1
|
Fu D, Shu X, Zhou G, Ji M, Liao G, Zou L. Connection between oral health and chronic diseases. MedComm (Beijing) 2025; 6:e70052. [PMID: 39811802 PMCID: PMC11731113 DOI: 10.1002/mco2.70052] [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: 06/12/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Chronic diseases have emerged as a paramount global health burden, accounting for 74% of global mortality and causing substantial economic losses. The oral cavity serves as a critical indicator of overall health and is inextricably linked to chronic disorders. Neglecting oral health can exacerbate localized pathologies and accelerate the progression of chronic conditions, whereas effective management has the potential to reduce their incidence and mortality. Nevertheless, limited resources and lack of awareness often impede timely dental intervention, delaying optimal therapeutic measures. This review provides a comprehensive analysis of the impact of prevalent chronic diseases-such as diabetes mellitus, rheumatoid arthritis, cardiovascular disorders, and chronic respiratory diseases-on oral health, along with an exploration of how changes in oral health affect these chronic conditions through both deterioration and intervention mechanisms. Additionally, novel insights into the underlying pathophysiological mechanisms governing these relationships are presented. By synthesizing these advancements, this review aims to illuminate the complex interrelationship between oral health and chronic diseases while emphasizing the urgent need for greater collaboration between dental practitioners and general healthcare providers to improve overall health outcomes.
Collapse
Affiliation(s)
- Di Fu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Xingyue Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Ge Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Mengzhen Ji
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
- Department of Information Management, Department of Stomatology Informatics, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
2
|
Karzoon A, Yerer MB, Cumaoğlu A. Empagliflozin demonstrates cytotoxicity and synergy with tamoxifen in ER-positive breast cancer cells: anti-proliferative and anti-survival effects. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:781-798. [PMID: 39066911 DOI: 10.1007/s00210-024-03316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
Accumulating evidence suggests that sodium-glucose cotransporter 2 (SGLT2) inhibitors may be effective at eliminating tumor cells. While empagliflozin exhibits nearly the highest selectivity for SGLT2 over SGLT1, its specific impact alone and in combination with tamoxifen remains largely unexplored in estrogen receptor α-positive (ERα +) breast cancer. This study investigated the anticancer effects of empagliflozin and its potential synergy with tamoxifen in MCF-7 breast cancer cells. The individual and combined cytotoxic effects of empagliflozin and tamoxifen were assessed using the xCELLigence system. The activities of AMP-activated protein kinase α (AMPKα), p38 mitogen-activated protein kinase (p38 MAPKα), p70-S6 kinase 1 (p70S6K1), and protein kinase B (Akt) were assessed using Western blotting. The gene expression levels of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) and Forkhead box O3a (FOXO3a) were assessed via qPCR. Our results revealed time- and concentration-dependent cytotoxic effects of empagliflozin and tamoxifen whether administered separately or in combination. While tamoxifen exhibits potency with an IC50 value of 17 μM, approximately ten times greater than that of empagliflozin (IC50 = 177 μM), synergistic effects are observed when the concentrations of the two agents approach their respective IC50 values. Additionally, empagliflozin significantly increases AMPKα activity while concurrently inhibiting Akt, p70S6K1, and p38 MAPKα, and these effects are significantly enhanced when empagliflozin is combined with tamoxifen. Moreover, empagliflozin modulates the gene expression, downregulating PGC-1α while upregulating FOXO3a. Empagliflozin exerts anti-proliferative and anti-survival effects by inhibiting mTOR, Akt, and PGC-1α, and it exhibits synergy with tamoxifen in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Ahmad Karzoon
- Department of Pharmacology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye.
| | - Mükerrem Betül Yerer
- Drug Application and Research Center (ERFARMA), Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Türkiye
| | - Ahmet Cumaoğlu
- Department of Biochemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Türkiye
| |
Collapse
|
3
|
Xu B, Zhou J. Sodium-glucose cotransporter 2 inhibitors and renal cancer in the US FDA adverse event reporting system. Eur J Clin Pharmacol 2024; 80:1959-1966. [PMID: 39285057 DOI: 10.1007/s00228-024-03759-6] [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: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 11/13/2024]
Abstract
BACKGROUND Recent evidence suggests an association between sodium-glucose cotransporter 2 (SGLT2) inhibitors and a higher risk of renal cancer. OBJECTIVE We conducted a pharmacovigilance analysis using the US FDA Adverse Event Reporting System (FAERS) to investigate the disproportionate association between SGLT2 inhibitors and renal cancer. METHODS We used AERSMine to mine data from FAERS, covering the period from 2014 Q1 to 2023 Q3. The control group was treated with other glucose-lowering medications (ATC-A10B). Disproportionality analysis results were performed using a proportional reporting ratio (PRR) with a 95% confidence interval (CI) and an information component (IC) with 95% credible interval. RESULTS Compared to the control group, the SGLT2 inhibitor group had a higher disproportionate renal cancer reporting frequency (0.92 vs 0.27/1000 reports; PRR 3.38; 95% CI 2.68-4.25; p < 0.001) with an IC of 1.36 (0.60-2.06), comprising dapagliflozin (PRR 4.14; 2.95-5.80; p < 0.001), empagliflozin (PRR 2.74; 1.94-3.89; p < 0.001), and canagliflozin (PRR 3.56; 2.48-5.12; p < 0.001). Consistent results were obtained in the diabetes indication with the primary outcomes only for the SGLT2 inhibitors group (not individual molecule). The results of the sensitivity analysis (excluding hypertension indication or antihypertensive drugs, obesity, smoking, alpha-1 blockers, or anti-renal cancer drugs) were highly consistent with the main outcomes, indicating good robustness of the results. The results from 2004 Q1 to 2023 Q3 were similar to those from 2014 Q1 to 2023 Q3, with the exception of empagliflozin. CONCLUSION There was a disproportionate association between SGLT2 inhibitors and renal cancer, which supports the current meta-analysis results indicating an increased risk of renal cancer associated with SGLT2 inhibitors.
Collapse
Affiliation(s)
- Bo Xu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, University of South China, Hengyang, 421001, Hunan, China
| | - Jiecan Zhou
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Pharmacy Department, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
4
|
D’Orazio G, La Ferla B. Synthesis of a Small Library of Glycoderivative Putative Ligands of SGLT1 and Preliminary Biological Evaluation. Molecules 2024; 29:5067. [PMID: 39519708 PMCID: PMC11547630 DOI: 10.3390/molecules29215067] [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: 09/18/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Sodium-glucose co-transporter 1 (SGLT1) is primarily expressed on the membrane of enterocytes, a type of epithelial cell found in the intestines, where it mediates the unidirectional absorption of glucose and galactose. Beyond its well-established role in nutrient absorption, SGLT1 also plays a protective role in maintaining the integrity of the intestinal barrier. Specifically, the natural ligand of SGLT1 (d-glucose) and a synthetic C-glucoside developed by our group can induce a protective anti-inflammatory effect on the intestinal epithelium. In this paper, we report the creation of a small library of C-glycoside, putative ligands for SGLT1, to gain further insights into its unclear mechanism of action. Preliminary biological experiments performed on an in vitro model of doxorubicin-induced mucositis, a severe intestinal inflammatory condition, indicate that the aromatic moiety present in all the compounds of the library is crucial for biological activity, while the sugar component appears to have less influence. These findings will be exploited to develop new, more potent anti-inflammatory compounds and to better understand and rationalize the protective mechanism of action.
Collapse
Affiliation(s)
- Giuseppe D’Orazio
- Department of Chemistry, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy
| | - Barbara La Ferla
- Department of Earth and Environmental Sciences DISAT, Università degli Studi di Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| |
Collapse
|
5
|
Xu B, Kang B, Li S, Fan S, Zhou J. Sodium-glucose cotransporter 2 inhibitors and cancer: a systematic review and meta-analysis. J Endocrinol Invest 2024; 47:2421-2436. [PMID: 38530620 DOI: 10.1007/s40618-024-02351-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/24/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND The effect of sodium-glucose cotransporter 2 (SGLT2) inhibitors on cancer has yet to be fully elucidated. OBJECTIVE This systematic review and meta-analysis investigated the effects of SGLT2 inhibitors on cancer. METHODS We searched the PubMed and ClinicalTrials.gov databases up to July 15, 2023, to identify eligible randomized, double-blind, placebo-controlled trials that lasted at least ≥24 weeks. The primary outcome was the overall cancer incidence, and the secondary outcomes were the incidences of various types of cancer. We used the Mantel-Haenszel method, fixed effects model, risk ratio (RR) and 95% confidence interval (CI) to analyze dichotomous variables. Subgroup analysis was performed based on the SGLT2 inhibitor type, baseline conditions, and follow-up duration. All meta-analyses were performed using RevMan5.4.1 and Stata MP 16.0. RESULTS A total of 58 publications (59 trials) were included, comprising 113,909 participants with type 2 diabetes mellitus and/or chronic kidney disease and/or high cardiovascular risk and/or heart failure (SGLT2 inhibitor group, 63864; placebo group, 50045). Compared to the placebo SGLT2 inhibitors did not significantly increase the overall incidence of cancer (RR 1.01; 95% CI 0.94-1.08; p = 0.82). However, ertugliflozin did significantly increase the overall incidence of cancer (RR 1.29; 95% CI 1.01-1.64; p = 0.04). SGLT2 inhibitors did not increase the risks of bladder or breast cancer. However, dapagliflozin did significantly reduce the risk of bladder cancer by 47% (RR 0.53; 95% CI 0.35-0.81; p = 0.003). SGLT2 inhibitors had no significant effect on the risks of gastrointestinal, thyroid, skin, respiratory, prostate, uterine/endometrial, hepatic and pancreatic cancers. Dapagliflozin reduced the risk of respiratory cancer by 26% (RR 0.74; 95% CI 0.55-1.00; p = 0.05). SGLT2 inhibitors (particularly mediated by dapagliflozin and ertugliflozin but not statistically significant) were associated with a greater risk of renal cancer than the placebo (RR 1.39; 95% CI 1.04-1.87; p = 0.03). CONCLUSION SGLT2 inhibitors did not significantly increase the overall risk of cancer or the risks of bladder and breast cancers. However, the higher risk of renal cancer associated with SGLT2 inhibitors warrants concern.
Collapse
Affiliation(s)
- B Xu
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - B Kang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - S Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The Affiliated Nanhua Hospital, Department of Docimasiology, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - S Fan
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - J Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
6
|
Ghezzi C, Ellingson BM, Lai A, Liu J, Barrio JR, Wright EM. Effect of Jardiance on glucose uptake into astrocytomas. J Neurooncol 2024; 169:437-444. [PMID: 39037688 PMCID: PMC11341586 DOI: 10.1007/s11060-024-04746-8] [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: 05/06/2024] [Accepted: 06/11/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE SGLT2, the sodium glucose cotransporter two, is expressed in human pancreatic, prostate and brain tumors, and in a mouse cancer model SGLT2 inhibitors reduce tumor glucose uptake and growth. In this study we have measured the effect of a specific SGLT2 inhibitor, Jardiance® (Empagliflozin), on glucose uptake into astrocytomas in patients. METHODS We have used a specific SGLT glucose tracer, α-methyl-4-[18F]fluoro-4-deoxy-α-D-glucopyranoside (Me4FDG), and Positron Emission Tomography (PET) to measure glucose uptake. Four of five patients enrolled had WHO grade IV glioblastomas, and one had a low grade WHO Grade II astrocytoma. Two dynamic brain PET scans were conducted on each patient, one before and one after treatment with a single oral dose of Jardiance, a specific SGLT2 inhibitor. As a control, we also determined the effect of oral Jardiance on renal SGLT2 activity. RESULTS In all five patients an oral dose (25 or 100 mg) of Jardiance reduced Me4FDG tumor accumulation, highly significant inhibition in four, and inhibited SGLT2 activity in the kidney. CONCLUSIONS These initial experiments show that SGLT2 is a functional glucose transporter in astocytomas, and Jardiance inhibited glucose uptake, a drug approved by the FDA to treat type 2 diabetes mellitus (T2DM), heart failure, and renal failure. We suggest that clinical trials be initiated to determine whether Jardiance reduces astrocytoma growth in patients.
Collapse
Affiliation(s)
- Chiara Ghezzi
- Department of Physiology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1751, USA
| | - Benjamin M Ellingson
- Department of Radiological Sciences, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Albert Lai
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Jie Liu
- Department of Molecular and Medical Pharmacology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Jorge R Barrio
- Department of Molecular and Medical Pharmacology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ernest M Wright
- Department of Physiology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1751, USA.
| |
Collapse
|
7
|
Kawabata-Iwakawa R, Iwasa N, Satoh K, Colinge J, Shimada M, Takeuchi S, Fujiwara H, Eguchi H, Oishi T, Sugiyama T, Suzuki M, Hasegawa K, Fujiwara K, Nishiyama M. Prediction of response to promising first-line chemotherapy in ovarian cancer patients with residual peritoneal tumors: practical biomarkers and robust multiplex models. Int J Clin Oncol 2024; 29:1334-1346. [PMID: 38767719 DOI: 10.1007/s10147-024-02552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Platinum/taxane (TC) chemotherapy with debulking surgery stays the mainstay of the treatment in ovarian cancer patients with peritoneal metastasis, and recently its novel modality, intraperitoneal carboplatin with dose-dense paclitaxel (ddTCip), was shown to have greater therapeutic impact. Nevertheless, the response varies among patients and consequent recurrence, or relapse often occurs. Discovery of therapeutic response predictor to ddTCip and/or TC therapy is eagerly awaited to improve the treatment outcome. METHODS Using datasets in 76 participants in our ddTCip study and published databases on patients received TC therapy, we first validated a total of 75 previously suggested markers, sought out more active biomarkers through the association analyses of genome-wide transcriptome and genotyping data with progression-free survival (PFS) and adverse events, and then developed multiplex statistical prediction models for PFS and toxicity by mainly using multiple regression analysis and the classification and regression tree (CART) algorithm. RESULTS The association analyses revealed that SPINK1 could be a possible biomarker of ddTCip efficacy, while ABCB1 rs1045642 and ERCC1 rs11615 would be a predictor of hematologic toxicity and peripheral neuropathy, respectively. Multiple regression analyses and CART algorithm finally provided a potent efficacy prediction model using 5 gene expression data and robust multiplex toxicity prediction models-CART models using a total of 4 genotype combinations and multiple regression models using 15 polymorphisms on 12 genes. CONCLUSION Biomarkers and multiplex models composed here could work well in the response prediction of ddTCip and/or TC therapy, which might contribute to realize optimal selection of the key therapy.
Collapse
Grants
- DOFMET-08 Development Organization for Frontier Medical Education and Therapeutics in Japan
- H21-3rd Comprehensive 10-year Strategy for Cancer Control-010 Ministry of Health, Labour and Welfare
- University Reform Action Plan "Gunma University Initiative for Advanced Research (GIAR) Ministry of Education, Culture, Sports, Science, and Technology (JP)
- University Reform Action Plan "Gunma University Initiative for Advanced Research (GIAR)" Ministry of Education, Culture, Sports, Science, and Technology (JP)
- Promotion Plan for the Platform of Human Resource Development for Cancer Ministry of Education, Culture, Sports, Science, and Technology (JP)
- the Fostering Health Professionals for Changing Needs of Cancer Ministry of Education, Culture, Sports, Science, and Technology (JP)
- New Paradigms - Establishing Center for Fostering Medical Researchers of the Future Programs Ministry of Education, Culture, Sports, Science, and Technology (JP)
Collapse
Affiliation(s)
- Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma, 371-8511, Japan
- Research Unit and Immunology and Inflammation, Department of Translational Research, Division of Sohyaku Innovative Research, Tanabe Mitsubishi Pharma, Osaka, Japan
| | - Norihiro Iwasa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
| | - Kenichi Satoh
- Faculty of Data Science, Shiga University, Hikone, Shiga, 522-8522, Japan
| | - Jacques Colinge
- Cancer Bioinformatics and System Biology, Institute of Cancer Research of Montpellier (IRCM), Inserm, University of Montpellier, ICM, 34298, Montpellier, France
| | - Muneaki Shimada
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
| | - Satoshi Takeuchi
- Department of Gynecology, Kobe Tokushukai Hospital, Kobe, Hyogo, 655-0017, Japan
- Department of Obstetrics and Gynecology, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hidetaka Eguchi
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
- Diagnosis and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Tetsuro Oishi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
- Department of Obstetrics and Gynecology, Matsue City Hospital, Matsue, Shimane, 690-8509, Japan
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
- Department of Obstetrics and Gynecology, St. Mary's Hospital, Kurume, Fukuoka, 830-8543, Japan
| | - Mitsuaki Suzuki
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Masahiko Nishiyama
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma, 371-8511, Japan.
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan.
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan.
- Laboratory for Analytical Instruments, Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
8
|
Shaaban A, Scott SS, Greenlee AN, Binda N, Noor A, Webb A, Guo S, Purdy N, Pennza N, Habib A, Mohammad SJ, Smith SA. Atrial fibrillation in cancer, anticancer therapies, and underlying mechanisms. J Mol Cell Cardiol 2024; 194:118-132. [PMID: 38897563 PMCID: PMC11500699 DOI: 10.1016/j.yjmcc.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Atrial fibrillation (AF) is a common arrhythmic complication in cancer patients and can be exacerbated by traditional cytotoxic and targeted anticancer therapies. Increased incidence of AF in cancer patients is independent of confounding factors, including preexisting myocardial arrhythmogenic substrates, type of cancer, or cancer stage. Mechanistically, AF is characterized by fast unsynchronized atrial contractions with rapid ventricular response, which impairs ventricular filling and results in various symptoms such as fatigue, chest pain, and shortness of breath. Due to increased blood stasis, a consequence of both cancer and AF, concern for stroke increases in this patient population. To compound matters, cardiotoxic anticancer therapies themselves promote AF; thereby exacerbating AF morbidity and mortality in cancer patients. In this review, we examine the relationship between AF, cancer, and cardiotoxic anticancer therapies with a focus on the shared molecular and electrophysiological mechanisms linking these disease processes. We also explore the potential role of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in the management of anticancer-therapy-induced AF.
Collapse
Affiliation(s)
- Adnan Shaaban
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Shane S Scott
- Medical Scientist Training Program, Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ashley N Greenlee
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nkongho Binda
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Ali Noor
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Averie Webb
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Shuliang Guo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Najhee Purdy
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nicholas Pennza
- Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Alma Habib
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA
| | - Somayya J Mohammad
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sakima A Smith
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
9
|
Agnes RS, Traughber BJ, Muzic RF. Development of a selective novel fluorescent substrate for sodium-dependent transporters. Life Sci 2024; 351:122847. [PMID: 38880166 DOI: 10.1016/j.lfs.2024.122847] [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: 03/27/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
AIM To synthesize, characterize, and validate 6FGA, a fluorescent glucose modified with a Cyanine5.5 at carbon-6 position, for probing the function of sodium-dependent glucose transporters, SGLT1 and SGLT2. MAIN METHODS The synthesis of fluorescent glucose analogue was achieved through "click chemistry" of Cyanine5.5-alkyne and 6-azido-6-deoxy-d-glucose. Cell system studies were conducted to characterize the in vivo transport properties. KEY FINDINGS Optical analyses revealed that 6FGA displayed similar spectral profiles to Cyanine5.5 in DMSO, allowing for concentration determination, thus supporting its utility in quantitative kinetic studies within biological assays. Uptake studies in cell system SGLT models, LLC-PK1 and HEK293 cells, exhibited concentration and time-dependent behavior, indicating saturation at specific concentrations and durations which are hallmarks of transported-mediated uptake. The results of cytotoxicity assays suggested cell viability at micromolar concentrations, enabling usage in assays for at least 1 h without significant toxicity. The dependence of 6FGA uptake on sodium, the co-transported cation, was demonstrated in LLC-PK1 and HEK293 cells. Fluorescence microscopy confirmed intracellular localization of 6FGA, particularly near the nucleus. Competition studies revealed that glucose tends to weakly reduce 6FGA uptake, although the effect did not achieve statistical significance. Assessments using standard SGLT and GLUT inhibitors highlighted 6FGA's sensitivity for probing SGLT-mediated transport. SIGNIFICANCE 6FGA is a new fluorescent glucose analog offering advantages over existing probes due to its improved photophysical properties, greater sensitivity, enabling subcellular resolution and efficient tissue penetration in near-infrared imaging. 6FGA presents practicality and cost-effectiveness, making it a promising tool for nonradioactive, microplate-based assays at investigating SGLT-mediated glucose transport mechanisms.
Collapse
Affiliation(s)
- Richard S Agnes
- Department of Radiology, University Hospitals of Cleveland and Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Bryan J Traughber
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Raymond F Muzic
- Department of Radiology, University Hospitals of Cleveland and Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
10
|
Lin Y, Zhang Y, Wang S, Cao L, Zhao R, Ma X, Yang Q, Zhang L, Yang Q. Pharmacological targets of SGLT2 inhibition on prostate cancer mediated by circulating metabolites: a drug-target Mendelian randomization study. Front Pharmacol 2024; 15:1443045. [PMID: 39166104 PMCID: PMC11333260 DOI: 10.3389/fphar.2024.1443045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024] Open
Abstract
Background The relationship between sodium-glucose cotransporter 2 (SGLT2) inhibitors and prostate cancer is still unknown. Although these inhibitors can influence tumor glycolysis, the underlying mechanism requires further exploration. Methods A two-sample two-step MR was used to determine 1) causal effects of SGLT2 inhibition on prostate cancer; 2) causal effects of 1,400 circulating metabolites or metabolite ratios on prostate cancer; and 3) mediation effects of these circulating metabolites. Genetic proxies for SGLT2 inhibition were identified as variants in the SLC5A2 gene and glycated hemoglobin level (HbA1c). Additionally, positive control analysis on type 2 diabetes mellitus (T2DM) was conducted to test the selection of genetic proxies. Phenome Wide Association Study (PheWAS) and MR-PheWAS analysis were used to explore potential treatable diseases and adverse outcomes of SGLT2 inhibitors. Results Genetically predicted SGLT2 inhibition (per 1 SD decrement in HbA1c) was associated with reduced risk of T2DM [odds ratio (OR) = 0.66 (95% CI 0.53, 0.82), P = 1.57 × 10-4]; prostate cancer [0.34 (0.23, 0.49), P = 2.21 × 10-8] and prostate-specific antigen [0.26 (0.08, 0.81), P = 2.07 × 10-2]. The effect of SGLT2 inhibition on prostate cancer was mediated by uridine level, with a mediated proportion of 9.34% of the total effect. In MR-PheWAS, 65 traits were found to be associated with SLGT2 inhibitors (P < 1.78 × 10-5), and among them, 13 were related to diabetes. Conclusion Our study suggested that SGLT2 inhibition could lower prostate cancer risk through uridine mediation. More mechanistic and clinical research is necessary to explore how uridine mediates the link between SGLT2 inhibition and prostate cancer.
Collapse
Affiliation(s)
- Yilong Lin
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Yue Zhang
- Department of Hematology, Xiangya Hospital, Xiangya School of Medicine, Central South University, Changsha, China
| | - Songsong Wang
- School of Medicine, Xiamen University, Xiamen, China
| | - Lin Cao
- The First Clinical College of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ruidan Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xilai Ma
- School of Medicine, Xiamen University, Xiamen, China
| | - Qiaolu Yang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Liyi Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qingmo Yang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Ionică LN, Buriman DG, Lința AV, Șoșdean R, Lascu A, Streian CG, Feier HB, Petrescu L, Mozoș IM, Sturza A, Muntean DM. Empagliflozin and dapagliflozin decreased atrial monoamine oxidase expression and alleviated oxidative stress in overweight non-diabetic cardiac patients. Mol Cell Biochem 2024:10.1007/s11010-024-05076-z. [PMID: 39042348 DOI: 10.1007/s11010-024-05076-z] [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: 05/24/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
The sodium-glucose-cotransporter 2 inhibitors (SGLT2i) are the blockbuster antidiabetic drugs that exert cardiovascular protection via pleiotropic effects. We have previously demonstrated that empagliflozin decreased monoamine oxidase (MAO) expression and oxidative stress in human mammary arteries. The present study performed in overweight, non-diabetic cardiac patients was aimed to assess whether the two widely prescribed SGLT2i decrease atrial MAO expression and alleviate oxidative stress elicited by exposure to angiotensin 2 (ANG2) and high glucose (GLUC). Right atrial appendages isolated during cardiac surgery were incubated ex vivo with either empagliflozin or dapagliflozin (1, 10 µm, 12 h) in the presence or absence of ANG2 (100 nm) and GLUC (400 mg/dL) and used for the evaluation of MAO-A and MAO-B expression and ROS production. Stimulation with ANG2 and GLUC increased atrial expression of both MAOs and oxidative stress; the effects were significantly decreased by the SGLT2i. Atrial oxidative stress positively correlated with the echocardiographic size of heart chambers and negatively with the left ventricular ejection fraction. In overweight patients, MAO contributes to cardiac oxidative stress in basal conditions and those that mimicked the renin-angiotensin system activation and hyperglycemia and can be targeted with empagliflozin and dapagliflozin, as novel off-target class effect of the SGLT2i.
Collapse
Affiliation(s)
- Loredana N Ionică
- Doctoral School Medicine-Pharmacy, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department V Internal Medicine - 1st Clinic of Medical Semiotics, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Darius G Buriman
- Doctoral School Medicine-Pharmacy, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department III Functional Sciences - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Adina V Lința
- Doctoral School Medicine-Pharmacy, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department III Functional Sciences - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Raluca Șoșdean
- Department VI Cardiology, Clinic of Cardiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Institute of Cardiovascular Diseases Timișoara, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Ana Lascu
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department III Functional Sciences - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Institute of Cardiovascular Diseases Timișoara, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Caius G Streian
- Institute of Cardiovascular Diseases Timișoara, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department VI Cardiology - Clinic of Cardiovascular Surgery, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Horea B Feier
- Institute of Cardiovascular Diseases Timișoara, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department VI Cardiology - Clinic of Cardiovascular Surgery, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Lucian Petrescu
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Ioana M Mozoș
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department III Functional Sciences - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| | - Adrian Sturza
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania.
- Department III Functional Sciences - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania.
| | - Danina M Muntean
- Centre for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
- Department III Functional Sciences - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy From Timișoara, Eftimie Murgu Sq. No. 2, 300041, Timișoara, Romania
| |
Collapse
|
12
|
Duan HY, Barajas-Martinez H, Antzelevitch C, Hu D. The potential anti-arrhythmic effect of SGLT2 inhibitors. Cardiovasc Diabetol 2024; 23:252. [PMID: 39010053 PMCID: PMC11251349 DOI: 10.1186/s12933-024-02312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/16/2024] [Indexed: 07/17/2024] Open
Abstract
Sodium-glucose cotransporter type 2 inhibitors (SGLT2i) were initially recommended as oral anti-diabetic drugs to treat type 2 diabetes (T2D), by inhibiting SGLT2 in proximal tubule and reduce renal reabsorption of sodium and glucose. While many clinical trials demonstrated the tremendous potential of SGLT2i for cardiovascular diseases. 2022 AHA/ACC/HFSA guideline first emphasized that SGLT2i were the only drug class that can cover the entire management of heart failure (HF) from prevention to treatment. Subsequently, the antiarrhythmic properties of SGLT2i have also attracted attention. Although there are currently no prospective studies specifically on the anti-arrhythmic effects of SGLT2i. We provide clues from clinical and fundamental researches to identify its antiarrhythmic effects, reviewing the evidences and mechanism for the SGLT2i antiarrhythmic effects and establishing a novel paradigm involving intracellular sodium, metabolism and autophagy to investigate the potential mechanisms of SGLT2i in mitigating arrhythmias.
Collapse
Affiliation(s)
- Hong-Yi Duan
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, Hubei, China
| | - Hector Barajas-Martinez
- Lankenau Institute for Medical Research, Lankenau Heart Institute, Wynnewood, PA, 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, 19107, USA
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Lankenau Heart Institute, Wynnewood, PA, 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, 19107, USA
| | - Dan Hu
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, Hubei, China.
| |
Collapse
|
13
|
Jiang Y, Yang J, Xia L, Wei T, Cui X, Wang D, Jin Z, Lin X, Li F, Yang K, Lang S, Liu Y, Hang J, Zhang Z, Hong T, Wei R. Gut Microbiota-Tryptophan Metabolism-GLP-1 Axis Participates in β-Cell Regeneration Induced by Dapagliflozin. Diabetes 2024; 73:926-940. [PMID: 38471012 PMCID: PMC11109800 DOI: 10.2337/db23-0553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors, efficacious antidiabetic agents that have cardiovascular and renal benefits, can promote pancreatic β-cell regeneration in type 2 diabetic mice. However, the underlying mechanism remains unclear. In this study, we aimed to use multiomics to identify the mediators involved in β-cell regeneration induced by dapagliflozin. We showed that dapagliflozin lowered blood glucose level, upregulated plasma insulin level, and increased islet area in db/db mice. Dapagliflozin reshaped gut microbiota and modulated microbiotic and plasmatic metabolites related to tryptophan metabolism, especially l-tryptophan, in the diabetic mice. Notably, l-tryptophan upregulated the mRNA level of glucagon-like peptide 1 (GLP-1) production-related gene (Gcg and Pcsk1) expression and promoted GLP-1 secretion in cultured mouse intestinal L cells, and it increased the supernatant insulin level in primary human islets, which was eliminated by GPR142 antagonist. Transplant of fecal microbiota from dapagliflozin-treated mice, supplementation of l-tryptophan, or treatment with dapagliflozin upregulated l-tryptophan, GLP-1, and insulin or C-peptide levels and promoted β-cell regeneration in db/db mice. Addition of exendin 9-39, a GLP-1 receptor (GLP-1R) antagonist, or pancreatic Glp1r knockout diminished these beneficial effects. In summary, treatment with dapagliflozin in type 2 diabetic mice promotes β-cell regeneration by upregulating GLP-1 production, which is mediated via gut microbiota and tryptophan metabolism. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yafei Jiang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Li Xia
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Tianjiao Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Xiaona Cui
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Dandan Wang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Zirun Jin
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Xiafang Lin
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Fei Li
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Kun Yang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Shan Lang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Ye Liu
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
| | - Jing Hang
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing, China
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Rui Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
14
|
Liu Z, Wang J, Tian P, Liu Y, Xing L, Fu C, Huang X, Liu P. Sodium-glucose cotransporter 1 promotes the biofunctions of perivascular preadipocytes mediated by Akt/mTOR/p70S6K signaling pathway. Am J Physiol Cell Physiol 2024; 326:C1611-C1624. [PMID: 38646789 PMCID: PMC11371362 DOI: 10.1152/ajpcell.00606.2023] [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: 11/09/2023] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/23/2024]
Abstract
The influence of SGLT-1 on perivascular preadipocytes (PVPACs) and vascular remodeling is not well understood. This study aimed to elucidate the role and mechanism of SGLT-1-mediated PVPACs bioactivity. PVPACs were cultured in vitro and applied ex vivo to the carotid arteries of mice using a lentivirus-based thermosensitive in situ gel (TISG). The groups were treated with Lv-SGLT1 (lentiviral vector, overexpression), Lv-siSGLT1 (RNA interference, knockdown), or specific signaling pathway inhibitors. Assays were conducted to assess changes in cell proliferation, apoptosis, glucose uptake, adipogenic differentiation, and vascular remodeling in the PVPACs. Protein expression was analyzed by Western blotting, immunocytochemistry, and/or immunohistochemistry. The methyl thiazolyl tetrazolium (MTT) assay and Hoechst 33342 staining indicated that SGLT-1 overexpression significantly promoted PVPACs proliferation and inhibited apoptosis in vitro. Conversely, SGLT-1 knockdown exerted the opposite effect. Oil Red O staining revealed that SGLT-1 overexpression facilitated adipogenic differentiation, while its inhibition mitigated these effects. 3H-labeled glucose uptake experiments demonstrated that SGLT-1 overexpression enhanced glucose uptake by PVPACs, whereas RNA interference-mediated SGLT-1 inhibition had no significant effect on glucose uptake. Moreover, RT-qPCR, Western blotting, and immunofluorescence analyses revealed that SGLT-1 overexpression upregulated FABP4 and VEGF-A levels and activated the Akt/mTOR/p70S6K signaling pathway, whereas SGLT-1 knockdown produced the opposite effects. In vivo studies corroborated these findings and indicated that SGLT-1 overexpression facilitated carotid artery remodeling. Our study demonstrates that SGLT-1 activation of the Akt/mTOR/p70S6K signaling pathway promotes PVPACs proliferation, adipogenesis, glucose uptake, glucolipid metabolism, and vascular remodeling.NEW & NOTEWORTHY SGLT-1 is expressed in PVPACs and can affect preadipocyte glucolipid metabolism and vascular remodeling. SGLT-1 promotes the biofunctions of PVPACs mediated by Akt/mTOR/p70S6K signaling pathway. Compared with caudal vein or intraperitoneal injection, the external application of lentivirus-based thermal gel around the carotid artery is an innovative attempt at vascular remodeling model, it may effectively avoid the transfection of lentiviral vector into the whole body of mice and the adverse effect on experimental results.
Collapse
Affiliation(s)
- Zhiquan Liu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
- Division of Life Sciences and Medicine, Department of Cardiology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jiayu Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| | - Peiqing Tian
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| | - Yixuan Liu
- Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Liyun Xing
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| | - Caihua Fu
- Department of Cardiology, Jinan Central Hospital Affiliated Shandong University, Jinan, China
| | - Xianwei Huang
- Department of Emergency, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ping Liu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
15
|
Zhang C, Wang Y, Guo X, Wang Z, Xiao J, Liu Z. SLC7A5 correlated with malignancies and immunotherapy response in bladder cancer. Cancer Cell Int 2024; 24:182. [PMID: 38790003 PMCID: PMC11127462 DOI: 10.1186/s12935-024-03365-7] [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: 02/23/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Metabolic reprogramming contributes to bladder cancer development. This study aimed to understand the role of SLC7A5 in bladder cancer. METHODS We systematically analyzed the correlation between SLC7A5 and bladder cancer through various approaches, including bioinformatics, western blotting, cell cycle analysis, cell proliferation assays, and invasion experiments. We also investigated the immunological features within the tumor microenvironment (TME), encompassing cancer immune cycles, immune modulators, immune checkpoints, tumor-infiltrating immune cells (TIIC), T cell inflammation scores, and treatment responses. Additionally, for a comprehensive assessment of the expression patterns and immunological roles of SLC7A5, pan-cancer analysis was performed using cancer genomics datasets. RESULTS SLC7A5 was associated with adverse prognosis in bladder cancer patients, activating the Wnt pathway and promoting bladder cancer cell cycle progression, proliferation, migration, and invasion. Based on the evidence that SLC7A5 positively correlated with immunomodulators, TIIC, the cancer immune cycle, immune checkpoint and T cell inflammation scores, we also found that SLC7A5 was associated with the inflammatory tumor immune microenvironment. EGFR-targeted therapy, cancer immunotherapy, and radiation therapy were effective for patients with high SLC7A5 expression in bladder cancer. Low SLC7A5 patients were, however, sensitive to targeted therapies and anti-angiogenic therapy, such as blocking β-catenin network, PPAR-γ and FGFR3 signaling. Anti-SLC7A5 combined with cancer immunotherapy may have greater effectiveness than either therapy alone. Furthermore, we observed specific overexpression of SLC7A5 in TME of various cancers. CONCLUSION SLC7A5 can predict therapeutic response to immunotherapy, radiotherapy and chemotherapy in bladder cancer patients. Targeting SLC7A5 in combination with immunotherapy may be a potentially appropriate treatment option.
Collapse
Affiliation(s)
- Chunyu Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiatong Xiao
- Departments of Urology, Xiangya Hospital, Central South University, Changsha, China.
| | - Zhi Liu
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
16
|
Huang H, Kung FL, Huang YW, Hsu CC, Guh JH, Hsu LC. Sensitization of cancer cells to paclitaxel-induced apoptosis by canagliflozin. Biochem Pharmacol 2024; 223:116140. [PMID: 38513740 DOI: 10.1016/j.bcp.2024.116140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
Cancer cells consume more glucose and usually overexpress glucose transporters which have become potential targets for the development of anticancer drugs. It has been demonstrated that selective SGLT2 inhibitors, such as canagliflozin and dapagliflozin, display anticancer activity. Here we demonstrated that canagliflozin and dapagliflozin synergistically enhanced the growth inhibitory effect of paclitaxel in cancer cells including ovarian cancer and oral squamous cell carcinoma cells. Canagliflozin also inhibited glucose uptake via GLUTs. The combination of paclitaxel and WZB117, a GLUT inhibitor, exhibited a strong synergy, supporting the notion that inhibition of GLUTs by canagliflozin may also account for the synergy between canagliflozin and paclitaxel. Mechanistic studies in ES-2 ovarian cancer cells revealed that canagliflozin potentiated paclitaxel-induced apoptosis and DNA damaging effect. Paclitaxel in the nanomolar range elevated abnormal mitotic cells as well as aneuploid cells, and canagliflozin further enhanced this effect. Furthermore, canagliflozin downregulated cyclin B1 and phospho-BUBR1 upon spindle assembly checkpoint (SAC) activation by paclitaxel, and may consequently impair SAC. Thus, paclitaxel disturbed microtubule dynamics and canagliflozin compromised SAC activity, together they may induce premature mitotic exit, accumulation of aneuploid cells with DNA damage, and ultimately apoptosis.
Collapse
Affiliation(s)
- Haoning Huang
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Fan-Lu Kung
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Yu-Wen Huang
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Chun-Chien Hsu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan.
| |
Collapse
|
17
|
Chen X, Yu X, Lian G, Tang H, Yan Y, Gao G, Huang B, Luo L, Xie L. Canagliflozin inhibits PASMCs proliferation via regulating SGLT1/AMPK signaling and attenuates artery remodeling in MCT-induced pulmonary arterial hypertension. Biomed Pharmacother 2024; 174:116505. [PMID: 38574614 DOI: 10.1016/j.biopha.2024.116505] [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: 01/09/2024] [Revised: 03/14/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) was a devastating disease characterized by artery remodeling, ultimately resulting in right heart failure. The aim of this study was to investigate the effects of canagliflozin (CANA), a sodium-glucose cotransporter 2 inhibitor (SGLT2i) with mild SGLT1 inhibitory effects, on rats with PAH, as well as its direct impact on pulmonary arterial smooth muscle cells (PASMCs). PAH rats were induced by injection of monocrotaline (MCT) (40 mg/kg), followed by four weeks of treatment with CANA (30 mg/kg/day) or saline alone. Pulmonary artery and right ventricular (RV) remodeling and dysfunction in PAH were alleviated with CANA, as assessed by echocardiography. Hemodynamic parameters and structural of pulmonary arteriole, including vascular wall thickness and wall area, were reduced by CANA. RV hypertrophy index, cardiomyocyte hypertrophy, and fibrosis were decreased with CANA treatment. PASMCs proliferation was inhibited by CANA under stimulation by platelet-derived growth factor (PDGF)-BB or hypoxia. Activation of AMP kinase (AMPK) was induced by CANA treatment in cultured PASMCs in a time- and concentration-dependent manner. These effects of CANA were attenuated when treatment with compound C, an AMPK inhibitor. Abundant expression of SGLT1 was observed in PASMCs and pulmonary arteries, while SGLT2 expression was undetectable. SGLT1 increased in response to PDGF-BB or hypoxia stimulation, while PASMCs proliferation was inhibited and beneficial effects of CANA were counteracted by knockdown of SGLT1. Our research demonstrated for the first time that CANA inhibited the proliferation of PASMCs by regulating SGLT1/AMPK signaling and thus exerted an anti-proliferative effect on MCT-induced PAH.
Collapse
Affiliation(s)
- Xiaojun Chen
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xing Yu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Guili Lian
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huibin Tang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yan Yan
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Gufeng Gao
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Bangbang Huang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
18
|
Fujiyoshi S, Honda S, Ara M, Kondo T, Kobayashi N, Taketomi A. SGLT2 is upregulated to acquire cisplatin resistance and SGLT2 inhibition reduces cisplatin resistance in hepatoblastoma. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:223-233. [PMID: 37927142 DOI: 10.1002/jhbp.1391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
BACKGROUND Cancer cells can alter glucose metabolism and regulate the expression of glucose transporters. Hepatoblastoma patients undergo cisplatin-based chemotherapy; however, 22.3% of patients develop cisplatin resistance and thus face a poor prognosis. We hypothesized that glucose transporters are associated with acquiring cisplatin resistance with increasing sugar intake inhibiting glucose transporters could reduce cisplatin resistance in hepatoblastoma patients. METHODS We established cisplatin-resistant HepG2 and HuH6 cells by continuous treatment with cisplatin. We evaluated the relationship between cisplatin resistance and glucose uptake. We used an expression array to select cisplatin-resistant associated glucose transporters and selected sodium-glucose cotransporter 2 (SGLT2). We used dapagliflozin as an SGLT2 inhibitor and evaluated glucose uptake and IC50 after dapagliflozin treatment in wild-type and resistant hepatoblastoma cells in vitro and in vivo. RESULTS We found a strong relationship between cisplatin resistance and glucose uptake. Additionally, SGLT2 was upregulated in resistant cells after cisplatin treatment. After dapagliflozin treatment, glucose uptake and cisplatin resistance decreased in resistant cells. CONCLUSIONS Cisplatin-resistant hepatoblastoma cells exhibited upregulated SGLT2 expression and activated glucose uptake to survive under cisplatin stress. SGLT2 inhibition decreased cellular resistance to cisplatin. SGLT2 inhibition with cisplatin therapy could be a novel therapeutic strategy for cisplatin-resistant hepatoblastoma patients.
Collapse
Affiliation(s)
- Sunao Fujiyoshi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Shohei Honda
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Momoko Ara
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Takafumi Kondo
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Nozomi Kobayashi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
19
|
Sun M, Sun J, Sun W, Li X, Wang Z, Sun L, Wang Y. Unveiling the anticancer effects of SGLT-2i: mechanisms and therapeutic potential. Front Pharmacol 2024; 15:1369352. [PMID: 38595915 PMCID: PMC11002155 DOI: 10.3389/fphar.2024.1369352] [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: 01/12/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
Cancer and diabetes are significant diseases that pose a threat to human health. Their interconnection is complex, particularly when they coexist, often necessitating multiple therapeutic approaches to attain remission. Sodium-glucose cotransporter protein two inhibitors (SGLT-2i) emerged as a treatment for hyperglycemia, but subsequently exhibited noteworthy extra-glycemic properties, such as being registered for the treatment of heart failure and chronic kidney disease, especially with co-existing albuminuria, prompting its assessment as a potential treatment for various non-metabolic diseases. Considering its overall tolerability and established use in diabetes management, SGLT-2i may be a promising candidate for cancer therapy and as a supplementary component to conventional treatments. This narrative review aimed to examine the potential roles and mechanisms of SGLT-2i in the management of diverse types of cancer. Future investigations should focus on elucidating the antitumor efficacy of individual SGLT-2i in different cancer types and exploring the underlying mechanisms. Additionally, clinical trials to evaluate the safety and feasibility of incorporating SGLT-2i into the treatment regimen of specific cancer patients and determining appropriate dosage combinations with established antitumor agents would be of significant interest.
Collapse
Affiliation(s)
- Min Sun
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| | - Jilei Sun
- Changchun Traditional Chinese Medicine Hospital, Changchun, China
| | - Wei Sun
- First Affiliated Hospital of Jilin University, Changchun, China
| | - Xiaonan Li
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| | - Zhe Wang
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
20
|
Yu XW, She PW, Chen FC, Chen YY, Zhou S, Wang XM, Lin XR, Liu QL, Huang ZJ, Qiu Y. Metabolic subtypes and immune landscapes in esophageal squamous cell carcinoma: prognostic implications and potential for personalized therapies. BMC Cancer 2024; 24:230. [PMID: 38373930 PMCID: PMC10875771 DOI: 10.1186/s12885-024-11890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND This study aimed to identify metabolic subtypes in ESCA, explore their relationship with immune landscapes, and establish a metabolic index for accurate prognosis assessment. METHODS Clinical, SNP, and RNA-seq data were collected from 80 ESCA patients from the TCGA database and RNA-seq data from the GSE19417 dataset. Metabolic genes associated with overall survival (OS) and progression-free survival (PFS) were selected, and k-means clustering was performed. Immune-related pathways, immune infiltration, and response to immunotherapy were predicted using bioinformatic algorithms. Weighted gene co-expression network analysis (WGCNA) was conducted to identify metabolic genes associated with co-expression modules. Lastly, cell culture and functional analysis were performed using patient tissue samples and ESCA cell lines to verify the identified genes and their roles. RESULTS Molecular subtypes were identified based on the expression profiles of metabolic genes, and univariate survival analysis revealed 163 metabolic genes associated with ESCA prognosis. Consensus clustering analysis classified ESCA samples into three distinct subtypes, with MC1 showing the poorest prognosis and MC3 having the best prognosis. The subtypes also exhibited significant differences in immune cell infiltration, with MC3 showing the highest scores. Additionally, the MC3 subtype demonstrated the poorest response to immunotherapy, while the MC1 subtype was the most sensitive. WGCNA analysis identified gene modules associated with the metabolic index, with SLC5A1, NT5DC4, and MTHFD2 emerging as prognostic markers. Gene and protein expression analysis validated the upregulation of MTHFD2 in ESCA. MTHFD2 promotes the progression of ESCA and may be a potential therapeutic target for ESCA. CONCLUSION The established metabolic index and identified metabolic genes offer potential for prognostic assessment and personalized therapeutic interventions for ESCA, underscoring the importance of targeting metabolism-immune interactions in ESCA. MTHFD2 promotes the progression of ESCA and may be a potential therapeutic target for ESCA.
Collapse
Affiliation(s)
- Xiao-Wan Yu
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China.
| | - Pei-Wei She
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, P. R. China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, 350001, Fuzhou, Fujian, P. R. China
| | - Fang-Chuan Chen
- Stomatology Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Ya-Yu Chen
- Stomatology Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Shuang Zhou
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Xi-Min Wang
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Xiao-Rong Lin
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Qiao-Ling Liu
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Zhi-Jun Huang
- Esophageal Surgery Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Yu Qiu
- Reproductive Center, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China.
| |
Collapse
|
21
|
Vaziri Z, Saleki K, Aram C, Alijanizadeh P, Pourahmad R, Azadmehr A, Ziaei N. Empagliflozin treatment of cardiotoxicity: A comprehensive review of clinical, immunobiological, neuroimmune, and therapeutic implications. Biomed Pharmacother 2023; 168:115686. [PMID: 37839109 DOI: 10.1016/j.biopha.2023.115686] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
Cancer and cardiovascular disorders are known as the two main leading causes of mortality worldwide. Cardiotoxicity is a critical and common adverse effect of cancer-related chemotherapy. Chemotherapy-induced cardiotoxicity has been associated with various cancer treatments, such as anthracyclines, immune checkpoint inhibitors, and kinase inhibitors. Different methods have been reported for the management of chemotherapy-induced cardiotoxicity. In this regard, sodium-glucose cotransporter-2 inhibitors (SGLT2i), a class of antidiabetic agents, have recently been applied to manage heart failure patients. Further, SGLT2i drugs such as EMPA exert protective cardiac and systemic effects. Moreover, it can reduce inflammation through the mediation of major inflammatory components, such as Nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasomes, Adenosine 5'-monophosphate-activated protein kinase (AMPK), and c-Jun N-terminal kinase (JNK) pathways, Signal transducer and activator of transcription (STAT), and overall decreasing transcription of proinflammatory cytokines. The clinical outcome of EMPA administration is related to improving cardiovascular risk factors, including body weight, lipid profile, blood pressure, and arterial stiffness. Intriguingly, SGLT2 suppressors can regulate microglia-driven hyperinflammation affecting neurological and cardiovascular disorders. In this review, we discuss the protective effects of EMPA in chemotherapy-induced cardiotoxicity from molecular, immunological, and neuroimmunological aspects to preclinical and clinical outcomes.
Collapse
Affiliation(s)
- Zahra Vaziri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Cena Aram
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Ramtin Pourahmad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran
| | - Naghmeh Ziaei
- Clinical Research Development unit of Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran; Department of Cardiology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
22
|
Azizogli AR, Vitti MR, Mishra R, Osorno L, Heffernan C, Kumar VA. Comparison of SGLT1, SGLT2, and Dual Inhibitor biological activity in treating Type 2 Diabetes Mellitus. ADVANCED THERAPEUTICS 2023; 6:2300143. [PMID: 38223846 PMCID: PMC10783160 DOI: 10.1002/adtp.202300143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Indexed: 01/16/2024]
Abstract
Diabetes Mellitus Type 2 (T2D) is an emerging health burden in the USand worldwide, impacting approximately 15% of Americans. Current front-line therapeutics for T2D patients include sulfonylureas that act to reduce A1C and/or fasting blood glucose levels, or Metformin that antagonizes the action of glucagon to reduce hepatic glucose production. Next generation glucomodulatory therapeutics target members of the high-affinity glucose transporter Sodium-Glucose-Linked-Transporter (SGLT) family. SGLT1 is primarily expressed in intestinal epithelium, whose inhibition reduces dietary glucose uptake, whilst SGLT2 is highly expressed in kidney - regulating glucose reabsorption. A number of SGLT2 inhibitors are FDA approved whilst SGLT1 and dual SGLT1 & 2 inhibitor are currently in clinical trials. Here, we discuss and compare SGLT2, SGLT1, and dual inhibitors' biochemical mechanism and physiological effects.
Collapse
Affiliation(s)
- Abdul-Rahman Azizogli
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Michael R Vitti
- University of Virginia School of Medicine, Charlottesville, VA, 22903
| | - Richa Mishra
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Laura Osorno
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103
| |
Collapse
|
23
|
Xu B, Li S, Kang B, Fan S, Chen C, Li W, Chen J, He Z, Tang F, Zhou J. Role of SLC5A2 polymorphisms and effects of genetic polymorphism on sodium glucose cotransporter 2 inhibitorsinhibitor response. Mol Biol Rep 2023; 50:9637-9647. [PMID: 37819499 DOI: 10.1007/s11033-023-08836-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a complex metabolic disease characterized by hyperglycaemia. T2DM is a highly heterogeneous polygenic disease. Due to genetic variation, variations in lifestyle and other environmental exposures, there are certain variations in the phenotype of T2DM patients. Sodium glucose cotransporter 2 (SGLT2) inhibitors are novel hypoglycaemic agents that increase urinary glucose excretion by inhibiting glucose reabsorption in the proximal tubules of the kidney. For glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, studies have confirmed a variety of gene variants that may modify their effects. For SGLT2 inhibitors, research has focused on the SLC5A2 gene encoding SGLT2 and UGT1A9 gene polymorphisms affecting SGLT2 inhibitor metabolism. The SLC5A2 polymorphism rs9934336 have been associated with decreased HbA1c during the oral glucose tolerance test. Common variants of the SLC5A2 gene are related to blood glucose and insulin concentrations, but not glucagon concentrations. SLC5A2 rs9934336 and rs3116150 are related to a lower risk of heart failure. SGLT2 inhibitor exposure of UGT1A9*3 carriers is commonly higher than that of noncarriers, while these effects commonly have no obvious clinical significance on SGLT2 inhibitor pharmacokinetics. In terms of efficacy, general SLC5A2 variants show no significant effect on the response to the SGLT2 inhibitor empagliflozin. At present, research on the relationship between genetic polymorphisms and the efficacy of SGLT2 inhibitors is limited. The main purpose of this review is to elucidate the general effects of SGLT2 polymorphisms and the association between polymorphisms and the treatment response to SGLT2 inhibitors.
Collapse
Affiliation(s)
- Bo Xu
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shaoqian Li
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Kang
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shangzhi Fan
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Canyu Chen
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Weiyi Li
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zunbo He
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Fan Tang
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
24
|
Saha S, Fang X, Green CD, Das A. mTORC1 and SGLT2 Inhibitors-A Therapeutic Perspective for Diabetic Cardiomyopathy. Int J Mol Sci 2023; 24:15078. [PMID: 37894760 PMCID: PMC10606418 DOI: 10.3390/ijms242015078] [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: 09/01/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic cardiomyopathy is a critical diabetes-mediated co-morbidity characterized by cardiac dysfunction and heart failure, without predisposing hypertensive or atherosclerotic conditions. Metabolic insulin resistance, promoting hyperglycemia and hyperlipidemia, is the primary cause of diabetes-related disorders, but ambiguous tissue-specific insulin sensitivity has shed light on the importance of identifying a unified target paradigm for both the glycemic and non-glycemic context of type 2 diabetes (T2D). Several studies have indicated hyperactivation of the mammalian target of rapamycin (mTOR), specifically complex 1 (mTORC1), as a critical mediator of T2D pathophysiology by promoting insulin resistance, hyperlipidemia, inflammation, vasoconstriction, and stress. Moreover, mTORC1 inhibitors like rapamycin and their analogs have shown significant benefits in diabetes and related cardiac dysfunction. Recently, FDA-approved anti-hyperglycemic sodium-glucose co-transporter 2 inhibitors (SGLT2is) have gained therapeutic popularity for T2D and diabetic cardiomyopathy, even acknowledging the absence of SGLT2 channels in the heart. Recent studies have proposed SGLT2-independent drug mechanisms to ascertain their cardioprotective benefits by regulating sodium homeostasis and mimicking energy deprivation. In this review, we systematically discuss the role of mTORC1 as a unified, eminent target to treat T2D-mediated cardiac dysfunction and scrutinize whether SGLT2is can target mTORC1 signaling to benefit patients with diabetic cardiomyopathy. Further studies are warranted to establish the underlying cardioprotective mechanisms of SGLT2is under diabetic conditions, with selective inhibition of cardiac mTORC1 but the concomitant activation of mTORC2 (mTOR complex 2) signaling.
Collapse
Affiliation(s)
- Sumit Saha
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Christopher D. Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
25
|
Schlessinger A, Zatorski N, Hutchinson K, Colas C. Targeting SLC transporters: small molecules as modulators and therapeutic opportunities. Trends Biochem Sci 2023; 48:801-814. [PMID: 37355450 PMCID: PMC10525040 DOI: 10.1016/j.tibs.2023.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/26/2023]
Abstract
Solute carrier (SLCs) transporters mediate the transport of a broad range of solutes across biological membranes. Dysregulation of SLCs has been associated with various pathologies, including metabolic and neurological disorders, as well as cancer and rare diseases. SLCs are therefore emerging as key targets for therapeutic intervention with several recently approved drugs targeting these proteins. Unlocking this large and complex group of proteins is essential to identifying unknown SLC targets and developing next-generation SLC therapeutics. Recent progress in experimental and computational techniques has significantly advanced SLC research, including drug discovery. Here, we review emerging topics in therapeutic discovery of SLCs, focusing on state-of-the-art approaches in structural, chemical, and computational biology, and discuss current challenges in transporter drug discovery.
Collapse
Affiliation(s)
- Avner Schlessinger
- Department of Pharmacological Sciences Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Nicole Zatorski
- Department of Pharmacological Sciences Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keino Hutchinson
- Department of Pharmacological Sciences Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Claire Colas
- University of Vienna, Department of Pharmaceutical Chemistry, Vienna, Austria.
| |
Collapse
|
26
|
Pillai U J, Ray A, Maan M, Dutta M. Repurposing drugs targeting metabolic diseases for cancer therapeutics. Drug Discov Today 2023; 28:103684. [PMID: 37379903 DOI: 10.1016/j.drudis.2023.103684] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/01/2023] [Accepted: 06/18/2023] [Indexed: 06/30/2023]
Abstract
Hurdles in the identification of new drugs for cancer treatment have made drug repurposing an increasingly appealing alternative. The approach involves the use of old drugs for new therapeutic purposes. It is cost-effective and facilitates rapid clinical translation. Given that cancer is also considered a metabolic disease, drugs for metabolic disorders are being actively repurposed for cancer therapeutics. In this review, we discuss the repurposing of such drugs approved for two major metabolic diseases, diabetes and cardiovascular disease (CVD), which have shown potential as anti-cancer treatment. We also highlight the current understanding of the cancer signaling pathways that these drugs target.
Collapse
Affiliation(s)
- Jisha Pillai U
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE
| | - Anindita Ray
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE
| | - Meenu Maan
- Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, UAE; New York University-Abu Dhabi, Abu Dhabi, UAE.
| | - Mainak Dutta
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE.
| |
Collapse
|
27
|
Rojas A, Schneider I, Lindner C, Gonzalez I, Morales MA. Association between diabetes and cancer. Current mechanistic insights into the association and future challenges. Mol Cell Biochem 2023; 478:1743-1758. [PMID: 36565361 DOI: 10.1007/s11010-022-04630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 11/30/2022] [Indexed: 12/25/2022]
Abstract
Compelling pieces of epidemiological, clinical, and experimental research have demonstrated that Diabetes mellitus (DM) is a major risk factor associated with increased cancer incidence and mortality in many human neoplasms. In the pathophysiology context of DM, many of the main classical actors are relevant elements that can fuel the different steps of the carcinogenesis process. Hyperglycemia, hyperinsulinemia, metabolic inflammation, and dyslipidemia are among the classic contributors to this association. Furthermore, new emerging actors have received particular attention in the last few years, and compelling data support that the microbiome, the epigenetic changes, the reticulum endoplasmic stress, and the increased glycolytic influx also play important roles in promoting the development of many cancer types. The arsenal of glucose-lowering therapeutic agents used for treating diabetes is wide and diverse, and a growing body of data raised during the last two decades has tried to clarify the contribution of therapeutic agents to this association. However, this research area remains controversial, because some anti-diabetic drugs are now considered as either promotors or protecting elements. In the present review, we intend to highlight the compelling epidemiological shreds of evidence that support this association, as well as the mechanistic contributions of many of these potential pathological mechanisms, some controversial points as well as future challenges.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| | - Ivan Schneider
- Medicine Faculty, Catholic University of Maule, Talca, Chile
| | | | - Ileana Gonzalez
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Miguel A Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
28
|
Wu W, Wang Y, Xie J, Fan S. Empagliflozin: a potential anticancer drug. Discov Oncol 2023; 14:127. [PMID: 37436535 DOI: 10.1007/s12672-023-00719-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
Empagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, is a highly effective and well-tolerated antidiabetic drug. In addition to hypoglycemic effects, empagliflozin has many other effects, such as being hypotensive and cardioprotective. It also has anti-inflammatory and antioxidative stress effects in diabetic nephropathy. Several studies have shown that empagliflozin has anticancer effects. SGLT2 is expressed in a variety of cancer cell lines. The SGLT2 inhibitor empagliflozin has significant inhibitory effects on certain types of tumor cells, such as inhibition of proliferation, migration and induction of apoptosis. In conclusion, empagliflozin has promising applications in cancer therapy as a drug for the treatment of diabetes and heart failure. This article provides a brief review of the anticancer effects of empagliflozin.
Collapse
Affiliation(s)
- Wenwen Wu
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Yanyan Wang
- Department of Ultrasonic Medicine, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China
| | - Jun Xie
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China.
| | - Shaohua Fan
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China.
| |
Collapse
|
29
|
Abdel-Qadir H, Carrasco R, Austin PC, Chen Y, Zhou L, Fang J, Su HM, Lega IC, Kaul P, Neilan TG, Thavendiranathan P. The Association of Sodium-Glucose Cotransporter 2 Inhibitors With Cardiovascular Outcomes in Anthracycline-Treated Patients With Cancer. JACC CardioOncol 2023; 5:318-328. [PMID: 37397088 PMCID: PMC10308059 DOI: 10.1016/j.jaccao.2023.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 07/04/2023] Open
Abstract
Background Sodium glucose cotransporter-2 inhibitors (SGLT2is) are hypothesized to reduce the risk of anthracycline-associated cardiotoxicity. Objectives This study sought to determine the association between SGLT2is and cardiovascular disease (CVD) after anthracycline-containing chemotherapy. Methods Using administrative data sets, we conducted a population-based cohort study of people >65 years of age with treated diabetes and no prior heart failure (HF) who received anthracyclines between January 1, 2016, and December 31, 2019. After estimating propensity scores for SGLT2i use, the average treatment effects for the treated weights were used to reduce baseline differences between SGLT2i-exposed and -unexposed controls. The outcomes were hospitalization for HF, incident HF diagnoses (in- or out-of-hospital), and documentation of any CVD in future hospitalizations. Death was treated as a competing risk. Cause-specific HRs for each outcome were determined for SGLT2i-treated people relative to unexposed controls. Results We studied 933 patients (median age 71.0 years, 62.2% female), 99 of whom were SGLT2i treated. During a median follow-up of 1.6 years, there were 31 hospitalizations for HF (0 in the SGLT2i group), 93 new HF diagnoses, and 74 hospitalizations with documented CVD. Relative to controls, SGLT2i exposure was associated with HR of 0 for HF hospitalization (P < 0.001) but no significant difference in incident HF diagnosis (HR: 0.55; 95% CI: 0.23-1.31; P = 0.18) or CVD diagnosis (HR: 0.39; 95% CI: 0.12-1.28; P = 0.12). There was no significant difference in mortality (HR: 0.63; 95% CI: 0.36-1.11; P = 0.11). Conclusions SGLT2is may reduce the rate of HF hospitalization after anthracycline-containing chemotherapy. This hypothesis warrants further testing in randomized controlled trials.
Collapse
Affiliation(s)
- Husam Abdel-Qadir
- Women’s College Hospital, Toronto, Ontario, Canada
- University Health Network, Toronto, Ontario, Canada
- ICES (formerly known as the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, Toronto, Ontario, Canada
| | - Rodrigo Carrasco
- Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, Toronto, Ontario, Canada
| | - Peter C. Austin
- ICES (formerly known as the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Yue Chen
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Limei Zhou
- ICES (formerly known as the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Jiming Fang
- ICES (formerly known as the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Henry M.H. Su
- Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, Toronto, Ontario, Canada
| | - Iliana C. Lega
- Women’s College Hospital, Toronto, Ontario, Canada
- ICES (formerly known as the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Padma Kaul
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
- Canadian Virtual Coordinating Centre for Global Collaborative Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | - Paaladinesh Thavendiranathan
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Cui W, Niu Y, Sun Z, Liu R, Chen L. Structures of human SGLT in the occluded state reveal conformational changes during sugar transport. Nat Commun 2023; 14:2920. [PMID: 37217492 DOI: 10.1038/s41467-023-38720-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
Sodium-Glucose Cotransporters (SGLT) mediate the uphill uptake of extracellular sugars and play fundamental roles in sugar metabolism. Although their structures in inward-open and outward-open conformations are emerging from structural studies, the trajectory of how SGLTs transit from the outward-facing to the inward-facing conformation remains unknown. Here, we present the cryo-EM structures of human SGLT1 and SGLT2 in the substrate-bound state. Both structures show an occluded conformation, with not only the extracellular gate but also the intracellular gate tightly sealed. The sugar substrate are caged inside a cavity surrounded by TM1, TM2, TM3, TM6, TM7, and TM10. Further structural analysis reveals the conformational changes associated with the binding and release of substrates. These structures fill a gap in our understanding of the structural mechanisms of SGLT transporters.
Collapse
Affiliation(s)
- Wenhao Cui
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Yange Niu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Zejian Sun
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
| | - Rui Liu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, 100871, Beijing, China.
- National Biomedical Imaging Center, Peking University, 100871, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China.
| |
Collapse
|
31
|
Bazzone A, Zerlotti R, Barthmes M, Fertig N. Functional characterization of SGLT1 using SSM-based electrophysiology: Kinetics of sugar binding and translocation. Front Physiol 2023; 14:1058583. [PMID: 36824475 PMCID: PMC9941201 DOI: 10.3389/fphys.2023.1058583] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Beside the ongoing efforts to determine structural information, detailed functional studies on transporters are essential to entirely understand the underlying transport mechanisms. We recently found that solid supported membrane-based electrophysiology (SSME) enables the measurement of both sugar binding and transport in the Na+/sugar cotransporter SGLT1 (Bazzone et al, 2022a). Here, we continued with a detailed kinetic characterization of SGLT1 using SSME, determining KM and KD app for different sugars, kobs values for sugar-induced conformational transitions and the effects of Na+, Li+, H+ and Cl- on sugar binding and transport. We found that the sugar-induced pre-steady-state (PSS) charge translocation varies with the bound ion (Na+, Li+, H+ or Cl-), but not with the sugar species, indicating that the conformational state upon sugar binding depends on the ion. Rate constants for the sugar-induced conformational transitions upon binding to the Na+-bound carrier range from 208 s-1 for D-glucose to 95 s-1 for 3-OMG. In the absence of Na+, rate constants are decreased, but all sugars bind to the empty carrier. From the steady-state transport current, we found a sequence for sugar specificity (Vmax/KM): D-glucose > MDG > D-galactose > 3-OMG > D-xylose. While KM differs 160-fold across tested substrates and plays a major role in substrate specificity, Vmax only varies by a factor of 1.9. Interestingly, D-glucose has the lowest Vmax across all tested substrates, indicating a rate limiting step in the sugar translocation pathway following the fast sugar-induced electrogenic conformational transition. SGLT1 specificity for D-glucose is achieved by optimizing two ratios: the sugar affinity of the empty carrier for D-glucose is similarly low as for all tested sugars (KD,K app = 210 mM). Affinity for D-glucose increases 14-fold (KD,Na app = 15 mM) in the presence of sodium as a result of cooperativity. Apparent affinity for D-glucose during transport increases 8-fold (KM = 1.9 mM) compared to KD,Na app due to optimized kinetics. In contrast, KM and KD app values for 3-OMG and D-xylose are of similar magnitude. Based on our findings we propose an 11-state kinetic model, introducing a random binding order and intermediate states corresponding to the electrogenic transitions detected via SSME upon substrate binding.
Collapse
Affiliation(s)
- Andre Bazzone
- Nanion Technologies GmbH, Munich, Germany,*Correspondence: Andre Bazzone,
| | - Rocco Zerlotti
- Nanion Technologies GmbH, Munich, Germany,Department of Structural Biology, Faculty of Biology and Pre-Clinics, Institute of Biochemistry, Genetics and Microbiology, University of Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
32
|
Ding L, Chen X, Zhang W, Dai X, Guo H, Pan X, Xu Y, Feng J, Yuan M, Gao X, Wang J, Xu X, Li S, Wu H, Cao J, He Q, Yang B. Canagliflozin primes antitumor immunity by triggering PD-L1 degradation in endocytic recycling. J Clin Invest 2023; 133:e154754. [PMID: 36594471 PMCID: PMC9797339 DOI: 10.1172/jci154754] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/26/2022] [Indexed: 01/04/2023] Open
Abstract
Understanding the regulatory mechanisms of PD-L1 expression in tumors provides key clues for improving immune checkpoint blockade efficacy or developing novel oncoimmunotherapy. Here, we showed that the FDA-approved sodium-glucose cotransporter-2 (SGLT2) inhibitor canagliflozin dramatically suppressed PD-L1 expression and enhanced T cell-mediated cytotoxicity. Mechanistic study revealed that SGLT2 colocalized with PD-L1 at the plasma membrane and recycling endosomes and thereby prevented PD-L1 from proteasome-mediated degradation. Canagliflozin disturbed the physical interaction between SGLT2 and PD-L1 and subsequently allowed the recognition of PD-L1 by Cullin3SPOP E3 ligase, which triggered the ubiquitination and proteasome-mediated degradation of PD-L1. In mouse models and humanized immune-transformation models, either canagliflozin treatment or SGLT2 silencing significantly reduced PD-L1 expression and limited tumor progression - to a level equal to the PD-1 mAb - which was correlated with an increase in the activity of antitumor cytotoxic T cells. Notably, prolonged progression-free survival and overall survival curves were observed in the group of PD-1 mAb-treated patients with non-small cell lung cancer with high expression of SGLT2. Therefore, our study identifies a regulator of cell surface PD-L1, provides a ready-to-use small-molecule drug for PD-L1 degradation, and highlights a potential therapeutic target to overcome immune evasion by tumor cells.
Collapse
Affiliation(s)
- Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Xi Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Wenxin Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Xiaoyang Dai
- Center of Drug Safety Evaluation and Research, Zhejiang University, Hangzhou, China
| | - Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Xiaohui Pan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Yanjun Xu
- Department of Medical Thoracic Oncology and
| | - Jianguo Feng
- Institute of Basic Medicine and Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China
| | - Meng Yuan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Xiaomeng Gao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Jian Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Xiaqing Xu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Sicheng Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
- The Innovation Institute for Artificial Intelligence in Medicine and
- Cancer Center of Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, and
- The Innovation Institute for Artificial Intelligence in Medicine and
| |
Collapse
|
33
|
Menchikov LG, Shestov AA, Popov AV. Warburg Effect Revisited: Embodiment of Classical Biochemistry and Organic Chemistry. Current State and Prospects. BIOCHEMISTRY (MOSCOW) 2023; 88:S1-S20. [PMID: 37069111 DOI: 10.1134/s0006297923140018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The Nobel Prize Winner (1931) Dr. Otto H. Warburg had established that the primary energy source of the cancer cell is aerobic glycolysis (the Warburg effect). He also postulated the hypothesis about "the prime cause of cancer", which is a matter of debate nowadays. Contrary to the hypothesis, his discovery was recognized entirely. However, the discovery had almost vanished in the heat of battle about the hypothesis. The prime cause of cancer is essential for the prevention and diagnosis, yet the effects that influence tumor growth are more important for cancer treatment. Due to the Warburg effect, a large amount of data has been accumulated on biochemical changes in the cell and the organism as a whole. Due to the Warburg effect, the recovery of normal biochemistry and oxygen respiration and the restoration of the work of mitochondria of cancer cells can inhibit tumor growth and lead to remission. Here, we review the current knowledge on the inhibition of abnormal glycolysis, neutralization of its consequences, and normalization of biochemical parameters, as well as recovery of oxygen respiration of a cancer cell and mitochondrial function from the point of view of classical biochemistry and organic chemistry.
Collapse
Affiliation(s)
- Leonid G Menchikov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, 119991, Russian Federation
| | - Alexander A Shestov
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, Perelman Center for Advanced Medicine, Philadelphia, PA 19104, USA
| | - Anatoliy V Popov
- University of Pennsylvania, Department of Radiology, Philadelphia, PA 19104, USA.
| |
Collapse
|
34
|
SGLT-2 Inhibitors in Cancer Treatment-Mechanisms of Action and Emerging New Perspectives. Cancers (Basel) 2022; 14:cancers14235811. [PMID: 36497303 PMCID: PMC9738342 DOI: 10.3390/cancers14235811] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
A new group of antidiabetic drugs, sodium-glucose cotransporter 2 inhibitors (SGLT-2 inhibitors), have recently been shown to have anticancer effects and their expression has been confirmed in many cancer cell lines. Given the metabolic reprogramming of these cells in a glucose-based model, the ability of SGLT-2 inhibitors to block the glucose uptake by cancer cells appears to be an attractive therapeutic approach. In addition to tumour cells, SGLT-2s are only found in the proximal tubules in the kidneys. Furthermore, as numerous clinical trials have shown, the use of SGLT-2 inhibitors is well-tolerated and safe in patients with diabetes and/or heart failure. In vitro cell culture studies and preclinical in vivo studies have confirmed that SGLT-2 inhibitors exhibit antiproliferative effects on certain types of cancer. However, the mechanisms of this action remain unclear. Even in those tumour cell types in which SGLT-2 is present, there is sometimes an SGLT-2-independent mechanism of anticancer action of this group of drugs. This article presents the current state of knowledge of the potential mechanisms of the anticancer action of SGLT-2 inhibitors and their possible future application in clinical oncology.
Collapse
|
35
|
Niu Y, Cui W, Liu R, Wang S, Ke H, Lei X, Chen L. Structural mechanism of SGLT1 inhibitors. Nat Commun 2022; 13:6440. [PMID: 36307403 PMCID: PMC9616851 DOI: 10.1038/s41467-022-33421-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/16/2022] [Indexed: 12/25/2022] Open
Abstract
Sodium glucose co-transporters (SGLT) harness the electrochemical gradient of sodium to drive the uphill transport of glucose across the plasma membrane. Human SGLT1 (hSGLT1) plays a key role in sugar uptake from food and its inhibitors show promise in the treatment of several diseases. However, the inhibition mechanism for hSGLT1 remains elusive. Here, we present the cryo-EM structure of the hSGLT1-MAP17 hetero-dimeric complex in the presence of the high-affinity inhibitor LX2761. LX2761 locks the transporter in an outward-open conformation by wedging inside the substrate-binding site and the extracellular vestibule of hSGLT1. LX2761 blocks the putative water permeation pathway of hSGLT1. The structure also uncovers the conformational changes of hSGLT1 during transitions from outward-open to inward-open states.
Collapse
Affiliation(s)
- Yange Niu
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China ,grid.11135.370000 0001 2256 9319National Biomedical Imaging Center, Peking University, Beijing, China
| | - Wenhao Cui
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China ,grid.11135.370000 0001 2256 9319National Biomedical Imaging Center, Peking University, Beijing, China ,grid.27255.370000 0004 1761 1174Taishan College, Shandong University, Qingdao, China
| | - Rui Liu
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China ,grid.11135.370000 0001 2256 9319National Biomedical Imaging Center, Peking University, Beijing, China
| | - Sanshan Wang
- grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China ,grid.454727.7Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing, China
| | - Han Ke
- grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China ,grid.454727.7Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing, China
| | - Xiaoguang Lei
- grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China ,grid.454727.7Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing, China
| | - Lei Chen
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China ,grid.11135.370000 0001 2256 9319National Biomedical Imaging Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
36
|
Nakhal MM, Aburuz S, Sadek B, Akour A. Repurposing SGLT2 Inhibitors for Neurological Disorders: A Focus on the Autism Spectrum Disorder. Molecules 2022; 27:7174. [PMID: 36364000 PMCID: PMC9653623 DOI: 10.3390/molecules27217174] [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: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 09/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a substantially increasing incidence rate. It is characterized by repetitive behavior, learning difficulties, deficits in social communication, and interactions. Numerous medications, dietary supplements, and behavioral treatments have been recommended for the management of this condition, however, there is no cure yet. Recent studies have examined the therapeutic potential of the sodium-glucose cotransporter 2 (SGLT2) inhibitors in neurodevelopmental diseases, based on their proved anti-inflammatory effects, such as downregulating the expression of several proteins, including the transforming growth factor beta (TGF-β), interleukin-6 (IL-6), C-reactive protein (CRP), nuclear factor κB (NF-κB), tumor necrosis factor alpha (TNF-α), and the monocyte chemoattractant protein (MCP-1). Furthermore, numerous previous studies revealed the potential of the SGLT2 inhibitors to provide antioxidant effects, due to their ability to reduce the generation of free radicals and upregulating the antioxidant systems, such as glutathione (GSH) and superoxide dismutase (SOD), while crossing the blood brain barrier (BBB). These properties have led to significant improvements in the neurologic outcomes of multiple experimental disease models, including cerebral oxidative stress in diabetes mellitus and ischemic stroke, Alzheimer's disease (AD), Parkinson's disease (PD), and epilepsy. Such diseases have mutual biomarkers with ASD, which potentially could be a link to fill the gap of the literature studying the potential of repurposing the SGLT2 inhibitors' use in ameliorating the symptoms of ASD. This review will look at the impact of the SGLT2 inhibitors on neurodevelopmental disorders on the various models, including humans, rats, and mice, with a focus on the SGLT2 inhibitor canagliflozin. Furthermore, this review will discuss how SGLT2 inhibitors regulate the ASD biomarkers, based on the clinical evidence supporting their functions as antioxidant and anti-inflammatory agents capable of crossing the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Mohammed Moutaz Nakhal
- Department of Biochemistry, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
| | - Salahdein Aburuz
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 17666, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
37
|
Li Y, Liu B, Yin X, Jiang Z, Fang C, Chen N, Zhang B, Dai L, Yin Y. Targeted demethylation of the SLC5A7 promotor inhibits colorectal cancer progression. Clin Epigenetics 2022; 14:92. [PMID: 35858918 PMCID: PMC9301853 DOI: 10.1186/s13148-022-01308-5] [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: 11/15/2021] [Accepted: 07/04/2022] [Indexed: 02/08/2023] Open
Abstract
Background SLC5A7 (solute carrier family 5 member 7), also known as choline transporter 1 (CHT1), is downregulated in colorectal cancer (CRC) and functions as a tumor suppressor. However, the mechanisms underlying the inactivation of SLC5A7 in CRC remain to be elucidated. Results In the present study, two broad-spectrum demethylation agents (azacitidine and decitabine) employed to treat CRC cells significantly upregulated SLC5A7 expression. Further results based on the CRC cohort and TCGA database indicated that SLC5A7 promoter methylation inversely correlated with SLC5A7 expression, and the status of SLC5A7 promotor methylation showed a promising prognostic value for patients with CRC. Next, the dCas9-multiGCN4/scFv-TET1CD-based precision demethylation system was constructed, which could significantly and specifically promote SLC5A7 expression in CRC cells through sgRNA targeting the SLC5A7 promoter. Both in vitro and in vivo experiments demonstrated that targeted demethylation of SLC5A7 by dCas9-multiGCN4/scFv-TET1CD-sgSLC5A7 inhibited tumor growth by stabilizing p53 and regulating downstream targets. Conclusions Collectively, DNA promoter methylation caused inactivation of SLC5A7 in CRC, and targeted demethylation of SLC5A7 might be a therapeutic target for CRC and other cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01308-5.
Collapse
Affiliation(s)
- Yang Li
- Department of Gastrointestinal Surgery, Guang'an People's Hospital, Guang'an, 638500, Sichuan, People's Republic of China.,Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Baike Liu
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhiyuan Jiang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.,School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
38
|
Wu W, Zhang Z, Jing D, Huang X, Ren D, Shao Z, Zhang Z. SGLT2 inhibitor activates the STING/IRF3/IFN-β pathway and induces immune infiltration in osteosarcoma. Cell Death Dis 2022; 13:523. [PMID: 35662245 PMCID: PMC9166744 DOI: 10.1038/s41419-022-04980-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023]
Abstract
SGLT2 (sodium-glucose cotransporter 2) is an important mediator of epithelial glucose transport and has been reported that SGLT2, robustly and diffusely expressed in malignant cancer cells, was overexpressed in various tumors, and inhibiting the SGLT2 expression significantly inhibited tumor progression. By blocking the functional activity of SGLT2, SGLT2 inhibitors have shown anticancer effects in several malignant cancers, including breast cancer, cervical cancer, hepatocellular cancer, prostate cancer, and lung cancer. However, the anticancer effect of SGLT2 inhibitors in osteosarcoma and the specific mechanism are still unclear. In the present study, we found that SGLT2 was overexpressed at the protein level in osteosarcoma. Furthermore, our results showed that the SGLT2 inhibitor significantly inhibited osteosarcoma tumor growth and induced infiltration of immune cells in vivo by upregulating STING expression and activating the IRF3/IFN-β pathway, which could attribute to the suppression of AKT phosphorylation. In addition, the combined treatment with SGLT2 inhibitor and STING agonist 2'3'-cGAMP exerted synergistic antitumor effects in osteosarcoma. Furthermore, the overexpression of SGLT2 at the protein level was correlated with the degradation of SGLT2 induced by TRIM21. This result demonstrated that SGLT2 is a novel therapeutic target of osteosarcoma, and that the SGLT2 inhibitor, especially in combination with 2'3'-cGAMP, is a potential therapeutic drug.
Collapse
Affiliation(s)
- Wei Wu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Zhenhao Zhang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Doudou Jing
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xin Huang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Dianyun Ren
- grid.33199.310000 0004 0368 7223Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Zengwu Shao
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Zhicai Zhang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
39
|
Shojaei Baghini S, Gardanova ZR, Abadi SAH, Zaman BA, İlhan A, Shomali N, Adili A, Moghaddar R, Yaseri AF. CRISPR/Cas9 application in cancer therapy: a pioneering genome editing tool. Cell Mol Biol Lett 2022; 27:35. [PMID: 35508982 PMCID: PMC9066929 DOI: 10.1186/s11658-022-00336-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
The progress of genetic engineering in the 1970s brought about a paradigm shift in genome editing technology. The clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9 (CRISPR/Cas9) system is a flexible means to target and modify particular DNA sequences in the genome. Several applications of CRISPR/Cas9 are presently being studied in cancer biology and oncology to provide vigorous site-specific gene editing to enhance its biological and clinical uses. CRISPR's flexibility and ease of use have enabled the prompt achievement of almost any preferred alteration with greater efficiency and lower cost than preceding modalities. Also, CRISPR/Cas9 technology has recently been applied to improve the safety and efficacy of chimeric antigen receptor (CAR)-T cell therapies and defeat tumor cell resistance to conventional treatments such as chemotherapy and radiotherapy. The current review summarizes the application of CRISPR/Cas9 in cancer therapy. We also discuss the present obstacles and contemplate future possibilities in this context.
Collapse
Affiliation(s)
- Sadegh Shojaei Baghini
- Plant Biotechnology Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zhanna R. Gardanova
- Department of Psychotherapy, Pirogov Russian National Research Medical University, 1 Ostrovityanova St., 117997 Moscow, Russia
| | - Saeme Azizi Hassan Abadi
- Department of Nursery and Midwifery, Faculty of Laboratory Science, Islamic Azad University of Chalous, Mazandaran, Iran
| | - Burhan Abdullah Zaman
- Basic Sciences Department, College of Pharmacy, University of Duhok, Kurdistan Region, Iraq
| | - Ahmet İlhan
- Department of Medical Biochemistry, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Navid Shomali
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Adili
- Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran
- Senior Adult Oncology Department, Moffitt Cancer Center, University of South Florida, Tampa, USA
| | - Roozbeh Moghaddar
- Department of Pediatric Hematology and Oncology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
40
|
Ginsenoside Rd protects cerebral endothelial cells from oxygen-glucose deprivation/reoxygenation induced pyroptosis via inhibiting SLC5A1 mediated sodium influx. J Ginseng Res 2022; 46:700-709. [PMID: 36090684 PMCID: PMC9459060 DOI: 10.1016/j.jgr.2022.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/21/2022] [Accepted: 05/12/2022] [Indexed: 12/03/2022] Open
Abstract
Background Ginsenoside Rd is a natural compound with promising neuroprotective effects. However, the underlying mechanisms are still not well-understood. In this study, we explored whether ginsenoside Rd exerts protective effects on cerebral endothelial cells after oxygen-glucose deprivation/reoxygenation (OGD/R) treatment and its potential docking proteins related to the underlying regulations. Method Commercially available primary human brain microvessel endothelial cells (HBMECs) were used for in vitro OGD/R studies. Cell viability, pyroptosis-associated protein expression and tight junction protein degradation were evaluated. Molecular docking proteins were predicted. Subsequent surface plasmon resonance (SPR) technology was utilized for validation. Flow cytometry was performed to quantify caspase-1 positive and PI positive (caspase-1+/PI+) pyroptotic cells. Results Ginsenoside Rd treatment attenuated OGD/R-induced damage of blood-brain barrier (BBB) integrity in vitro. It suppressed NLRP3 inflammasome activation (increased expression of NLRP3, cleaved caspase-1, IL-1β and GSDMD-N terminal (NT)) and subsequent cellular pyroptosis (caspase-1+/PI + cells). Ginsenoside Rd interacted with SLC5A1 with a high affinity and reduced OGD/R-induced sodium influx and potassium efflux in HBMECs. Inhibiting SLC5A1 using phlorizin suppressed OGD/R-activated NLRP3 inflammasome and pyroptosis in HBMECs. Conclusion Ginsenoside Rd protects HBMECs from OGD/R-induced injury partially via binding to SLC5A1, reducing OGD/R-induced sodium influx and potassium efflux, thereby alleviating NLRP3 inflammasome activation and pyroptosis.
Collapse
|
41
|
Papaioannou L, Avgoustakis K. Responsive nanomedicines enhanced by or enhancing physical modalities to treat solid cancer tumors: Preclinical and clinical evidence of safety and efficacy. Adv Drug Deliv Rev 2022; 181:114075. [PMID: 34883140 DOI: 10.1016/j.addr.2021.114075] [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: 07/01/2021] [Revised: 09/22/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022]
Abstract
Nanomedicine has improved cancer treatment but not to the extent anticipated. Responsive nanomedicines enhanced by physical modalities (radiation, ultrasounds, alternating magnetic fields) or enhancing the activity of physical modalities such as radiotherapy to kill cancer represents an important approach in improving the safety and anticancer effectiveness. Importantly, the combined treatments have shown promise for the treatment of difficult to treat tumors, such as tumors that are resistant to chemotherapy (multi drug resistant, MDR) or radiotherapy and hypoxic tumors, and for the prevention of tumor metastasis. In this review, the mechanisms of responsive nanomedicines activity enhancement by physical means and vice versa are presented and preclinical and, most importantly, clinical evidence of the safety and efficacy of nanomedicines enhanced by or enhancing by physical modalities in treating solid tumors are critically discussed.
Collapse
Affiliation(s)
- Ligeri Papaioannou
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Konstantinos Avgoustakis
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, 26504 Patras, Greece; Biomedical Research Foundation Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, Athens 11527, Greece.
| |
Collapse
|
42
|
Du J, Gu J, Deng J, Kong L, Guo Y, Jin C, Bao Y, Fu D, Li J. The expression and survival significance of sodium glucose transporters in pancreatic cancer. BMC Cancer 2022; 22:116. [PMID: 35090421 PMCID: PMC8796473 DOI: 10.1186/s12885-021-09060-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/25/2021] [Indexed: 12/31/2022] Open
Abstract
Background Sodium glucose transporters (SGLTs) play vital roles in glucose uptake in many solid cancers, including pancreatic cancer (PC). However, their expression profile in pancreatic cancer and correlation with prognosis are not clear. Thus, we aimed to analyse the expression profile and prognostic significance of SGLT-1 and SGLT-2 in PC. Methods Eighty-eight patients with pancreatic ductal adenocarcinoma (PDAC) undergoing surgery in Huashan Hospital, Fudan University, from July 2017 to June 2020 were enrolled in the study. Specimens for immunohistochemistry were obtained through surgical resection. Bioinformatics analysis was performed based on the Gene Expression Omnibus (GEO), Oncomine and The Cancer Genome Atlas (TCGA) databases. The statistics were calculated using IBM SPSS Statistics, version 20 and R 4.1.1. P values lower than 0.05 were considered to indicate statistical significance. Results SGLT-1 but not SGLT-2 was significantly overexpressed in PDAC. Survival analysis showed that the median overall survival (OS) and progression-free survival (PFS) of patients with high SGLT-1 expression were significantly longer than that of patients with low SGLT-1 expression. Cox regression indicated that high SGLT-1 expression was an independent predictor for a better prognosis, while residual tumour status (R1 and R2) was an independent risk factor for a poor prognosis. Finally, PDZK1-interacting protein 1 (PDZK1IP1), a protein participating in the generation of reactive oxygen species, was overexpressed in PDAC and its expression was significantly correlated with SGLT-1. Conclusions SGLT-1 but not SGLT-2 was overexpressed in PDAC, and the overexpression of SGLT-1 could be a predictor of a better prognosis. Residual tumour status (R1 and R2) was a risk factor for poor prognosis and disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09060-4.
Collapse
|
43
|
Structural basis of the selective sugar transport in sodium-glucose cotransporters. J Mol Biol 2022; 434:167464. [DOI: 10.1016/j.jmb.2022.167464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/23/2022]
|
44
|
Wang X, Zhang Y, Li Y, Tang M, Deng Q, Mao J, Du L. Estrogen Regulates Glucose Metabolism in Cattle Neutrophils Through Autophagy. Front Vet Sci 2021; 8:773514. [PMID: 34912878 PMCID: PMC8666889 DOI: 10.3389/fvets.2021.773514] [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: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Hypoglycemia resulting from a negative energy balance (NEB) in periparturient cattle is the major reason for a reduced glycogen content in polymorphonuclear neutrophils (PMNs). The lack of glycogen induces PMNs dysfunction and is responsible for the high incidence of perinatal diseases. The perinatal period is accompanied by dramatic changes in sex hormones levels of which estrogen (17β-estradiol, E2) has been shown to be closely associated with PMNs function. However, the precise regulatory mechanism of E2 on glucose metabolism in cattle PMNs has not been elucidated. Cattle PMNs were cultured in RPMI 1640 with 2.5 (LG), 5.5 (NG) and 25 (HG) mM glucose and E2 at 20 (EL), 200 (EM) and 450 (EH) pg/mL. We found that E2 maintained PMNs viability in different glucose conditions, and promoted glycogen synthesis by inhibiting PFK1, G6PDH and GSK-3β activity in LG while enhancing PFK1 and G6PDH activity and inhibiting GSK-3β activity in HG. E2 increased the ATP content in LG but decreased it in HG. This indicated that the E2-induced increase/decrease of ATP content may be independent of glycolysis and the pentose phosphate pathway (PPP). Further analysis showed that E2 promoted the activity of hexokinase (HK) and GLUT1, GLUT4 and SGLT1 expression in LG, while inhibiting GLUT1, GLUT4 and SGLT1 expression in HG. Finally, we found that E2 increased LC3, ATG5 and Beclin1 expression, inhibited p62 expression, promoting AMPK-dependent autophagy in LG, but with the opposite effect in HG. Moreover, E2 increased the Bcl-2/Bax ratio and decreased the apoptosis rate of PMNs in LG but had the opposite effect in HG. These results showed that E2 could promote AMPK-dependent autophagy and inhibit apoptosis in response to glucose-deficient environments. This study elucidated the detailed mechanism by which E2 promotes glycogen storage through enhancing glucose uptake and retarding glycolysis and the PPP in LG. Autophagy is essential for providing ATP to maintain the survival and immune potential of PMNs. These results provided significant evidence for further understanding the effects of E2 on PMNs immune potential during the hypoglycemia accompanying perinatal NEB in cattle.
Collapse
Affiliation(s)
- Xinbo Wang
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Yuming Zhang
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Yansong Li
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Mingyu Tang
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Qinghua Deng
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Jingdong Mao
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Liyin Du
- Clinical Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| |
Collapse
|
45
|
Lau KTK, Ng L, Wong JWH, Loong HHF, Chan WWL, Lee CH, Wong CKH. Repurposing sodium-glucose co-transporter 2 inhibitors (SGLT2i) for cancer treatment - A Review. Rev Endocr Metab Disord 2021; 22:1121-1136. [PMID: 34272645 DOI: 10.1007/s11154-021-09675-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2021] [Indexed: 01/24/2023]
Abstract
Developed as an antidiabetic drug, recent evidence suggests that several sodium-glucose co-transporter 2 inhibitors (SGLT2i), especially canagliflozin and dapagliflozin, may exhibit in vitro and in vivo anticancer activities in selected cancer types, including an inhibition of tumor growth and induction of cell death. When used in combination with chemotherapy or radiotherapy, SGLT2i may offer possible synergistic effects in enhancing their treatment efficacy while alleviating associated side effects. Potential mechanisms include a reduction of glucose uptake into cancer cells, systemic glucose restriction, modulation of multiple signaling pathways, and regulation of different gene and protein expression. Furthermore, preliminary clinical findings have reported potential anticancer properties of canagliflozin and dapagliflozin in patients with liver and colon cancers respectively, with reference to decreases in their tumor marker levels. Given its general tolerability and routine use in diabetes management, SGLT2i may be a good candidate for drug repurposing in cancer treatment and as adjunct to conventional therapies. While current evidence reveals that only certain SGLT2i appear to be effective against selected cancer types, further studies are needed to explore the antitumor abilities of each SGLT2i in various cancers. Moreover, clinical trials are called for to evaluate the safety and feasibility of introducing SGLT2i in the treatment regimen of patients with specific cancers, and to identify the preferred route of drug administration for targeted delivery to selected tumor sites.
Collapse
Affiliation(s)
- Kristy T K Lau
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lui Ng
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jason W H Wong
- School of Biomedical Sciences, Faculty of Medicine, Li Ka Shing, The University of Hong Kong, Hong Kong SAR, China
| | - Herbert H F Loong
- Department of Clinical Oncology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wendy W L Chan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chi Ho Lee
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Carlos K H Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
46
|
Ho H, Kikuchi K, Oikawa D, Watanabe S, Kanemitsu Y, Saigusa D, Kujirai R, Ikeda‐Ohtsubo W, Ichijo M, Akiyama Y, Aoki Y, Mishima E, Ogata Y, Oikawa Y, Matsuhashi T, Toyohara T, Suzuki C, Suzuki T, Mano N, Kagawa Y, Owada Y, Katayama T, Nakayama T, Tomioka Y, Abe T. SGLT-1-specific inhibition ameliorates renal failure and alters the gut microbial community in mice with adenine-induced renal failure. Physiol Rep 2021; 9:e15092. [PMID: 34921520 PMCID: PMC8683788 DOI: 10.14814/phy2.15092] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/20/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022] Open
Abstract
Sodium-dependent glucose cotransporters (SGLTs) have attracted considerable attention as new targets for type 2 diabetes mellitus. In the kidney, SGLT2 is the major glucose uptake transporter in the proximal tubules, and inhibition of SGLT2 in the proximal tubules shows renoprotective effects. On the other hand, SGLT1 plays a role in glucose absorption from the gastrointestinal tract, and the relationship between SGLT1 inhibition in the gut and renal function remains unclear. Here, we examined the effect of SGL5213, a novel and potent intestinal SGLT1 inhibitor, in a renal failure (RF) model. SGL5213 improved renal function and reduced gut-derived uremic toxins (phenyl sulfate and trimethylamine-N-oxide) in an adenine-induced RF model. Histological analysis revealed that SGL5213 ameliorated renal fibrosis and inflammation. SGL5213 also reduced gut inflammation and fibrosis in the ileum, which is a primary target of SGL5213. Examination of the gut microbiota community revealed that the Firmicutes/Bacteroidetes ratio, which suggests gut dysbiosis, was increased in RF and SGL5213 rebalanced the ratio by increasing Bacteroidetes and reducing Firmicutes. At the genus level, Allobaculum (a major component of Erysipelotrichaceae) was significantly increased in the RF group, and this increase was canceled by SGL5213. We also measured the effect of SGL5213 on bacterial phenol-producing enzymes that catalyze tyrosine into phenol, following the reduction of phenyl sulfate, which is a novel marker and a therapeutic target for diabetic kidney disease DKD. We found that the enzyme inhibition was less potent, suggesting that the change in the microbial community and the reduction of uremic toxins may be related to the renoprotective effect of SGL5213. Because SGL5213 is a low-absorbable SGLT1 inhibitor, these data suggest that the gastrointestinal inhibition of SGLT1 is also a target for chronic kidney diseases.
Collapse
Affiliation(s)
- Hsin‐Jung Ho
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Koichi Kikuchi
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Medical MegabankTohoku UniversitySendaiJapan
| | - Daiki Oikawa
- Department of Biomolecular Engineering Applied Life ChemistryTohoku University Graduate School of EngineeringSendaiJapan
| | - Shun Watanabe
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | | | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Ryota Kujirai
- Laboratory of OncologyPharmacy Practice and SciencesTohoku University Graduate School of Pharmaceutical SciencesSendaiJapan
| | - Wakako Ikeda‐Ohtsubo
- Laboratory of Animal Products ChemistryGraduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Mariko Ichijo
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yukako Akiyama
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yuichi Aoki
- Department of Integrative Genomics, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Eikan Mishima
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshiaki Ogata
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshitsugu Oikawa
- Department of PediatricsTohoku University Graduate School of MedicineSendaiJapan
| | - Tetsuro Matsuhashi
- Department of PediatricsTohoku University Graduate School of MedicineSendaiJapan
| | - Takafumi Toyohara
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Chitose Suzuki
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Takehiro Suzuki
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Nariyasu Mano
- Department of Pharmaceutical SciencesTohoku University HospitalSendaiJapan
| | - Yoshiteru Kagawa
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Yuji Owada
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Takane Katayama
- Laboratory of Molecular Biology of BioresponseGraduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Toru Nakayama
- Department of Biomolecular Engineering Applied Life ChemistryTohoku University Graduate School of EngineeringSendaiJapan
| | - Yoshihisa Tomioka
- Laboratory of Molecular Biology of BioresponseGraduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Takaaki Abe
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
47
|
Vrhovac Madunić I, Karin-Kujundžić V, Madunić J, Šola IM, Šerman L. Endometrial Glucose Transporters in Health and Disease. Front Cell Dev Biol 2021; 9:703671. [PMID: 34552924 PMCID: PMC8450505 DOI: 10.3389/fcell.2021.703671] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022] Open
Abstract
Pregnancy loss is a frequent occurrence during the peri-implantation period, when there is high glucose demand for embryonic development and endometrial decidualization. Glucose is among the most essential uterine fluid components required for those processes. Numerous studies associate abnormal glucose metabolism in the endometrium with a higher risk of adverse pregnancy outcomes. The endometrium is incapable of synthesizing glucose, which thus must be delivered into the uterine lumen by glucose transporters (GLUTs) and/or the sodium-dependent glucose transporter 1 (SGLT1). Among the 26 glucose transporters (14 GLUTs and 12 SGLTs) described, 10 (9 GLUTs and SGLT1) are expressed in rodents and 8 (7 GLUTs and SGLT1) in the human uterus. This review summarizes present knowledge on the most studied glucose transporters in the uterine endometrium (GLUT1, GLUT3, GLUT4, and GLUT8), whose data regarding function and regulation are still lacking. We present the recently discovered SGLT1 in the mouse and human endometrium, responsible for controlling glycogen accumulation essential for embryo implantation. Moreover, we describe the epigenetic regulation of endometrial GLUTs, as well as signaling pathways included in uterine GLUT’s expression. Further investigation of the GLUTs function in different endometrial cells is of high importance, as numerous glucose transporters are associated with infertility, polycystic ovary syndrome, and gestational diabetes.
Collapse
Affiliation(s)
- Ivana Vrhovac Madunić
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Valentina Karin-Kujundžić
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence in Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Josip Madunić
- Biochemistry and Organic Analytical Chemistry Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Ida Marija Šola
- Department of Gynecology and Obstetrics, Sisters of Charity University Hospital, Zagreb, Croatia
| | - Ljiljana Šerman
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence in Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
48
|
Yin Y, Jiang Z, Fu J, Li Y, Fang C, Yin X, Chen Y, Chen N, Li J, Ji Y, Su X, Qiu M, Huang W, Zhang B, Deng H, Dai L. Choline-induced SLC5A7 impairs colorectal cancer growth by stabilizing p53 protein. Cancer Lett 2021; 525:55-66. [PMID: 34562520 DOI: 10.1016/j.canlet.2021.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023]
Abstract
The members of the solute carrier (SLC) superfamily are vital membrane transporters in human cells. In the present study, we determine the expression and function of SLC5 family members in colorectal cancer (CRC). Expression analysis based on The Cancer Genome Atlas database and potential clinical relation analysis based on the Oncomine database indicate that SLC5A7 is downregulated and is predicted to correlate with the staging, and prognosis response of CRC. Additional results demonstrate that SLC5A7 is downregulated and correlates with good prognosis in patients with CRC. Ectopic expression of SLC5A7 either by overexpression, or uptake of choline efficiently inhibits CRC growth. Examination of the molecular mechanism reveals that SLC5A7 promotes p53 protein expression by directly interacting with and modifying p53 and disrupting the interaction between p53 and MDM2 in wild type p53 CRC cells. Our findings establish the clear correlation between SLC5A7 and tumour growth, providing a novel potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Zhiyuan Jiang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Jiamei Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yang Li
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China; Department of Gastrointestinal Surgery, Guang'an People's Hospital, Guang'an, Sichuan, 638500, People's Republic of China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ye Chen
- Department of Medical Oncology, Cancer Center, The State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Junshu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanhong Ji
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Meng Qiu
- Department of Medical Oncology, Cancer Center, The State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
49
|
Expression of SGLT1 in the Mouse Endometrial Epithelium and its Role in Early Embryonic Development and Implantation. Reprod Sci 2021; 28:3094-3108. [PMID: 34460091 DOI: 10.1007/s43032-021-00480-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/31/2021] [Indexed: 10/20/2022]
Abstract
Many functional activities of endometrium epithelium are energy consuming which are very important for maintaining intrauterine environment needed by early embryonic development and establishment of implantation window. Glucose is a main energy supplier and one of the main components of intrauterine fluid. Obviously, glucose transports in endometrium epithelium involve in for these activities but their functions have not been elucidated. In this research, we observed a spatiotemporal pattern of sodium glucose transporter 1 (SGLT1) expression in the mouse endometrium. We also determined that progesterone can promote the expression of SGLT1 in the mouse endometrial epithelium in response to the action of oestrogen. Treatment with the SGLT1 inhibitor phlorizin or small interfering RNA specific for SGLT1 (SGLT1-siRNA) altered glucose uptake in primary cultured endometrial epithelial cells, which exhibited reduced ATP levels and AMPK activation. The injection of phlorizin or SGLT1-siRNA into one uterine horn of each mouse on day 2 of pregnancy led to an increased glucose concentration in the uterine fluid and decreased number of harvested normal blastocysts and decreased expression of integrin αVβ3 in endometrial epithelium and increased expression of mucin 1 and lactoferrin in endometrial epithelium and the uterine homogenates exhibited activated AMPK, a decreased ATP level on day 4, and a decreased number of implantation sites on day 5. In embryo transfer experiments, pre-treatment of the uterine horn with phlorizin or SGLT1-siRNA during the implantation window led to a decreased embryo implantation rate on day 5 of pregnancy, even when embryos from normal donor mice were used. In conclusion, SGLT1, which participates in glucose transport in the mouse endometrial epithelium, inhibition and/or reduced expression of SGLT1 affects early embryo development by altering the glucose concentration in the uterine fluid. Inhibition and/or reduced expression of SGLT1 also affects embryo implantation by influencing energy metabolism in epithelial cells, which consequently influences implantation-related functional activities.
Collapse
|
50
|
Ren D, Sun Y, Zhang D, Li D, Liu Z, Jin X, Wu H. SGLT2 promotes pancreatic cancer progression by activating the Hippo signaling pathway via the hnRNPK-YAP1 axis. Cancer Lett 2021; 519:277-288. [PMID: 34314754 DOI: 10.1016/j.canlet.2021.07.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/03/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022]
Abstract
SGLT2 is overexpressed in various cancers, including pancreatic cancer. However, the mechanisms underlying the tumorigenic effects of SGLT2 in pancreatic cancer remain unclear. In this study, we demonstrated that SGLT2 inhibition significantly suppressed the growth of pancreatic cancer cells in vitro and in vivo. RNA sequencing, real-time PCR, and Western blot analyses revealed that SGLT2 silencing or inhibition suppressed Hippo signaling activation by downregulating YAP1 expression. Liquid chromatography-mass spectrometry and immunoprecipitation analyses showed that SGLT2 interacted with hnRNPK, promoting its nuclear translocation and thereby enhancing hnRNPK-induced YAP1 transcription. Importantly, YAP1 inhibitor enhanced the anti-pancreatic cancer effect of SGLT2 inhibitor in mice bearing pancreatic tumors. These findings suggest that SGLT2 promotes pancreatic cancer progression by activating the Hippo signaling pathway through the hnRNPK-YAP1 axis. Hence, SGLT2 inhibition alone or combined with YAP1 inhibition may represent a promising therapeutic approach for pancreatic cancer.
Collapse
Affiliation(s)
- Dianyun Ren
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yan Sun
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Dan Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Dan Li
- Cardiovascular Medicine Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhiqiang Liu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xin Jin
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha,Hunan, 410011, China.
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|