrated ADAM12 and COL1A2 up-regulation among 66 overexpressed CSPs caused resistance to Venetoclax and Cyclophosphamide with a high estimate, respectively. Many CSPs are deregulated in CRC, and can be valuable candidates as biomarkers for diagnosis, prognosis, and drug resistance.
Collapse
Affiliation(s)
- Nasrin Nazempour
- Department of Chemistry, Shahreza Branch, Islamic Azad University, Shahreza, Isfahan, Iran; Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | - Mohammad Hossein Taleqani
- Department of Biology, Faculty of Science, University of Yazd, Yazd, Iran; Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | - Navid Taheri
- Department of Microbiology, Zanjan Branch, Islamic Azad University, Zanjan, Iran; Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | | | - Alireza Shokrollahi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Atefeh Zamani
- Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | | | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Sharekord, Iran.
| | - Mohammad Mahdevar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
44
|
Berkman AM, Hildebrandt MA, Landstrom AP. The genetic underpinnings of anthracycline-induced cardiomyopathy predisposition. Clin Genet 2021; 100:132-143. [PMID: 33871046 PMCID: PMC9902211 DOI: 10.1111/cge.13968] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Anthracyclines, chemotherapeutic agents that have contributed to significant improvements in cancer survival, also carry risk of both acute and chronic cardiotoxicity. This has led to significantly elevated risks of cardiac morbidity and mortality among cancer survivors treated with these agents. Certain treatment related, demographic, and medical factors increase an individual's risk of anthracycline induced cardiotoxicity; however, significant variability among those affected suggests that there is an underlying genetic predisposition to anthracycline induced cardiotoxicity. The current narrative review seeks to summarize the literature to date that has identified genetic variants associated with anthracycline induced cardiotoxicity. These include variants found in genes that encode proteins associated with anthracycline transportation and metabolism, those that encode proteins associated with the generation of reactive oxygen species, and those known to be associated with cardiac disease. While there is strong evidence that susceptibility to anthracycline induced cardiotoxicity has genetic underpinnings, the majority of work to date has been candidate gene analyses. Future work should focus on genome-wide analyses including genome-wide association and sequencing-based studies to confirm and expand these findings.
Collapse
Affiliation(s)
- Amy M. Berkman
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
| | - Michelle A.T. Hildebrandt
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas, United States
| | - Andrew P. Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
- Department of Cell Biology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
| |
Collapse
|
45
|
van Deventer R, Rhode C, Marx M, Roodt-Wilding R. Elucidation of coat colour genetics in blue wildebeest. Mamm Biol 2021. [DOI: 10.1007/s42991-021-00126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
46
|
Pasquariello KZ, Dey JM, Sprowl JA. Current Understanding of Membrane Transporters as Regulators or Targets for Cisplatin-Induced Hearing Loss. Mol Pharmacol 2021; 100:348-355. [PMID: 34330821 DOI: 10.1124/molpharm.121.000274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/22/2021] [Indexed: 11/22/2022] Open
Abstract
Cisplatin is a platinum-based drug which remains among the most efficacious anticancer treatment options. Unfortunately, use of cisplatin is hindered by dose-limiting toxicities, including irreversible hearing loss, which can grossly affect patient quality of life. Cisplatin-induced ototoxicity is the result of cochlear hair cell damage through a mechanism that is poorly understood. However, cisplatin cytotoxicity is reliant on intracellular accumulation, a process that is largely dependent on the presence of particular membrane transporters. This review will provide an update on our current understanding of the various transporters known to be involved in the disposition and cytotoxicity of platinum drugs or their metabolites, as well as their role in mediating cisplatin-induced hearing loss. We also provide a summary of the successes and opportunities in therapeutically targeting membrane transporters to alleviate platinum-induced hearing loss. Moreover, we describe how this approach could be used to reduce the severity or onset of other adverse events associated with exposure to various forms of platinum drugs, without diminishing anti-tumor efficacy. Significance Statement Cisplatin-induced hearing loss is a dose limiting and irreversible adverse event with no current preventative or curative treatment measures. Pharmacological targeting of membrane transporters that regulate platinum uptake into cochlear hair cells, if conducted appropriately, may alleviate this devastating side effect and could be applied to alleviate other platinum-induced toxicities.
Collapse
Key Words
- Uptake transporters (OATP, OAT, OCT, PEPT, MCT, NTCP, ASBT, etc.)
- cancer chemotherapy
- efflux transporters (P-gp, BCRP, MRP, MATE, BSEP, etc)
- ototoxicity
Collapse
Affiliation(s)
| | | | - Jason A Sprowl
- School of Pharmacy, University of Buffalo, United States
| |
Collapse
|
47
|
Smagin DA, Babenko VN, Redina OE, Kovalenko IL, Galyamina AG, Kudryavtseva NN. Reduced Expression of Slc Genes in the VTA and NAcc of Male Mice with Positive Fighting Experience. Genes (Basel) 2021; 12:genes12071099. [PMID: 34356115 PMCID: PMC8306410 DOI: 10.3390/genes12071099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/27/2021] [Accepted: 07/16/2021] [Indexed: 12/28/2022] Open
Abstract
A range of several psychiatric medications targeting the activity of solute carrier (SLC) transporters have proved effective for treatment. Therefore, further research is needed to elucidate the expression profiles of the Slc genes, which may serve as markers of altered brain metabolic processes and neurotransmitter activities in psychoneurological disorders. We studied the Slc differentially expressed genes (DEGs) using transcriptomic profiles in the ventral tegmental area (VTA), nucleus accumbens (NAcc), and prefrontal cortex (PFC) of control and aggressive male mice with psychosis-like behavior induced by repeated experience of aggression accompanied with wins in daily agonistic interactions. The majority of the Slc DEGs were shown to have brain region-specific expression profiles. Most of these genes in the VTA and NAcc (12 of 17 and 25 of 26, respectively) were downregulated, which was not the case in the PFC (6 and 5, up- and downregulated, respectively). In the VTA and NAcc, altered expression was observed for the genes encoding the transporters of neurotransmitters as well as inorganic and organic ions, amino acids, metals, glucose, etc. This indicates an alteration in transport functions for many substrates, which can lead to the downregulation or even disruption of cellular and neurotransmitter processes in the VTA and NAcc, which are attributable to chronic stimulation of the reward systems induced by positive fighting experience. There is not a single Slc DEG common to all three brain regions. Our findings show that in male mice with repeated experience of aggression, altered activity of neurotransmitter systems leads to a restructuring of metabolic and neurotransmitter processes in a way specific for each brain region. We assume that the scoring of Slc DEGs by the largest instances of significant expression co-variation with other genes may outline a candidate for new prognostic drug targets. Thus, we propose that the Slc genes set may be treated as a sensitive genes marker scaffold in brain RNA-Seq studies.
Collapse
Affiliation(s)
- Dmitry A. Smagin
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Vladimir N. Babenko
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
| | - Olga E. Redina
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
| | - Irina L. Kovalenko
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Anna G. Galyamina
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Natalia N. Kudryavtseva
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
48
|
ActTRANS: Functional classification in active transport proteins based on transfer learning and contextual representations. Comput Biol Chem 2021; 93:107537. [PMID: 34217007 DOI: 10.1016/j.compbiolchem.2021.107537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 05/09/2021] [Accepted: 06/26/2021] [Indexed: 01/08/2023]
Abstract
MOTIVATION Primary and secondary active transport are two types of active transport that involve using energy to move the substances. Active transport mechanisms do use proteins to assist in transport and play essential roles to regulate the traffic of ions or small molecules across a cell membrane against the concentration gradient. In this study, the two main types of proteins involved in such transport are classified from transmembrane transport proteins. We propose a Support Vector Machine (SVM) with contextualized word embeddings from Bidirectional Encoder Representations from Transformers (BERT) to represent protein sequences. BERT is a powerful model in transfer learning, a deep learning language representation model developed by Google and one of the highest performing pre-trained model for Natural Language Processing (NLP) tasks. The idea of transfer learning with pre-trained model from BERT is applied to extract fixed feature vectors from the hidden layers and learn contextual relations between amino acids in the protein sequence. Therefore, the contextualized word representations of proteins are introduced to effectively model complex structures of amino acids in the sequence and the variations of these amino acids in the context. By generating context information, we capture multiple meanings for the same amino acid to reveal the importance of specific residues in the protein sequence. RESULTS The performance of the proposed method is evaluated using five-fold cross-validation and independent test. The proposed method achieves an accuracy of 85.44 %, 88.74 % and 92.84 % for Class-1, Class-2, and Class-3, respectively. Experimental results show that this approach can outperform from other feature extraction methods using context information, effectively classify two types of active transport and improve the overall performance.
Collapse
|
49
|
Mohammadi F, Rostami G, Assad D, Shafiei M, Hamid M, Jalaeikhoo H. Association of SLC22A1,SLCO1B3 Drug Transporter Polymorphisms and Smoking with Disease Risk and Cytogenetic Response to Imatinib in Patients with Chronic Myeloid Leukemia. Lab Med 2021; 52:584-596. [PMID: 34128532 DOI: 10.1093/labmed/lmab023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To determine whether polymorphisms of SLC22A1 and SLCO1B3 genes could predict imatinib (IM) response and chronic myeloid leukemia (CML) risk. METHODS We genotyped SLC22A1 (c.480G > C, c.1222A > G) and SLCO1B3 (c.334T > G, c.699G > A) polymorphisms in 132 patients with CML and 109 sex- and age-matched healthy subjects. The patients were evaluated for cytogenetic response by standard chromosome banding analysis (CBA). RESULTS Polymorphism analysis showed significant increased risk of IM resistance for SLC22A1c.1222AG (P = .03; OR = 2.2), SLCO1B3c.334TT/TG genotypes (P = .007; OR = 4.37) and 334T allele (P = .03; OR = 2.86). The double combinations of SLC22A1c.480CC and c.1222AG polymorphisms with SLCO1B3c.334TT/TG were significantly associated with complete cytogenetic response (CCyR) (P <.05; OR> 7). The interaction between all polymorphisms and smoking were associated with CML development and IM resistance (P ≤.04; OR> 3). CONCLUSIONS Our study results suggest the influence of SLC22A1 and SLCO1B3 polymorphisms and the interaction of smoking on CML development and IM response.
Collapse
Affiliation(s)
- Fatemeh Mohammadi
- Department of Biology, School of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Golale Rostami
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Dlnya Assad
- Department of Biology, College of Science, Sulaimani University, Sulaymanyah, Iraq
| | - Mohammad Shafiei
- Department of Biology, School of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Biotechnology and Biological Science Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Hamid
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Jalaeikhoo
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Gargiuli C, Sepe P, Tessari A, Sheetz T, Colecchia M, de Braud FGM, Procopio G, Sensi M, Verzoni E, Dugo M. Integrative Transcriptomic Analysis Reveals Distinctive Molecular Traits and Novel Subtypes of Collecting Duct Carcinoma. Cancers (Basel) 2021; 13:2903. [PMID: 34200770 PMCID: PMC8230422 DOI: 10.3390/cancers13122903] [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: 04/30/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Collecting duct carcinoma (CDC) is a rare and highly aggressive kidney cancer subtype with poor prognosis and no standard treatments. To date, only a few studies have examined the transcriptomic portrait of CDC. Through integration of multiple datasets, we compared CDC to normal tissue, upper-tract urothelial carcinomas, and other renal cancers, including clear cell, papillary, and chromophobe histologies. Association between CDC gene expression signatures and in vitro drug sensitivity data was evaluated using the Cancer Therapeutic Response Portal, Genomics of Drug Sensitivity in Cancer datasets, and connectivity map. We identified a CDC-specific gene signature that predicted in vitro sensitivity to different targeted agents and was associated to worse outcome in clear cell renal cell carcinoma. We showed that CDC are transcriptionally related to the principal cells of the collecting ducts providing evidence that this tumor originates from this normal kidney cell type. Finally, we proved that CDC is a molecularly heterogeneous disease composed of at least two subtypes distinguished by cell signaling, metabolic and immune-related alterations. Our findings elucidate the molecular features of CDC providing novel biological and clinical insights. The identification of distinct CDC subtypes and their transcriptomic traits provides the rationale for patient stratification and alternative therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Gargiuli
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Pierangela Sepe
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
| | - Anna Tessari
- Department of Cancer Biology and Genetics, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.T.); (T.S.)
| | - Tyler Sheetz
- Department of Cancer Biology and Genetics, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.T.); (T.S.)
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Maurizio Colecchia
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Filippo Guglielmo Maria de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20133 Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
| | - Marialuisa Sensi
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
| | - Matteo Dugo
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| |
Collapse
|