1
|
Wei Y, Li Z, Yu T, Chen Y, Yang Q, Wen K, Liao J, Li L. Ultrasound-activated piezoelectric biomaterials for cartilage regeneration. ULTRASONICS SONOCHEMISTRY 2025; 117:107353. [PMID: 40250302 DOI: 10.1016/j.ultsonch.2025.107353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Due to the low density of chondrocytes and limited ability to repair damaged extracellular matrix (ECM) in cartilage, many patients with congenital or acquired craniofacial trauma require filler graft materials to support facial structure, restore function, improve self-confidence, and regain socialization. Ultrasound has the capacity to stimulate piezoelectric materials, converting mechanical energy into electrical signals that can regulate the metabolism, proliferation, and differentiation of chondrocytes. This unique property has sparked growing interest in using piezoelectric biomaterials in regenerative medicine. In this review, we first explain the principle behind ultrasound-activated piezoelectric materials and how they generate piezopotential. We then review studies demonstrating how this bioelectricity promotes chondrocyte regeneration, stimulates the secretion of key extracellular components and supports cartilage regeneration by activating relevant signaling pathways. Next, we discuss the properties, synthesis, and modification strategies of various piezoelectric biomaterials. We further discuss recent progresses in the development of ultrasound-activated piezoelectric biomaterials specifically designed for cartilage regeneration. Lastly, we discuss future research challenges facing this technology, ultrasound-activated piezoelectric materials for cartilage regeneration engineering. While the technology holds great promise, certain obstacles remain, including issues related to material stability, precise control over ultrasound parameters, and the integration of these systems into clinical settings. The combination of ultrasound-activated piezoelectric technology with other emerging fields, such as Artificial Intelligence (AI) and cartilage organoid chips, may open new frontiers in regenerative medicine. We hope this review encourages further exploration of ultrasound-activated strategies for piezoelectric materials and their future applications in regenerative medicines.
Collapse
Affiliation(s)
- Yangchen Wei
- The First Affiliated Hospital, Center of Burn & Plastic and Wound Repair, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China
| | - Zhengyang Li
- The First Affiliated Hospital, Center of Burn & Plastic and Wound Repair, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China
| | - Tianjing Yu
- The First Affiliated Hospital, Center of Burn & Plastic and Wound Repair, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yan Chen
- The First Affiliated Hospital, Center of Burn & Plastic and Wound Repair, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Qinglai Yang
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kaikai Wen
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Junlin Liao
- The First Affiliated Hospital, Center of Burn & Plastic and Wound Repair, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
2
|
Artusa P, White JH. Vitamin D and its analogs in immune system regulation. Pharmacol Rev 2025; 77:100032. [PMID: 40148037 DOI: 10.1016/j.pharmr.2024.100032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Vitamin D was discovered as the cure for nutritional rickets, a disease of bone growth arising from inadequate intestinal calcium absorption, and for much of the 20th century, it was studied for its critical role in calcium homeostasis. However, we now recognize that the vitamin D receptor and vitamin D metabolic enzymes are expressed in numerous tissues unrelated to calcium homeostasis. Notably, vitamin D signaling can induce cellular differentiation and cell cycle arrest. Moreover, the vitamin D receptor and the enzyme CYP27B1, which produces the hormonal form of vitamin D, 1,25-dihydroxyvitamin D (1,25D), are expressed throughout the immune system. In addition, CYP27B1 expression in immune cells is regulated by physiological inputs independent of those controlling its expression in calcium homeostatic tissues. These observations have driven the development of 1,25D-like secosteroidal analogs and nonsecosteroidal analogs to separate the effects of vitamin D on cell differentiation and function from its calcemic activities. Notably, some of these analogs have had considerable success in the clinic in the treatment of inflammatory and immune-related disorders. In this review, we described in detail the mechanisms of vitamin D signaling and the physiological signals controlling 1,25D synthesis and catabolism, with a focus on the immune system. We also surveyed the effects of 1,25D and its analogs on the regulation of immune system function and their implications for human immune-related disorders. Finally, we described the potential of vitamin D analogs as anticancer therapeutics, in particular, their use as adjuncts to cancer immunotherapy. SIGNIFICANCE STATEMENT: Vitamin D signaling is active in both the innate and adaptive arms of the immune system. Numerous vitamin D analogs, developed primarily to minimize the dose-limiting hypercalcemia of the active form of vitamin D, have been used widely in preclinical and clinical studies of immune system regulation. This review presents a description of the mechanisms of action of vitamin D signaling, an overview of analog development, and an in-depth discussion of the immunoregulatory roles of vitamin D analogs.
Collapse
Affiliation(s)
- Patricio Artusa
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - John H White
- Department of Physiology, McGill University, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Hahn JM, Combs KA, Phillips CM, Warner PM, Qazi UA, Powell HM, Supp DM. CYP24A1 is overexpressed in keloid keratinocytes and its inhibition alters profibrotic gene expression. BURNS & TRAUMA 2025; 13:tkae063. [PMID: 39822648 PMCID: PMC11736898 DOI: 10.1093/burnst/tkae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 01/19/2025]
Abstract
Background Keloids are disfiguring, fibrotic scar-like lesions that are challenging to treat and commonly recur after therapy. A deeper understanding of the mechanisms driving keloid formation is necessary for the development of more effective therapies. Reduced vitamin D receptor (VDR) expression has been observed in keloids, implicating vitamin D signaling in keloid pathology. Vitamin D exhibits anti-proliferative and anti-inflammatory properties, suggesting it could have therapeutic utility in keloid disorder. The current study investigated vitamin D-regulated gene expression in keloid keratinocytes and the effects of inhibiting an enzyme involved in vitamin D metabolism on the phenotype of keloid-derived keratinocytes. Methods Normal and keloid-derived primary keratinocytes were isolated from normal skin and keloid lesions, respectively, and were cultured in the absence or presence of vitamin D. In some experiments, inhibitors of the vitamin D metabolizing enzyme CYP24A1, ketoconazole or VID400 were added in the absence or presence of vitamin D. Cellular proliferation, migration and gene expression were measured. Results We observed significant overexpression of CYP24A1 mRNA in keloid versus normal keratinocytes and increased CYP24A1 protein levels in keloids versus normal skin. CYP24A1 encodes 24 hydroxylase and is induced by vitamin D in a feedback loop that regulates vitamin D levels; thus, inhibition of CYP24A1 activity may locally increase active vitamin D levels. Ketoconazole, a non-specific cytochrome P-450 inhibitor, reduced proliferation of keloid and normal keratinocytes, but VID400, a specific CYP24A1 inhibitor, only significantly affected keloid keratinocyte proliferation. Neither inhibitor significantly reduced keratinocyte migration. The two inhibitors had different effects on vitamin D target gene expression in keratinocytes. Specifically, ketoconazole treatment reduced CYP24A1 expression in normal and keloid keratinocytes, whereas VID400 increased CYP24A1 expression. Both inhibitors decreased expression of profibrotic genes, including periostin and hyaluronan synthase 2, in keloid-derived cells. Combined treatment of keloid keratinocytes with vitamin D and ketoconazole or VID400 increased the effects of vitamin D treatment on target genes, although the effects were gene- and cell type-specific. Conclusions The data suggest that reduction of vitamin D inactivation with CYP24A1 inhibitors may reduce profibrotic gene expression in keloid-derived cells. Therefore, CYP24A1 inhibitors may serve as adjunctive therapies to suppress keloid-associated gene expression changes.
Collapse
Affiliation(s)
- Jennifer M Hahn
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Kelly A Combs
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Caitlin M Phillips
- Clinical Research Department, Shriners Children’s Ohio, One Children's Plaza - 2 West, Dayton, OH, 45404, USA
| | - Petra M Warner
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Uzair A Qazi
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Heather M Powell
- Clinical Research Department, Shriners Children’s Ohio, One Children's Plaza - 2 West, Dayton, OH, 45404, USA
- Department of Materials Science and Engineering, The Ohio State University, 140 W. 19th Avenue, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 W. 19th Avenue, Columbus, OH, 43210, USA
| | - Dorothy M Supp
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
- Clinical Research Department, Shriners Children’s Ohio, One Children's Plaza - 2 West, Dayton, OH, 45404, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| |
Collapse
|
4
|
Chao G, Lin A, Bao Y. A study of the association of vitamin D receptor (VDR) as a predictive biomarker for immune checkpoint inhibitor therapy with immune invasion in colon adenocarcinoma. J Pharm Biomed Anal 2025; 252:116510. [PMID: 39378759 DOI: 10.1016/j.jpba.2024.116510] [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: 06/01/2024] [Revised: 09/21/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024]
Abstract
Colon adenocarcinoma(COAD) is a primary and aggressive malignancy with the fifth highest mortality rate among cancers, and it is important to discover new strategies. The online database was used to analyze the correlation between Vitamin D receptor (VDR) and COAD, and further explore the immune infiltration and related gene networks.The expression and methylation levels of VDR was analyzed by using Timer database, GEPIA platform and UALCAN database. GeneMANIA database was used to analyze and obtain gene networks that are closely linked to VDR. UALCAN database was used to score the gene effects of VDR in colorectal cancer cell lines. The cBioPortal database was used for the detection of gene mutations. The survival curve analysis was carried out using the GEPIA database. The relationship between VDR expression and immune cell infiltration was analyzed by using the timer database and TISIDB database. TISIDB database was used to obtain VDR-related drug targets.The expression of VDR was significantly lower in COAD(p<0.05). The methylation level of VDR was significantly higher in COAD (p<0.05). The gene mutation rate of VDR in COAD was 2 %. OS and DFS were not associated with changes in the VDR gene in patients with COAD. VDR expression was correlated with CD4+T cell infiltration, macrophage infiltration, neutrophil infiltration, and dendritic cell infiltration. VDR has a clear correlation with ADORA2A, BTLA, CD160, CD244, CD274, CD96, CSF1R, CTLA4, HAVCR2, IL10, IDO1, LAG3, LGALS9, PDCD1, PDCD1LG2, PVRL2, TGFB1, TGFBR1, TIGIT and VTCN1.The expression of VDR is associated with immune infiltration in patients with COAD. VDR may be a new candidate biomarker for determining the level of immune infiltration and predicting immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, China.
| | - Ailing Lin
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, China
| | - Yang Bao
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, China
| |
Collapse
|
5
|
De Smedt J, Van Kelst S, Janssen L, Marasigan V, Boecxstaens V, Bogaerts K, Belmans A, Vanderschueren D, Vandenberghe K, Bechter O, Aura C, Lambrechts D, Strobbe T, Emri G, Nikkels A, Garmyn M. High-dose vitamin D supplementation does not improve outcome in a cutaneous melanoma population: results of a randomized double-blind placebo-controlled study (ViDMe trial). Br J Dermatol 2024; 191:886-896. [PMID: 38913652 DOI: 10.1093/bjd/ljae257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Observational studies in cutaneous melanoma (CM) have indicated an inverse relationship between levels of 25-hydroxyvitamin D and Breslow thickness, in addition to a protective effect of high 25-hydroxyvitamin D levels on clinical outcome. OBJECTIVES To evaluate whether high-dose vitamin D supplementation in curatively resected CM reduces melanoma relapse. METHODS In a prospective randomized double-blind placebo-controlled trial, 436 patients with resected CM stage IA to III (8th American Joint Committee on Cancer staging) were randomized. Among them, 218 received a placebo while 218 received monthly 100 000 IU cholecalciferol for a minimum of 6 months and a maximum of 42 months (treatment arm). Following randomization, patients were followed for a median of 52 months, with a maximum follow-up of 116 months. The primary endpoint was relapse-free survival. Secondary endpoints were melanoma-related mortality, overall survival, and the evolution of 25-hydroxyvitamin D serum levels over time. RESULTS In our population (mean age 55 years, 54% female sex) vitamin D supplementation increased 25-hydroxyvitamin D serum levels after 6 months of supplementation in the treatment arm by a median 17 ng mL-1 [95% confidence interval (CI) 9-26] compared with 0 ng mL-1 (95% CI 6-8) in the placebo arm (P < 0.001, Wilcoxon test) and remained at a steady state during the whole treatment period. The estimated event rate for relapse-free survival at 72 months after inclusion was 26.51% in the vitamin D supplemented arm (95% CI 19.37-35.64) vs. 20.70% (95% CI 14.26-29.52) in the placebo arm (hazard ratio 1.27, 95% CI 0.79-2.03; P = 0.32). After adjusting for confounding factors (including baseline stage, body mass index, age, sex and baseline season), the hazard ratio was 1.20 (95% CI 0.74-1.94, P = 0.46). The number of deaths from progression of CM and nonmelanoma-related deaths was similar in both the vitamin D supplemented and placebo groups (deaths from progression of CM, n = 10 and n = 11, respectively; nonmelanoma-related deaths, n = 3 and n = 2, respectively). No major adverse events were observed during the study. CONCLUSIONS In patients with CM, monthly high-dose vitamin D supplementation was safe, resulted in a sustained increase in 25-hydroxyvitamin D levels during the treatment period, but did not improve relapse-free survival, melanoma-related death or overall survival.
Collapse
Affiliation(s)
- Julie De Smedt
- Laboratory of Dermatology, Department of Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| | - Sofie Van Kelst
- Laboratory of Dermatology, Department of Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| | - Laudine Janssen
- Laboratory of Dermatology, Department of Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| | - Vivien Marasigan
- Department of Surgery, South Infirmary Victoria University Hospital, Cork, Ireland
| | - Veerle Boecxstaens
- Oncological Surgery, Department of Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| | - Kris Bogaerts
- Leuven Biostatistics and Statistical Bioinformatics Centre (L-BioStat), KU Leuven, Leuven, Belgium
| | - Ann Belmans
- Leuven Biostatistics and Statistical Bioinformatics Centre (L-BioStat), KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronical Illness and Metabolism, KU Leuven, UZ Leuven, Leuven, Belgium
| | | | - Oliver Bechter
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| | - Claudia Aura
- Conway Institute of Biomolecular and Biomedical Research, Pathology, University College Dublin, Ireland
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Tinne Strobbe
- Department of Dermatology, Imeldaziekenhuis, Bonheiden, Belgium
| | - Gabriella Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Arjen Nikkels
- CHU Sart Tilman, University of Liège, Department of Dermatology, Liège, Belgium
| | - Marjan Garmyn
- Laboratory of Dermatology, Department of Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| |
Collapse
|
6
|
van Doorn R. Supplemental vitamin D on trial: no evidence of benefit as an adjuvant treatment for melanoma. Br J Dermatol 2024; 191:858-859. [PMID: 39041830 DOI: 10.1093/bjd/ljae302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/23/2024] [Indexed: 07/24/2024]
Affiliation(s)
- Remco van Doorn
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Dermatology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Conway K, Edmiston SN, Vondras A, Reiner A, Corcoran DL, Shen R, Parrish EA, Hao H, Lin L, Kenney JM, Ilelaboye G, Kostrzewa CE, Kuan PF, Busam KJ, Lezcano C, Lee TK, Hernando E, Googe PB, Ollila DW, Moschos S, Gorlov I, Amos CI, Ernstoff MS, Cust AE, Wilmott JS, Scolyer RA, Mann GJ, Vergara IA, Ko J, Rees JR, Yan S, Nagore E, Bosenberg M, Rothberg BG, Osman I, Lee JE, Saenger Y, Bogner P, Thompson CL, Gerstenblith M, Holmen SL, Funchain P, Brunsgaard E, Depcik-Smith ND, Luo L, Boyce T, Orlow I, Begg CB, Berwick M, Thomas NE. DNA Methylation Classes of Stage II and III Primary Melanomas and Their Clinical and Prognostic Significance. JCO Precis Oncol 2024; 8:e2400375. [PMID: 39509669 PMCID: PMC11737429 DOI: 10.1200/po-24-00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/05/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024] Open
Abstract
PURPOSE Patients with stage II and III cutaneous primary melanoma vary considerably in their risk of melanoma-related death. We explore the ability of methylation profiling to distinguish primary melanoma methylation classes and their associations with clinicopathologic characteristics and survival. MATERIALS AND METHODS InterMEL is a retrospective case-control study that assembled primary cutaneous melanomas from American Joint Committee on Cancer (AJCC) 8th edition stage II and III patients diagnosed between 1998 and 2015 in the United States and Australia. Cases are patients who died of melanoma within 5 years from original diagnosis. Controls survived longer than 5 years without evidence of melanoma recurrence or relapse. Methylation classes, distinguished by consensus clustering of 850K methylation data, were evaluated for their clinicopathologic characteristics, 5-year survival status, and differentially methylated gene sets. RESULTS Among 422 InterMEL melanomas, consensus clustering revealed three primary melanoma methylation classes (MethylClasses): a CpG island methylator phenotype (CIMP) class, an intermediate methylation (IM) class, and a low methylation (LM) class. CIMP and IM were associated with higher AJCC stage (both P = .002), Breslow thickness (CIMP P = .002; IM P = .006), and mitotic index (both P < .001) compared with LM, while IM had higher N stage than CIMP (P = .01) and LM (P = .007). CIMP and IM had a 2-fold higher likelihood of 5-year death from melanoma than LM (CIMP odds ratio [OR], 2.16 [95% CI, 1.18 to 3.96]; IM OR, 2.00 [95% CI, 1.12 to 3.58]) in a multivariable model adjusted for age, sex, log Breslow thickness, ulceration, mitotic index, and N stage. Despite more extensive CpG island hypermethylation in CIMP, CIMP and IM shared similar patterns of differential methylation and gene set enrichment compared with LM. CONCLUSION Melanoma MethylClasses may provide clinical value in predicting 5-year death from melanoma among patients with primary melanoma independent of other clinicopathologic factors.
Collapse
Affiliation(s)
- Kathleen Conway
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC
- Department of Dermatology, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sharon N. Edmiston
- Department of Dermatology, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Amanda Vondras
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Allison Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David L. Corcoran
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ronglai Shen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eloise A. Parrish
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Honglin Hao
- Department of Dermatology, University of North Carolina, Chapel Hill, NC
| | - Lan Lin
- Department of Dermatology, University of North Carolina, Chapel Hill, NC
| | - Jessica M Kenney
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gbemisola Ilelaboye
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Caroline E. Kostrzewa
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pei Fen Kuan
- Department of Applied Mathematics and Statistics, State University of New York, Stony Brook, NY
| | - Klaus J. Busam
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Cecilia Lezcano
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tim K. Lee
- British Columbia Cancer Research Center, Vancouver, BC, Canada
| | - Eva Hernando
- Grossman School of Medicine, New York University, New York, NY
| | - Paul B. Googe
- Department of Dermatology, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David W. Ollila
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Stergios Moschos
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ivan Gorlov
- Department of Medicine, Baylor Medical Center, Houston, TX
| | | | | | - Anne E. Cust
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, Australia
- Melanoma Institute of Australia, The University of Sydney, New South Wales, Australia
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - James S. Wilmott
- Melanoma Institute of Australia, The University of Sydney, New South Wales, Australia
| | - Richard A. Scolyer
- Melanoma Institute of Australia, The University of Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Graham J. Mann
- Melanoma Institute of Australia, The University of Sydney, New South Wales, Australia
- John Curtin School of Medical Research, Australian National University, ACT 2601, Australia
| | - Ismael A. Vergara
- Melanoma Institute of Australia, The University of Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Judy R. Rees
- Department of Epidemiology, Dartmouth Medical School, Lebanon NH
| | - Shaofeng Yan
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon NH
| | - Eduardo Nagore
- Instituto Valenciano de Oncologia, Valencia, Spain
- Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | | | | | - Iman Osman
- Grossman School of Medicine, New York University, New York, NY
| | - Jeffrey E. Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yvonne Saenger
- Columbia University Medical School, New York, NY
- Albert Einstein School of Medicine, New York, NY
| | - Paul Bogner
- Departments of Pathology and Dermatology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Cheryl L. Thompson
- Case Western Reserve University, Cleveland, OH
- Penn State University, Hershey, PA
| | | | - Sheri L. Holmen
- Department of Surgery, University of Utah Health Sciences Center and Huntsman Cancer Institute, Salt Lake City, UT
| | | | - Elise Brunsgaard
- Department of Dermatology, Rush University Medical Center, Chicago, Il
| | | | - Li Luo
- Department of Internal Medicine and the UNM Comprehensive Cancer Center, Albuquerque, NM
| | - Tawny Boyce
- Department of Internal Medicine and the UNM Comprehensive Cancer Center, Albuquerque, NM
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Colin B. Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marianne Berwick
- Department of Internal Medicine and the UNM Comprehensive Cancer Center, Albuquerque, NM
| | - Nancy E. Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | |
Collapse
|
8
|
Randerson-Moor J, Davies J, Harland M, Nsengimana J, Bigirumurame T, Walker C, Laye J, Appleton ES, Ball G, Cook GP, Bishop DT, Salmond RJ, Newton-Bishop J. Systemic Inflammation, the Peripheral Blood Transcriptome, and Primary Melanoma. J Invest Dermatol 2024; 144:2513-2529.e17. [PMID: 38583742 DOI: 10.1016/j.jid.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 04/09/2024]
Abstract
Peripheral blood transcriptomes from 383 patients with newly diagnosed melanoma were subjected to differential gene expression analysis. The hypotheses were that impaired systemic immunity is associated with poorer prognosis (thicker tumors and fewer tumor-infiltrating lymphocytes) and evidence of systemic inflammation (high-sensitivity CRP and fibrinogen levels). Higher fibrinogen levels were associated with thicker primary tumors. In single-gene analysis, high-sensitivity CRP levels were significantly associated with higher blood CD274 expression (coding for PD-L1), but each was independently prognostic, with high-sensitivity CRP associated with increased mortality and higher CD274 protective, independent of age. Pathway analysis identified downregulation of immune cell signaling pathways in the blood of people with thicker tumors and notable upregulation of signal transducer and activator of transcription 1 gene STAT1 in people with brisk tumor-infiltrating lymphocytes. Transcriptomic data provided evidence for increased NF-kB signaling with higher inflammatory markers but with reduction in expression of HLA class II molecules and higher CD274, suggesting that aberrant systemic inflammation is a significant mediator of reduced immune function in melanoma. In summary, transcriptomic data revealed evidence of reduced immune function in patients with thicker tumors and fewer tumor-infiltrating lymphocytes at diagnosis. Inflammatory markers were associated with thicker primaries and independently with death from melanoma, suggesting that systemic inflammation contributes to that reduced immune function.
Collapse
Affiliation(s)
- Juliette Randerson-Moor
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - John Davies
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom; Leeds Institute of Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Mark Harland
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jérémie Nsengimana
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Theophile Bigirumurame
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Christopher Walker
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jon Laye
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Elizabeth S Appleton
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom; Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Graham P Cook
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - D Timothy Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Robert J Salmond
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Julia Newton-Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
9
|
Li J, Hou Y, Ding H, Wang P, Li B. 1α,25-hydroxyvitamin D/VDR suppresses stem-like properties of ovarian cancer cells by restraining nuclear translocation of β-catenin. Steroids 2024; 211:109488. [PMID: 39151767 DOI: 10.1016/j.steroids.2024.109488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
Several studies have indicated that 1α,25-hydroxyvitamin D [1α,25(OH)2D3] inhibits the proliferation and metastasis of cancer cells through suppressing epithelial-mesenchymal transition. However, its influence on the translocation of β-catenin remains unclear. In the present study, ovarian cancer stem-like cells (CSCs), including side population (SP) and CD44+/CD117+, were isolated from mouse ovarian surface epithelial (MOSE) cells with malignant transformation. The findings revealed that 1α,25(OH)2D3 obviously reduced the sphere-forming ability, as well as Notch1 and Klf levels. Moreover, the limiting dilution assay demonstrated that 1α,25(OH)2D3 effectively hindered the tumorigenesis of ovarian CSCs in vitro. Notably, treatment with 1α,25(OH)2D3 led to a substantial increase in the cell population of CD44+/CD117+ forming one tumor from ≤ 100 to 445 in orthotopic transplanted model, indicating a pronounced suppression of stemness of ovarian CSCs. Additionally, 1α,25(OH)2D3 robustly promoted the translocation of β-catenin from the nuclear to the cytoplasm through directly binding to VDR, which resulted in decreased levels of c-Myc and CyclinD1 within late MOSE cells. Taken together, these results strongly supported the role of 1α,25(OH)2D3 in inhibiting stem-like properties in ovarian cancer cells by restraining nuclear translocation of β-catenin, thereby offering a promising target for cancer therapeutics.
Collapse
Affiliation(s)
- Jie Li
- Deparment of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yongfeng Hou
- Deparment of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Beijing 100037, China
| | - Hongmei Ding
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou 215123, China.
| | - Ping Wang
- Deparment of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| | - Bingyan Li
- Deparment of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
10
|
Gieniusz E, Skrzydlewska E, Łuczaj W. Current Insights into the Role of UV Radiation-Induced Oxidative Stress in Melanoma Pathogenesis. Int J Mol Sci 2024; 25:11651. [PMID: 39519202 PMCID: PMC11546485 DOI: 10.3390/ijms252111651] [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: 10/04/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Cutaneous melanoma accounts for the majority of skin cancer-related deaths, and its incidence increases each year. The growing number of melanoma cases, especially in advanced stages, poses a significant socio-medical challenge throughout the world. Extensive research on melanoma pathogenesis identifies UV radiation as the most important factor in melanocytic transformation. Oxidative effects of UV irradiation exert their influence on melanoma pathogenesis primarily through modification of nucleic acids, proteins, and lipids, further disrupting cellular signaling and cell cycle regulation. Its effects extend beyond melanocytes, leading to immunosuppression in the exposed skin tissue, which consequently creates conditions for immune surveillance evasion and further progression. In this review, we focus on the specific molecular changes observed in the UV-dependent oxidative stress environment and their biological consequences in the course of the disease, which have not been considered in previous reviews on melanoma. Nonetheless, data show that the exact role of oxidative stress in melanoma initiation and progression remains unclear, as it affects cancerous cells differently depending on the specific context. A better understanding of the pathophysiological basis of melanoma development holds promise for identifying potential targets, which could lead to effective melanoma prevention strategies.
Collapse
Affiliation(s)
| | | | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (E.G.); (E.S.)
| |
Collapse
|
11
|
Domżalski P, Piotrowska A, Tuckey RC, Zmijewski MA. Anticancer Activity of Vitamin D, Lumisterol and Selected Derivatives against Human Malignant Melanoma Cell Lines. Int J Mol Sci 2024; 25:10914. [PMID: 39456696 PMCID: PMC11506961 DOI: 10.3390/ijms252010914] [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: 09/11/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Despite the recent development of improved methods of treating melanoma such as targeted therapy, immunotherapy or combined treatment, the number of new cases worldwide is increasing. It is well known that active metabolites of vitamin D3 and lumisterol (L3) exert photoprotective and antiproliferative effects on the skin, while UV radiation is a major environmental risk factor for melanoma. Thus, many natural metabolites and synthetic analogs of steroidal and secosteroidal molecules have been tested on various cancer cells and in animal models. In this study, we tested the anti-melanoma properties of several natural derivatives of vitamin D3 and L3 in comparison to 1,25-dihydroxyvitamin D3 (1,25(OH)2D3). A significant decrease in melanoma cell proliferation and cell mobility was observed for selected derivatives, with (25R)-27-hydroxyL3 showing the highest potency (lowest IC50) in A375 cells but lower potency in SK-MEL-28 cells, whereas the parent L3 failed to inhibit proliferation. The efficacy (% inhibition) by 1,24,25(OH)3D3 and 1,25(OH)2D3 were similar in both cell types. 1,25(OH)2D3 showed higher potency than 1,24,25(OH)3D3 in SK-MEL-28 cells, but lower potency in A375 cells for the inhibition of proliferation. As for 1,25(OH)2D3, but not the other derivatives tested, treatment of melanoma cells with 1,24,25(OH)3D3 markedly increased the expression of CYP24A1, enhanced translocation of the vitamin D receptor (VDR) from the cytoplasm to the nucleus and also decreased the expression of the proliferation marker Ki67. The effects of the other compounds tested were weaker and occurred only under certain conditions. Our data indicate that 1,24,25(OH)3D3, which has undergone the first step in 1,25(OH)2D3 inactivation by being hydroxylated at C24, still shows anti-melanoma properties, displaying higher potency than 1,25(OH)2D3 in SK-MEL-28 cells. Furthermore, hydroxylation increases the potency of some of the lumisterol hydroxy-derivatives, as in contrast to L3, (25R)-27(OH)L3 effectively inhibits proliferation and migration of the human malignant melanoma cell line A375.
Collapse
Affiliation(s)
- Paweł Domżalski
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland; (P.D.); (A.P.)
| | - Anna Piotrowska
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland; (P.D.); (A.P.)
| | - Robert C. Tuckey
- School of Molecular Sciences, University of Western Australia, Perth, WA 6009, Australia;
| | - Michał A. Zmijewski
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland; (P.D.); (A.P.)
| |
Collapse
|
12
|
Slominski AT, Kim TK, Janjetovic Z, Slominski RM, Li W, Jetten AM, Indra AK, Mason RS, Tuckey RC. Biological Effects of CYP11A1-Derived Vitamin D and Lumisterol Metabolites in the Skin. J Invest Dermatol 2024; 144:2145-2161. [PMID: 39001720 PMCID: PMC11416330 DOI: 10.1016/j.jid.2024.04.022] [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: 02/01/2024] [Revised: 04/06/2024] [Accepted: 04/29/2024] [Indexed: 07/15/2024]
Abstract
Novel pathways of vitamin D3, lumisterol 3 (L3), and tachysterol 3 (T3) activation have been discovered, initiated by CYP11A1 and/or CYP27A1 in the case of L3 and T3. The resulting hydroxymetabolites enhance protection of skin against DNA damage and oxidative stress; stimulate keratinocyte differentiation; exert anti-inflammatory, antifibrogenic, and anticancer activities; and inhibit cell proliferation in a structure-dependent manner. They act on nuclear receptors, including vitamin D receptor, aryl hydrocarbon receptor, LXRα/β, RAR-related orphan receptor α/γ, and peroxisome proliferator-activated receptor-γ, with selectivity defined by their core structure and distribution of hydroxyl groups. They can activate NRF2 and p53 and inhibit NF-κB, IL-17, Shh, and Wnt/β-catenin signaling. Thus, they protect skin integrity and physiology.
Collapse
Affiliation(s)
- Andrzej T Slominski
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA; Cancer Chemoprevention Program, Comprehensive Cancer Center, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA; Veterans Administration Medical Center, Birmingham, Alabama, USA.
| | - Tae-Kang Kim
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zorica Janjetovic
- Department of Dermatology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Radomir M Slominski
- Department of Genetics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei Li
- Drug Discovery Center, Department of Pharmaceutical Sciences, University of Tennessee Health Science Center College of Pharmacy, Memphis, Tennessee, USA
| | - Anton M Jetten
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA; Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon; USA
| | - Rebecca S Mason
- School of Life and Environmental Sciences, The University of Sydney, Australia; Charles Perkins Centre, The University of Sydney, Australia
| | - Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| |
Collapse
|
13
|
Mikulić P, Ogorevc M, Petričević M, Kaličanin D, Tafra R, Saraga-Babić M, Mardešić S. SOX2, JAGGED1, β-Catenin, and Vitamin D Receptor Expression Patterns during Early Development and Innervation of the Human Inner Ear. Int J Mol Sci 2024; 25:8719. [PMID: 39201406 PMCID: PMC11354891 DOI: 10.3390/ijms25168719] [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: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Sensorineural hearing loss can be caused by lesions to the inner ear during development. Understanding the events and signaling pathways that drive inner ear formation is crucial for determining the possible causes of congenital hearing loss. We have analyzed the innervation and expression of SOX2, JAGGED1, β-catenin (CTNNB1), and vitamin D receptor (VDR) in the inner ears of human conceptuses aged 5 to 10 weeks after fertilization (W) using immunohistochemistry. The prosensory domains of the human inner ear displayed SOX2 and JAGGED1 expression throughout the analyzed period, with SOX2 expression being more extensive in all the analyzed timepoints. Innervation of vestibular prosensory domains was present at 6 W and extensive at 10 W, while nerve fibers reached the base of the cochlear prosensory domain at 7-8 W. CTNNB1 and VDR expression was mostly membranous and present during all analyzed timepoints in the inner ear, being the strongest in the non-sensory epithelium. Their expression was stronger in the vestibular region compared to the cochlear duct. CTNNB1 and VDR expression displayed opposite expression trends during the analyzed period, but additional studies are needed to elucidate whether they interact during inner ear development.
Collapse
Affiliation(s)
- Petra Mikulić
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia; (P.M.); (R.T.)
| | - Marin Ogorevc
- Division of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.O.); (S.M.)
| | - Marin Petričević
- Institute of Emergency Medicine of Split-Dalmatia County, Spinčićeva 1, 21000 Split, Croatia;
| | - Dean Kaličanin
- Department of Medical Biology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia;
| | - Robert Tafra
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia; (P.M.); (R.T.)
| | - Mirna Saraga-Babić
- Division of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.O.); (S.M.)
| | - Snježana Mardešić
- Division of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.O.); (S.M.)
| |
Collapse
|
14
|
Mansilla-Polo M, Luque-Luna M, Morgado-Carrasco D. [Translated article] Vitamin D and Skin Cancer: A Controversial Society. Literature Update and Review. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:T679-T692. [PMID: 38823772 DOI: 10.1016/j.ad.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 06/03/2024] Open
Abstract
Vitamin D (VD) deficiency has been associated with various tumors. However, the association between VD and skin cancer is controversial. Although in non-melanoma skin cancer, adequate or even high levels of VD can be associated with a higher risk of developing tumors, this could be biased by the direct association between sun exposure and VD levels. Regarding melanoma, results are contradictory. Most studies analyzed state that higher levels of VD could reduce the risk of melanoma, be associated with melanomas with better prognosis and with an enhanced antitumor response, and also with fewer adverse events associated with melanoma immunotherapy. However, prospective studies of adequate methodological quality are still needed to assess the association between VD levels and its supplementation and development/prognosis in skin cancer.
Collapse
Affiliation(s)
- M Mansilla-Polo
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Instituto de Investigación Sanitaria (IIS) La Fe, Valencia, Spain
| | - M Luque-Luna
- Servicio de Dermatología, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - D Morgado-Carrasco
- Servicio de Dermatología, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Servicio de Dermatología, Hospital de Figueres, Fundació Salut Empordà, Figueres, Girona, Spain.
| |
Collapse
|
15
|
Mansilla-Polo M, Luque-Luna M, Morgado-Carrasco D. Vitamin D and Skin Cancer: A Controversial Society. Literature Update and Review. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:679-692. [PMID: 38556198 DOI: 10.1016/j.ad.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 04/02/2024] Open
Abstract
Vitamin D (VD) deficiency has been associated with various tumors. However, the association between VD and skin cancer is controversial. Although in non-melanoma skin cancer, adequate or even high levels of VD can be associated with a higher risk of developing tumors, this could be biased by the direct association between sun exposure and VD levels. Regarding melanoma, results are contradictory. Most studies analyzed state that higher levels of VD could reduce the risk of melanoma, be associated with melanomas with better prognosis and with an enhanced antitumor response, and also with fewer adverse events associated with melanoma immunotherapy. However, prospective studies of adequate methodological quality are still needed to assess the association between VD levels and its supplementation and development/prognosis in skin cancer.
Collapse
Affiliation(s)
- M Mansilla-Polo
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Instituto de Investigación Sanitaria (IIS) La Fe, Valencia, España
| | - M Luque-Luna
- Servicio de Dermatología, Hospital Clínic, Universitat de Barcelona, Barcelona, España
| | - D Morgado-Carrasco
- Servicio de Dermatología, Hospital Clínic, Universitat de Barcelona, Barcelona, España; Servicio de Dermatología, Hospital de Figueres, Fundació Salut Empordà, Figueres, Girona, España.
| |
Collapse
|
16
|
Slominski RM, Kim TK, Janjetovic Z, Brożyna AA, Podgorska E, Dixon KM, Mason RS, Tuckey RC, Sharma R, Crossman DK, Elmets C, Raman C, Jetten AM, Indra AK, Slominski AT. Malignant Melanoma: An Overview, New Perspectives, and Vitamin D Signaling. Cancers (Basel) 2024; 16:2262. [PMID: 38927967 PMCID: PMC11201527 DOI: 10.3390/cancers16122262] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Melanoma, originating through malignant transformation of melanin-producing melanocytes, is a formidable malignancy, characterized by local invasiveness, recurrence, early metastasis, resistance to therapy, and a high mortality rate. This review discusses etiologic and risk factors for melanoma, diagnostic and prognostic tools, including recent advances in molecular biology, omics, and bioinformatics, and provides an overview of its therapy. Since the incidence of melanoma is rising and mortality remains unacceptably high, we discuss its inherent properties, including melanogenesis, that make this disease resilient to treatment and propose to use AI to solve the above complex and multidimensional problems. We provide an overview on vitamin D and its anticancerogenic properties, and report recent advances in this field that can provide solutions for the prevention and/or therapy of melanoma. Experimental papers and clinicopathological studies on the role of vitamin D status and signaling pathways initiated by its active metabolites in melanoma prognosis and therapy are reviewed. We conclude that vitamin D signaling, defined by specific nuclear receptors and selective activation by specific vitamin D hydroxyderivatives, can provide a benefit for new or existing therapeutic approaches. We propose to target vitamin D signaling with the use of computational biology and AI tools to provide a solution to the melanoma problem.
Collapse
Affiliation(s)
- Radomir M. Slominski
- Department of Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Tae-Kang Kim
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Zorica Janjetovic
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Ewa Podgorska
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Katie M. Dixon
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Rebecca S. Mason
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Robert C. Tuckey
- School of Molecular Sciences, University of Western Australia, Perth, WA 6009, Australia;
| | - Rahul Sharma
- Department of Biomedical Informatics and Data Science, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - David K. Crossman
- Department of Genetics and Bioinformatics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Craig Elmets
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Chander Raman
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anton M. Jetten
- Cell Biology Section, NIEHS—National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrzej T. Slominski
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, Veteran Administration Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
17
|
Zeng JY, Wang Y, Hong FY, Miao M, Jiang YY, Qiao ZX, Wang YT, Bao XR. Tanshinone IIA is superior to paricalcitol in ameliorating tubulointerstitial fibrosis through regulation of VDR/Wnt/β-catenin pathway in rats with diabetic nephropathy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3959-3977. [PMID: 37991543 PMCID: PMC11111530 DOI: 10.1007/s00210-023-02853-3] [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: 08/16/2023] [Accepted: 11/11/2023] [Indexed: 11/23/2023]
Abstract
Glomerulosclerosis and tubulointerstitial fibrosis (TIF) are closely involved in the development of diabetic nephropathy (DN). Moreover, the development of TIF is closely related to epithelial-to-mesenchymal transition (EMT). Tanshinone IIA (Tan) has various pharmacological effects, especially the anti-fibrotic effect. And it is mainly used in the clinical treatment of cardiovascular diseases. Currently, the protective effect of Tan on DN and its possible mechanism have not been clearly elucidated. Our previous studies illustrated that Tan could improve the EMT of HK-2 cells induced by high glucose by regulating the vitamin D receptor (VDR)/Wnt/β-catenin pathway. Here, we collected demographic information and laboratory results from the National Health and Nutrition Examination Survey (NHANES) database in order to investigate the relationship between VD and DN. Then, we established a DN model and treated DN rats with Tan and paricalcitol (Par) for 6 weeks. We subsequently compared the changes in general condition, renal function, pathological changes, and TIF-related protein expression levels of control rats, DN rats induced by STZ, DN rats with Tan at 5.4 mg/kg, DN rats with Tan at 10.8 mg/kg, and DN rats with Par at 0.054 µg/kg, to explore the effect and mechanism of Tan and Par on DN rats. The results showed that VD had a protective effect against DN in diabetic patients. And we found that Tan had a protective effect on renal fibrosis in DN rats, which was superior to Par in improving the symptoms of "three more and one less," reducing fasting blood glucose level, improving renal index, BUN/SCr, and UACR, reducing histopathological damage of kidney, and improving the expression of fibrosis-related proteins in kidney tissue by regulating VDR/Wnt/β-catenin pathway. Tan was superior to Par in ameliorating tubulointerstitial fibrosis by regulating VDR/Wnt/β-catenin pathway in rats with diabetic nephropathy.
Collapse
Affiliation(s)
- Jing-Yi Zeng
- Department of Nephrology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yu Wang
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Fu-Yuan Hong
- Department of Nephrology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Miao Miao
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yu-Ying Jiang
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Zi-Xuan Qiao
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yun-Tao Wang
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xiao-Rong Bao
- Department of Nephrology, Jinshan Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Wang R, Chen Y, Xie Y, Ma X, Liu Y. Deciphering and overcoming Anti-PD-1 resistance in Melanoma: A comprehensive review of Mechanisms, biomarker Developments, and therapeutic strategies. Int Immunopharmacol 2024; 132:111989. [PMID: 38583243 DOI: 10.1016/j.intimp.2024.111989] [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: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
Worldwide, tens of thousands of people die from melanoma each year, making it the most frequently fatal form of cutaneous cancer. Immunotherapeutic advancements, particularly with anti-PD-1 medications, have significantly enhanced treatment outcomes over recent decades. With the broad application of anti-PD-1 therapies, insights into the mechanisms of resistance have evolved. Despite the development of combination treatments and early predictive biomarkers, a comprehensive synthesis of these advancements is absent in the current literature. This review underscores the prevailing knowledge of anti-PD-1 resistance mechanisms and underscores the critical role of robust predictive biomarkers in stratifying patients for targeted combinations of anti-PD-1 and other conventional or innovative therapeutic approaches. Additionally, we offer insights that may shape future melanoma treatment strategies.
Collapse
Affiliation(s)
- Ruoqi Wang
- Shanghai Skin Disease Hospital, Shanghai Clinical College of Dermatology, Fifth Clinical Medical College, Anhui Medical University, Shanghai 200443, China
| | - Yanbin Chen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yongyi Xie
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xin Ma
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Yeqiang Liu
- Shanghai Skin Disease Hospital, Shanghai Clinical College of Dermatology, Fifth Clinical Medical College, Anhui Medical University, Shanghai 200443, China; Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
19
|
Giampazolias E, da Costa MP, Lam KC, Lim KHJ, Cardoso A, Piot C, Chakravarty P, Blasche S, Patel S, Biram A, Castro-Dopico T, Buck MD, Rodrigues RR, Poulsen GJ, Palma-Duran SA, Rogers NC, Koufaki MA, Minutti CM, Wang P, Vdovin A, Frederico B, Childs E, Lee S, Simpson B, Iseppon A, Omenetti S, Kelly G, Goldstone R, Nye E, Suárez-Bonnet A, Priestnall SL, MacRae JI, Zelenay S, Patil KR, Litchfield K, Lee JC, Jess T, Goldszmid RS, Sousa CRE. Vitamin D regulates microbiome-dependent cancer immunity. Science 2024; 384:428-437. [PMID: 38662827 PMCID: PMC7615937 DOI: 10.1126/science.adh7954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 03/04/2024] [Indexed: 05/03/2024]
Abstract
A role for vitamin D in immune modulation and in cancer has been suggested. In this work, we report that mice with increased availability of vitamin D display greater immune-dependent resistance to transplantable cancers and augmented responses to checkpoint blockade immunotherapies. Similarly, in humans, vitamin D-induced genes correlate with improved responses to immune checkpoint inhibitor treatment as well as with immunity to cancer and increased overall survival. In mice, resistance is attributable to the activity of vitamin D on intestinal epithelial cells, which alters microbiome composition in favor of Bacteroides fragilis, which positively regulates cancer immunity. Our findings indicate a previously unappreciated connection between vitamin D, microbial commensal communities, and immune responses to cancer. Collectively, they highlight vitamin D levels as a potential determinant of cancer immunity and immunotherapy success.
Collapse
Affiliation(s)
- Evangelos Giampazolias
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | | | - Khiem C. Lam
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology (LICI), Center for Cancer Research (CCR), National Cancer Institute (NCI), 37 Convent Drive, Bethesda, MD 20892-0001, USA
| | - Kok Haw Jonathan Lim
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College, London, UK
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Cécile Piot
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sonja Blasche
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Swara Patel
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Adi Biram
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Tomas Castro-Dopico
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael D. Buck
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Richard R. Rodrigues
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Microbiome and Genetics Core, LICI, CCR, NCI, 37 Convent Drive, Bethesda, MD 20892-0001, USA
| | - Gry Juul Poulsen
- National Center of Excellence for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Faculty of Medicine, Aalborg University, Department of Gastroenterology and Hepatology, Aalborg University Hospital, A.C. Meyers Vænge 15, A DK-2450 Copenhagen, Denmark
| | | | - Neil C. Rogers
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Maria A. Koufaki
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Carlos M. Minutti
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Pengbo Wang
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Alexander Vdovin
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Bruno Frederico
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Eleanor Childs
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sonia Lee
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ben Simpson
- Tumor ImmunoGenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, 72 Huntley St, London WC1E 6DD, UK
| | - Andrea Iseppon
- AhRimmunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sara Omenetti
- AhRimmunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Gavin Kelly
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Robert Goldstone
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emma Nye
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alejandro Suárez-Bonnet
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - Simon L. Priestnall
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - James I. MacRae
- Metabolomics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Santiago Zelenay
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Kiran Raosaheb Patil
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Kevin Litchfield
- Tumor ImmunoGenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, 72 Huntley St, London WC1E 6DD, UK
| | - James C. Lee
- Genetic Mechanisms of Disease Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Institute of Liver and Digestive Health, Division of Medicine, Royal Free Hospital, University College London, London, NW3 2QG, UK
| | - Tine Jess
- National Center of Excellence for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Faculty of Medicine, Aalborg University, Department of Gastroenterology and Hepatology, Aalborg University Hospital, A.C. Meyers Vænge 15, A DK-2450 Copenhagen, Denmark
| | - Romina S. Goldszmid
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology (LICI), Center for Cancer Research (CCR), National Cancer Institute (NCI), 37 Convent Drive, Bethesda, MD 20892-0001, USA
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
20
|
Peña-Oyarzún D, Guzmán C, Kretschmar C, Torres VA, Maturana-Ramirez A, Aitken J, Reyes M. Calcitriol Treatment Decreases Cell Migration, Viability and β-Catenin Signaling in Oral Dysplasia. Curr Issues Mol Biol 2024; 46:3050-3062. [PMID: 38666921 PMCID: PMC11049246 DOI: 10.3390/cimb46040191] [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: 02/09/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Nearly 90% of oral cancers are characterized as oral squamous cell carcinoma (OSCC), representing the sixth most common type of cancer. OSCC usually evolves from oral potentially malignant disorders that, in some cases, are histologically consistent with a oral dysplasia. The levels of 1α,25 dihydroxyvitamin D3 (1,25-(OH)2D3; calcitriol), the active form of vitamin D3, have been shown to be decreased in patients with oral dysplasia and OSCC. Moreover, treatment with 1,25-(OH)2D3 has been proven beneficial in OSCC by inhibiting the Wnt/β-catenin pathway, a signaling route that promotes cell migration, proliferation, and viability. However, whether this inhibition mechanism occurs in oral dysplasia is unknown. To approach this question, we used dysplastic oral keratinocyte cultures and oral explants (ex vivo model of oral dysplasia) treated with 1,25-(OH)2D3 for 48 h. Following treatment with 1,25-(OH)2D3, both in vitro and ex vivo models of oral dysplasia showed decreased levels of nuclear β-catenin by immunofluorescence (IF) and immunohistochemistry (IHC). Consistently, reduced protein and mRNA levels of the Wnt/β-catenin target gene survivin were observed after treatment with 1,25-(OH)2D3. Moreover, 1,25-(OH)2D3 promoted membranous localization of E-cadherin and nuclear localization of vitamin D receptor (VDR). Functionally, DOK cells treated with 1,25-(OH)2D3 displayed diminished cell migration and viability in vitro.
Collapse
Affiliation(s)
- Daniel Peña-Oyarzún
- Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastián, Los Leones Campus, Santiago 7510157, Chile
| | - Constanza Guzmán
- Department of Pathology and Oral Medicine, Faculty of Odontology, Universidad de Chile, Santiago 8380544, Chile
| | - Catalina Kretschmar
- Institute for Research in Dental Sciences, Faculty of Odontology, Universidad de Chile, Santiago 8380544, Chile
| | - Vicente A. Torres
- Institute for Research in Dental Sciences, Faculty of Odontology, Universidad de Chile, Santiago 8380544, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago 8380453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago 8380494, Chile
| | - Andrea Maturana-Ramirez
- Department of Pathology and Oral Medicine, Faculty of Odontology, Universidad de Chile, Santiago 8380544, Chile
| | - Juan Aitken
- Department of Pathology and Oral Medicine, Faculty of Odontology, Universidad de Chile, Santiago 8380544, Chile
| | - Montserrat Reyes
- Department of Pathology and Oral Medicine, Faculty of Odontology, Universidad de Chile, Santiago 8380544, Chile
| |
Collapse
|
21
|
Wang P, Nie J, Li J, Ye C, Chen J, Zhang Z, Li B. VDRA downregulate β-catenin/Smad3 and DNA damage and repair associated with improved prognosis in ccRCC patients. Int J Biol Macromol 2024; 263:130405. [PMID: 38403213 DOI: 10.1016/j.ijbiomac.2024.130405] [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: 07/31/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
The clear cell renal cell carcinoma (ccRCC) spotlighted the poorest survival, while chromophobe renal cell carcinoma (chRCC) was associated with the best survival. Earlier studies corroborated vitamin D receptor (VDR) was a promising molecular for improving the prognosis of RCC. In contrast to VDRA, the one of VDR isoforms, VDRB1 (VDR isoform B1) has an N-terminal extension of 50 amino acids and is less ligand-dependent. However, the functional differences between VDRA and VDRB1, and their roles in the prognosis of ccRCC and chRCC, have not been investigated. In the present study, we uncovered that the transcripts related to vitamin D pathway and cellular calcium signaling were effectively decreased in the context of ccRCC, yet failed to exert a comparable effect within chRCC. Specially, minimally levels of VDRA wherein kidneys of patients suffering from ccRCC predict shorter survival time. In addition, the protein expressions for β-catenin/Smad3 pathway and DNA damage and repair pathways were obviously impeded in VDRA-overexpressed ccRCC cells, yet this inhibitory effect was conspicuously absent in enable VDRB1 cells. Our results provide a new idea to improve the prognosis of ccRCC via VDRA upregulation.
Collapse
Affiliation(s)
- Ping Wang
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jin Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Jiafu Li
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Caiyong Ye
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Jianwu Chen
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Fujian Province, China.
| | - Zengli Zhang
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, Suzhou, China.
| | - Bingyan Li
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
22
|
Cai F, Li J, Zhang Y, Huang S, Liu W, Zhuo W, Qiu C. Interaction between Wnt/β-catenin signaling pathway and EMT pathway mediates the mechanism of sunitinib resistance in renal cell carcinoma. BMC Cancer 2024; 24:175. [PMID: 38317072 PMCID: PMC10840195 DOI: 10.1186/s12885-024-11907-5] [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: 04/04/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Targeted drugs are the main methods of RCC treatment. However, drug resistance is common in RCC patients, in-depth study of the drug-resistant mechanism is essential. METHODS We constructed sunitinib resistant and Twist overexpressed A498 cells, and studied its mechanisms in vitro and in vivo. RESULTS In cell research, we found that either sunitinib resistance or Twist overexpression can activate Wnt/β-catenin and EMT signaling pathway, and the sunitinib resistance may work through β-catenin/TWIST/TCF4 trimer. In zebrafish research, we confirmed the similarity of Twist overexpression and sunitinib resistance, and the promoting effect of Twist overexpression on drug resistance. CONCLUSIONS Sunitinib resistance and Twist overexpression can activate Wnt/β-catenin signaling pathway and EMT to promote the growth and metastasis of RCC cells.
Collapse
Affiliation(s)
- Fangzhen Cai
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jianwei Li
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yanmei Zhang
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Sihuai Huang
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Wenbin Liu
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weifeng Zhuo
- Department of Urology, JinJiang Municipal Hospital, Quanzhou, Fujian, China
| | - Chengzhi Qiu
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| |
Collapse
|
23
|
Sobhi P, Bahrami M, Mahdizadeh F, Fazaeli A, Babaei G, Rezagholizadeh L. Vitamin D and potential effects on cancers: a review. Mol Biol Rep 2024; 51:190. [PMID: 38270702 DOI: 10.1007/s11033-023-09111-y] [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: 09/01/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Cancer is characterized by the abnormal and uncontrollable division and growth of cells that can infiltrate tissues and alter normal physiological function, which will become crucial and life-threatening if left untreated. Cancer can be a result of genetics, such as mutations or environmental causes, including smoking, lack of physical activity, as well as nutritional imbalance in the body. Vitamin D is one of the foremost nutrients that play a crucial role in a variety of biochemical pathways, and it is an important key factor in several diseases. Vitamin D is an essential nutrient for preventing malignancies and a complementary treatment for cancer through direct and indirect biochemical pathways. In this article, we summarized the correlation between vitamin D and various cancers using an extensive search on PubMed, Google Scholar, and Scopus. This paper reviews the role of vitamin D in different types of cancer.
Collapse
Affiliation(s)
- Pouria Sobhi
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Bahrami
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Faraz Mahdizadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Aliakbar Fazaeli
- Department of Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Lotfollah Rezagholizadeh
- Department of Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
24
|
Yan Q, Su X, Chen Y, Wang Z, Han W, Xia Q, Mao Y, Si J, Li H, Duan S. LINC00941: a novel player involved in the progression of human cancers. Hum Cell 2024; 37:167-180. [PMID: 37995050 DOI: 10.1007/s13577-023-01002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023]
Abstract
LINC00941, also known as lncRNA-MUF, is an intergenic non-coding RNA located on chromosome 12p11.21. It actively participates in a complex competing endogenous RNA network, regulating the expression of microRNA and its downstream proteins. Through transcriptional and post-transcriptional regulation, LINC00941 plays a vital role in multiple signaling pathways, influencing cell behaviors such as tumor cell proliferation, epithelial-mesenchymal transition, migration, and invasion. Noteworthy is its consistently high expression in various tumor types, closely correlating with clinicopathological features and cancer prognoses. Elevated LINC00941 levels are associated with adverse clinical outcomes, including increased tumor size, extensive lymphatic metastasis, and distant metastasis, leading to poorer survival rates across different cancers. Additionally, LINC00941 and its associated genes are linked to various targeted drugs available in the market. In this comprehensive review, we systematically summarize existing studies, detailing LINC00941's differential expression, clinicopathological and prognostic implications, regulatory mechanisms, and associated therapeutic drugs. Our analysis includes relevant charts and incorporates bioinformatics analyses to verify LINC00941's differential expression in pan-cancer and explore potential transcriptional regulation patterns of downstream targets. This work not only establishes a robust data foundation but also guides future research directions. Given its potential as a significant cancer biomarker and therapeutic target, further investigation into LINC00941's differential expression and regulatory mechanisms is essential.
Collapse
Affiliation(s)
- Qibin Yan
- Institute of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmacy, Hangzhou City University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinming Su
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Yunzhu Chen
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zehua Wang
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Wenbo Han
- Department of Pharmacy, Hangzhou City University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Qing Xia
- Institute of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Yunan Mao
- Department of Pharmacy, Hangzhou City University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiahua Si
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hanbing Li
- Institute of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang, China.
| | - Shiwei Duan
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
25
|
Dong Y, Wei J, Yang F, Qu Y, Huang J, Shi D. Nutrient-Based Approaches for Melanoma: Prevention and Therapeutic Insights. Nutrients 2023; 15:4483. [PMID: 37892558 PMCID: PMC10609833 DOI: 10.3390/nu15204483] [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: 09/22/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Melanoma, a prevalent and lethal form of skin cancer, remains a formidable challenge in terms of prevention and treatment. While significant progress has been made in understanding its pathogenesis and treatment, the quest for effective prevention strategies and therapeutic approaches remains ongoing. Considering the increased advancements in understanding the dynamic interplay between nutrients and melanoma, we aim to offer a refreshed perspective on nutrient-based approaches for melanoma prevention and adjunctive therapy. In contrast to other studies, we have innovatively provided a detailed exposition of the nutrients' influences on melanoma prognosis and treatment. This review firstly examines various nutrients, including antioxidants (namely vitamins A, D, C, and E; selenium; and caffeine), polyunsaturated fatty acids, and flavonoids, for their effects and underlying mechanisms in reducing melanoma risk. Among these nutrients, caffeine shows the most promising potential, as it is supported by multiple cohort studies for its protective effect against melanoma. In contrast, there is a certain degree of inconsistency in the research of other nutrients, possibly due to inherent differences between animal studies and epidemiological research, as well as variations in the definition of nutrient intake. To comprehensively investigate the impact of nutrients on melanoma progression and therapeutic approaches, the following sections will explore how nutrients influence immune responses and other physiological processes. While there is robust support from cell and animal studies regarding the immunomodulatory attributes of vitamins D and zinc, the anti-angiogenic potential of polyphenols, and the cell growth-inhibitory effects of flavonoids, the limited availability of human-based research substantially constrains their practical relevance in clinical contexts. As for utilizing nutrients in adjuvant melanoma treatments, multiple approaches have garnered clinical research support, including the utilization of vitamin D to decrease the postoperative recurrence rates among melanoma patients and the adoption of a high-fiber diet to enhance the effectiveness of immunotherapy. In general, the effects of most nutrients on reducing the risk of melanoma are not entirely clear. However, several nutrients, including vitamin D and dietary fiber, have demonstrated their potential to improve the melanoma prognosis and enhance the treatment outcomes, making them particularly deserving of clinical attention. A personalized and interdisciplinary approach, involving dermatologists, oncologists, nutritionists, and researchers, holds the promise of optimizing melanoma treatment strategies.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Yang Qu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Di Shi
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| |
Collapse
|
26
|
Donati S, Palmini G, Aurilia C, Falsetti I, Marini F, Giusti F, Iantomasi T, Brandi ML. Calcifediol: Mechanisms of Action. Nutrients 2023; 15:4409. [PMID: 37892484 PMCID: PMC10610216 DOI: 10.3390/nu15204409] [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: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Due to its essential role in calcium and phosphate homeostasis, the secosteroid hormone calcitriol has received growing attention over the last few years. Calcitriol, like other steroid hormones, may function through both genomic and non-genomic mechanisms. In the traditional function, the interaction between the biologically active form of vitamin D and the vitamin D receptor (VDR) affects the transcription of thousands of genes by binding to repeated sequences present in their promoter region, named vitamin D-responsive elements (VDREs). Non-transcriptional effects, on the other hand, occur quickly and are unaffected by inhibitors of transcription and protein synthesis. Recently, calcifediol, the immediate precursor metabolite of calcitriol, has also been shown to bind to the VDR with weaker affinity than calcitriol, thus exerting gene-regulatory properties. Moreover, calcifediol may also trigger rapid non-genomic responses through its interaction with specific membrane vitamin D receptors. Membrane-associated VDR (mVDR) and protein disulfide isomerase family A member 3 (Pdia3) are the best-studied candidates for mediating these rapid responses to vitamin D metabolites. This paper provides an overview of the calcifediol-related mechanisms of action, which may help to better understand the vitamin D endocrine system and to identify new therapeutic targets that could be important for treating diseases closely associated with vitamin D deficiency.
Collapse
Affiliation(s)
- Simone Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Cinzia Aurilia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Irene Falsetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Francesca Marini
- Fondazione Italiana Ricerca Sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy;
| | - Francesca Giusti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Maria Luisa Brandi
- Fondazione Italiana Ricerca Sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy;
- Donatello Bone Clinic, Villa Donatello Hospital, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
27
|
Burgermeister E. Mitogen-Activated Protein Kinase and Nuclear Hormone Receptor Crosstalk in Cancer Immunotherapy. Int J Mol Sci 2023; 24:13661. [PMID: 37686465 PMCID: PMC10488039 DOI: 10.3390/ijms241713661] [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: 06/28/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The three major MAP-kinase (MAPK) pathways, ERK1/2, p38 and JNK/SAPK, are upstream regulators of the nuclear "hormone" receptor superfamily (NHRSF), with a prime example given by the estrogen receptor in breast cancer. These ligand-activated transcription factors exert non-genomic and genomic functions, where they are either post-translationally modified by phosphorylation or directly interact with components of the MAPK pathways, events that govern their transcriptional activity towards target genes involved in cell differentiation, proliferation, metabolism and host immunity. This molecular crosstalk takes place not only in normal epithelial or tumor cells, but also in a plethora of immune cells from the adaptive and innate immune system in the tumor-stroma tissue microenvironment. Thus, the drugability of both the MAPK and the NHRSF pathways suggests potential for intervention therapies, especially for cancer immunotherapy. This review summarizes the existing literature covering the expression and function of NHRSF subclasses in human tumors, both solid and leukemias, and their effects in combination with current clinically approved therapeutics against immune checkpoint molecules (e.g., PD1).
Collapse
Affiliation(s)
- Elke Burgermeister
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| |
Collapse
|
28
|
Ito S, Liu X, Ishikawa Y, Conti DD, Otomo N, Kote-Jarai Z, Suetsugu H, Eeles RA, Koike Y, Hikino K, Yoshino S, Tomizuka K, Horikoshi M, Ito K, Uchio Y, Momozawa Y, Kubo M, Kamatani Y, Matsuda K, Haiman CA, Ikegawa S, Nakagawa H, Terao C. Androgen receptor binding sites enabling genetic prediction of mortality due to prostate cancer in cancer-free subjects. Nat Commun 2023; 14:4863. [PMID: 37612283 PMCID: PMC10447511 DOI: 10.1038/s41467-023-39858-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 06/27/2023] [Indexed: 08/25/2023] Open
Abstract
Prostate cancer (PrCa) is the second most common cancer worldwide in males. While strongly warranted, the prediction of mortality risk due to PrCa, especially before its development, is challenging. Here, we address this issue by maximizing the statistical power of genetic data with multi-ancestry meta-analysis and focusing on binding sites of the androgen receptor (AR), which has a critical role in PrCa. Taking advantage of large Japanese samples ever, a multi-ancestry meta-analysis comprising more than 300,000 subjects in total identifies 9 unreported loci including ZFHX3, a tumor suppressor gene, and successfully narrows down the statistically finemapped variants compared to European-only studies, and these variants strongly enrich in AR binding sites. A polygenic risk scores (PRS) analysis restricting to statistically finemapped variants in AR binding sites shows among cancer-free subjects, individuals with a PRS in the top 10% have a strongly higher risk of the future death of PrCa (HR: 5.57, P = 4.2 × 10-10). Our findings demonstrate the potential utility of leveraging large-scale genetic data and advanced analytical methods in predicting the mortality of PrCa.
Collapse
Affiliation(s)
- Shuji Ito
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Bone and Joint Diseases, Yokohama, Japan
- Department of Orthopedic Surgery, Shimane University, Izumo, Japan
| | - Xiaoxi Liu
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Yuki Ishikawa
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - David D Conti
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Nao Otomo
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
- Department of Orthopedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | | | - Hiroyuki Suetsugu
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Yoshinao Koike
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Keiko Hikino
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Pharmacogenomics, Yokohama, Japan
| | - Soichiro Yoshino
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Tomizuka
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Momoko Horikoshi
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Genomics of Diabetes and Metabolism, Yokohama, Japan
| | - Kaoru Ito
- RIKEN Center for Integrative Medical Sciences, The Cardiovascular Genomics and Informatics, Yokohama, Japan
| | - Yuji Uchio
- Department of Orthopedic Surgery, Shimane University, Izumo, Japan
| | - Yukihide Momozawa
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Genotyping Development, Yokohama, Japan
| | | | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Institute of Medical Science, The University of Tokyo, Laboratory of Genome Technology, Human Genome Center, Tokyo, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Tokyo, Japan
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shiro Ikegawa
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Bone and Joint Diseases, Yokohama, Japan
| | - Hidewaki Nakagawa
- RIKEN Center for Integrative Medical Sciences, Laboratory for Cancer Genomics, Yokohama, Japan
| | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan.
- Shizuoka General Hospital, The Clinical Research Center, Shizuoka, Japan.
- School of Pharmaceutical Sciences, University of Shizuoka, The Department of Applied Genetics, Shizuoka, Japan.
| |
Collapse
|
29
|
Gong D, Adomako-Bonsu AG, Wang M, Li J. Three specific gut bacteria in the occurrence and development of colorectal cancer: a concerted effort. PeerJ 2023; 11:e15777. [PMID: 37554340 PMCID: PMC10405800 DOI: 10.7717/peerj.15777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/29/2023] [Indexed: 08/10/2023] Open
Abstract
Colorectal cancer (CRC), which develops from the gradual evolution of tubular adenomas and serrated polyps in the colon and rectum, has a poor prognosis and a high mortality rate. In addition to genetics, lifestyle, and chronic diseases, intestinal integrity and microbiota (which facilitate digestion, metabolism, and immune regulation) could promote CRC development. For example, enterotoxigenic Bacteroides fragilis, genotoxic Escherichia coli (pks+ E. coli), and Fusobacterium nucleatum, members of the intestinal microbiota, are highly correlated in CRC. This review describes the roles and mechanisms of these three bacteria in CRC development. Their interaction during CRC initiation and progression has also been proposed. Our view is that in the precancerous stage of colorectal cancer, ETBF causes inflammation, leading to potential changes in intestinal ecology that may provide the basic conditions for pks+ E. coli colonization and induction of oncogenic mutations, when cancerous intestinal epithelial cells can further recruit F. nucleatum to colonise the lesion site and F. nucleatum may contribute to CRC advancement by primarily the development of cancer cells, stemization, and proliferation, which could create new and tailored preventive, screening and therapeutic interventions. However, there is the most dominant microbiota in each stage of CRC development, not neglecting the possibility that two or even all three bacteria could be engaged at any stage of the disease. The relationship between the associated gut microbiota and CRC development may provide important information for therapeutic strategies to assess the potential use of the associated gut microbiota in CRC studies, antibiotic therapy, and prevention strategies.
Collapse
Affiliation(s)
- Dengmei Gong
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Amma G Adomako-Bonsu
- Institute of Toxicology and Pharmacology, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Maijian Wang
- Gastrointestinal Surgery, Affiliate Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jida Li
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
30
|
Shellenberger RA, Gowda S, Kurn H, Albright J, Mayo MH. Vitamin D insufficiency and serum levels related to the incidence and stage of cutaneous melanoma: a systematic review and meta-analysis. Melanoma Res 2023; 33:265-274. [PMID: 37199748 DOI: 10.1097/cmr.0000000000000897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Cutaneous melanoma (CM) is the deadliest skin cancer, and vitamin D insufficiency has been suggested as a risk factor. We evaluated the relationship between both vitamin D insufficiency and 25-hydroxy vitamin D levels with the incidence and stage of CM. Five databases were searched from inception until 11 July 2022. Inclusion criteria were cohort and case-control studies that reported mean 25-hydroxy vitamin D levels or the presence of vitamin D insufficiency in CM patients and compared with healthy controls; or those that reported vitamin D insufficiency and Breslow tumor depth or the development of metastasis in CM. Fourteen studies were included in the analysis. Statistically significant relationships were found between vitamin D level <20 ng/dl and incidence of CM [pooled RR 1.45, 95% confidence interval (CI) 1.04-2.02]; lower mean vitamin D level and Breslow depth >1 mm (SMD 0.19, 95% CI 0.11-0.28); and vitamin D level >20 ng/dl and Breslow depth <1 mm (pooled RR 0.69, 95% CI 0.58-0.82). Statistical significance was not found in the relationships between vitamin D levels and the presence of metastasis (pooled SMD -0.13, 95% CI, -0.38 to 0.12); or mean vitamin D level and the incidence of CM (pooled SMD -0.39, 95% CI, -0.80 to 0.01). We identified an association of increased incidence of CM and vitamin D insufficiency, as well as less favorable Breslow tumor depth with lower levels of vitamin D and the presence of vitamin D insufficiency.
Collapse
Affiliation(s)
| | - Sunaina Gowda
- Department of Internal Medicine, Trinity Health Ann Arbor Hospital, Ann Arbor, Michigan
| | - Heidi Kurn
- Department of Internal Medicine, Trinity Health Ann Arbor Hospital, Ann Arbor, Michigan
| | | | - MacKenzie H Mayo
- Department of Internal Medicine, Trinity Health Ann Arbor Hospital, Ann Arbor, Michigan
| |
Collapse
|
31
|
Nautiyal R, Bhatnagar P. Deciphering the role of vitamin D on skin cancers and tumour microenvironment. Indian J Dermatol Venereol Leprol 2023; 0:1-10. [PMID: 37609744 DOI: 10.25259/ijdvl_1236_2021] [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: 12/31/2021] [Accepted: 05/15/2023] [Indexed: 08/24/2023]
Abstract
Skin cancer is a significant health burden being the fourth most common cancer globally and accounts for 6.2% of the total combined cancer cases. However, mortality rates due to skin cancer are less when compared with other cancers, but it is significantly high in the Asian population (43%). DNA mutations and environmental and genetic factors are linked with skin cancer prognosis; however, long-term exposure to ultraviolet (UV) radiation remains one of the leading factors worldwide. Sun exposure is a major environmental risk factor for skin cancers but is also an essential source of vitamin D. On the other hand, studies exploring the relationship between skin cancer risk and vitamin D show mixed, somewhat conflicting results. This study investigates the role of vitamin D and skin carcinogenesis to clarify the associations. Moreover, in addition to suppressing cancer stem cells, it has been observed that vitamin D also regulates tumor initiation and metastasis. In conclusion, the incorporation of well-designed studies on the metabolism of vitamin D from a genotypic and phenotypic perspective is required to understand the intricate mechanisms linking the role of vitamin D in skin carcinogenesis. These new findings will open up new pathways in targeting the disease and lead to novel opportunities for its treatment and cure.
Collapse
Affiliation(s)
- Rohit Nautiyal
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Hyderabad, India
| | - Priyanka Bhatnagar
- Disease Biology Segment, Quick IsCool, Aitele Research LLP, Bihar, India
| |
Collapse
|
32
|
Shang L, Li J, Zhou F, Zhang M, Wang S, Yang S. MiR-874-5p targets VDR/NLRP3 to reduce intestinal pyroptosis and improve intestinal barrier damage in sepsis. Int Immunopharmacol 2023; 121:110424. [PMID: 37315369 DOI: 10.1016/j.intimp.2023.110424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Vitamin D receptor (VDR) is associated with intestinal barrier damage in sepsis. However, the mechanism of action of miR-874-5p/VDR/NLRP3 axis in disease has not been clearly explained. Therefore, the main content of this study is to explore the mechanism of this axis in intestinal barrier damage in sepsis. METHODS In order to confirm the progress of miR-874-5p regulation of VDR/NLRP3 pathway and its involvement in intestinal barrier damage in sepsis, a series of molecular biology and cell biology methods were carried out in this study. These include the establishment of cecal ligation puncture model, Western blot, RT-qPCR, hematoxylin and eosin staining, double luciferase reporting method, Fluorescence in situ hybridization, immunohistochemistry, and enzyme-linked immunosorption assay. RESULTS The expression level of miR-874-5p was higher and that of VDR was lower in sepsis. miR-874-5p was negatively correlated with VDR. Inhibition of miR-874-5p expression increased the expression of VDR, decreased the expression of NLRP3, reduced caspase-1 activation and IL-1β secretion, reduced pyroptosis and inflammatory response, and thus protected the intestinal barrier damage in sepsis, all of which were reversed by the downregulation of VDR. CONCLUSIONS This study suggested that down-regulation of miR-874-5p or up-regulation of VDR could reduce intestinal barrier damage in sepsis, which may provide potential biomarkers and therapeutic targets for intestinal barrier damage in sepsis.
Collapse
Affiliation(s)
- Luorui Shang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxiao Li
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangyuan Zhou
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengqi Zhang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuhan Wang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenglan Yang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
33
|
Starska-Kowarska K. Role of Vitamin D in Head and Neck Cancer-Immune Function, Anti-Tumour Effect, and Its Impact on Patient Prognosis. Nutrients 2023; 15:nu15112592. [PMID: 37299554 DOI: 10.3390/nu15112592] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/13/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) describes a heterogeneous group of human neoplasms of the head and neck with high rates of morbidity and mortality, constituting about 3% of all cancers and ~1.5% of all cancer deaths. HNSCC constituted the seventh most prevalent human malignancy and the most common human cancer in the world in 2020, according to multi-population observations conducted by the GLOBOCAN group. Since approximately 60-70% of patients present with stage III/IV neoplastic disease, HNSCC is still one of the leading causes of death in cancer patients worldwide, with an overall survival rate that is too low, not exceeding 40-60% of these patients. Despite the application of newer surgical techniques and the implementation of modern combined oncological treatment, the disease often follows a fatal course due to frequent nodal metastases and local neoplastic recurrences. The role of micronutrients in the initiation, development, and progression of HNSCC has been the subject of considerable research. Of particular interest has been vitamin D, the pleiotropic biologically active fat-soluble family of secosteroids (vitamin-D-like steroids), which constitutes a key regulator of bone, calcium, and phosphate homeostasis, as well as carcinogenesis and the further development of various neoplasms. Considerable evidence suggests that vitamin D plays a key role in cellular proliferation, angiogenesis, immunity, and cellular metabolism. A number of basic science, clinical, and epidemiological studies indicate that vitamin D has multidirectional biological effects and influences anti-cancer intracellular mechanisms and cancer risk, and that vitamin D dietary supplements have various prophylactic benefits. In the 20th century, it was reported that vitamin D may play various roles in the protection and regulation of normal cellular phenotypes and in cancer prevention and adjunctive therapy in various human neoplasms, including HNSCC, by regulating a number of intracellular mechanisms, including control of tumour cell expansion and differentiation, apoptosis, intercellular interactions, angio- and lymphogenesis, immune function, and tumour invasion. These regulatory properties mainly occur indirectly via epigenetic and transcriptional changes regulating the function of transcription factors, chromatin modifiers, non-coding RNA (ncRNAs), and microRNAs (miRs) through protein-protein interactions and signalling pathways. In this way, calcitriol enhances intercellular communication in cancer biology, restores the connection with the extracellular matrix, and promotes the epithelial phenotype; it thus counteracts the tumour-associated detachment from the extracellular matrix and inhibits the formation of metastases. Furthermore, the confirmation that the vitamin D receptor (VDR) is present in many human tissues confirmed the physiopathological significance of vitamin D in various human tumours. Recent studies indicate quantitative associations between exposure to vitamin D and the incidence of HNC, i.e., cancer risk assessment included circulating calcidiol plasma/serum concentrations, vitamin D intake, the presence of the VDR gene polymorphism, and genes involved in the vitamin D metabolism pathway. Moreover, the chemopreventive efficacy of vitamin D in precancerous lesions of the head and neck and their role as predictors of mortality, survival, and recurrence of head and neck cancer are also widely discussed. As such, it may be considered a promising potential anti-cancer agent for developing innovative methods of targeted therapy. The proposed review discusses in detail the mechanisms regulating the relationship between vitamin D and HNSCC. It also provides an overview of the current literature, including key opinion-forming systematic reviews as well as epidemiological, prospective, longitudinal, cross-sectional, and interventional studies based on in vitro and animal models of HNSCC, all of which are accessible via the PubMed/Medline/EMBASE/Cochrane Library databases. This article presents the data in line with increasing clinical credibility.
Collapse
Affiliation(s)
- Katarzyna Starska-Kowarska
- Department of Physiology, Pathophysiology and Clinical Immunology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Clinical Physiology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Otorhinolaryngology, EnelMed Center Expert, Lodz, Drewnowska 58, 91-001 Lodz, Poland
| |
Collapse
|
34
|
Gao W, Liang Y, Wu D, Deng S, Qiu R. Graphene quantum dots enhance the osteogenic differentiation of PDLSCs in the inflammatory microenvironment. BMC Oral Health 2023; 23:331. [PMID: 37244994 DOI: 10.1186/s12903-023-03026-7] [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: 11/29/2022] [Accepted: 05/09/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Graphene quantum dots (GQDs), a type of carbon-based nanomaterial, have remarkable biological, physical, and chemical properties. This study investigated the biological mechanisms of the proliferation and osteogenic differentiation of human periodontal ligament stem cells (PDLSCs) induced by GQDs in an inflammatory microenvironment. MATERIALS AND METHODS PDLSCs were cultured in osteogenic-induced medium with various concentrations of GQDs in standard medium or medium mimicking a proinflammatory environment. The effects of GQDs on the proliferation and osteogenic differentiation activity of PDLSCs were tested by CCK-8 assay, Alizarin Red S staining, and qRT‒PCR. In addition, Wnt/β-catenin signalling pathway-related gene expression was measured by qRT‒PCR. RESULTS Compared with the control group, the mRNA expression levels of ALP, RUNX2, and OCN and the number of mineralized nodules were all increased in PDLSCs after treatment with GQDs. Moreover, during the osteogenic differentiation of PDLSCs, the expression levels of LRP6 and β-catenin, which are Wnt/β-catenin signalling pathway-related genes, were upregulated. CONCLUSION In the inflammatory microenvironment, GQDs might promote the osteogenic differentiation ability of PDLSCs by activating the Wnt/β-catenin signalling pathway.
Collapse
Grants
- No.2021KY0119 Project of Basic Research Capacity Improvement in Young and Middle-aged Teachers in Guangxi universities
- No.2021KY0119 Project of Basic Research Capacity Improvement in Young and Middle-aged Teachers in Guangxi universities
- No.2021KY0119 Project of Basic Research Capacity Improvement in Young and Middle-aged Teachers in Guangxi universities
- No.2021KY0119 Project of Basic Research Capacity Improvement in Young and Middle-aged Teachers in Guangxi universities
- No.2021KY0119 Project of Basic Research Capacity Improvement in Young and Middle-aged Teachers in Guangxi universities
- NO.S2020041 Guangxi Medical and Health appropriate Technology Development and Promotion and Application Project
- NO.S2020041 Guangxi Medical and Health appropriate Technology Development and Promotion and Application Project
- NO.S2020041 Guangxi Medical and Health appropriate Technology Development and Promotion and Application Project
- NO.S2020041 Guangxi Medical and Health appropriate Technology Development and Promotion and Application Project
- NO.S2020041 Guangxi Medical and Health appropriate Technology Development and Promotion and Application Project
- NO.2020039 Science and Technology Plan Project of Qingxiu District, Nanning City, Guangxi
- NO.2020039 Science and Technology Plan Project of Qingxiu District, Nanning City, Guangxi
- NO.2020039 Science and Technology Plan Project of Qingxiu District, Nanning City, Guangxi
- NO.2020039 Science and Technology Plan Project of Qingxiu District, Nanning City, Guangxi
- NO.2020039 Science and Technology Plan Project of Qingxiu District, Nanning City, Guangxi
- NO. 2021AB11097 Key R & D projects of Guangxi science and Technology Department
- NO. 2021AB11097 Key R & D projects of Guangxi science and Technology Department
- NO. 2021AB11097 Key R & D projects of Guangxi science and Technology Department
- NO. 2021AB11097 Key R & D projects of Guangxi science and Technology Department
- NO. 2021AB11097 Key R & D projects of Guangxi science and Technology Department
Collapse
Affiliation(s)
- Wanshan Gao
- College of Stomatology, Hospital of Stomatology Guangxi Medical University , Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, 530021, Guangxi, China
| | - Yan Liang
- College of Stomatology, Hospital of Stomatology Guangxi Medical University , Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, 530021, Guangxi, China
| | - Dongyan Wu
- College of Stomatology, Hospital of Stomatology Guangxi Medical University , Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, 530021, Guangxi, China
| | - Sicheng Deng
- College of Stomatology, Hospital of Stomatology Guangxi Medical University , Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, 530021, Guangxi, China
| | - Rongmin Qiu
- College of Stomatology, Hospital of Stomatology Guangxi Medical University , Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, 530021, Guangxi, China.
- Key Laboratory of Research and Application of Stomatological Equipment College of Stomatology Hospital of Stomatology Guangxi Medical University, Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China, 530021.
| |
Collapse
|
35
|
Donati S, Marini F, Giusti F, Palmini G, Aurilia C, Falsetti I, Iantomasi T, Brandi ML. Calcifediol: Why, When, How Much? Pharmaceuticals (Basel) 2023; 16:ph16050637. [PMID: 37242420 DOI: 10.3390/ph16050637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Vitamin D deficiency is a constantly growing health problem worldwide. Adults affected with hypovitaminosis D could experience negative consequences on their musculoskeletal system and extra-skeletal health. In fact, an optimal vitamin D status is essential to ensure the correct bone, calcium, and phosphate homeostasis. To improve vitamin D status, it is important to not only increase the intake of food fortified with vitamin D, but also to administer vitamin D supplementation when required. Vitamin D3 (cholecalciferol) is the most widely used supplement. In recent years, the administration of calcifediol (25(OH)D3), the direct precursor of the biologically active form of vitamin D3, as oral vitamin D supplementation has progressively grown. Here, we report the potential medical benefits of some peculiar biological actions of calcifediol, discussing the possible specific clinical scenarios in which the oral intake of calcifediol could be most effective to restore the correct serum levels of 25(OH)D3. In summary, the aim of this review is to provide insights into calcifediol-related rapid non-genomic responses and the possible use of this vitamin D metabolite as a supplement for the treatment of people with a higher risk of hypovitaminosis D.
Collapse
Affiliation(s)
- Simone Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Francesca Marini
- Fondazione Italiana Ricerca Sulle Malattie dell'Osso (FIRMO Onlus), 50129 Florence, Italy
| | - Francesca Giusti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Cinzia Aurilia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Irene Falsetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Maria Luisa Brandi
- Fondazione Italiana Ricerca Sulle Malattie dell'Osso (FIRMO Onlus), 50129 Florence, Italy
| |
Collapse
|
36
|
Yu J, Sun Q, Hui Y, Xu J, Shi P, Chen Y, Chen Y. Vitamin D receptor prevents tumour development by regulating the Wnt/β-catenin signalling pathway in human colorectal cancer. BMC Cancer 2023; 23:336. [PMID: 37046222 PMCID: PMC10091620 DOI: 10.1186/s12885-023-10690-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 02/28/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common disease threatening human lives worldwide, and vitamin D receptor (VDR) contributes protective roles in this disease. However, the molecular mechanisms underlying VDR protection in CRC progression require further investigation. METHODS In this study, we statistically analyzed the relationship between VDR expression and CRC development in patients and detected invasion and apoptosis in CRC cells with VDR overexpression and interference. We also detected the expression of key genes involved in Wnt/β-catenin signalling (β-catenin, lymphoid enhancer factor (LEF)-1 and cyclin D1) in SW480 cells and nude mice injected with VDR-overexpressing SW480 cells and observed tumour development. Additionally, we performed Co-immunoprecipitation (Co-IP) and glutathione-S-transferase (GST) pull-down assays to identify the protein interactions of VDR with β-catenin, dual luciferase (LUC) and chromatin immunoprecipitation (ChIP) to detect the activation of LEF-1 by VDR. RESULTS The VDR level was closely related to the development and prognosis of CRC patients. VDR overexpression inhibited invasion but promoted apoptosis in cancer cells. β-catenin shRNA contributed oppositely to cancer cell activity with VDR shRNA. Additionally, VDR interacted with β-catenin at the protein level and blocked its nuclear accumulation. VDR regulated the expression of β-catenin, cyclin D1 and LEF-1 and directly activated LEF-1 transcription in vitro. Furthermore, nude mice injected with VDR-overexpressing SW480 cells revealed suppression of tumour growth and decreased expression of β-catenin, cyclin D1 and LEF-1. CONCLUSIONS This study indicated that VDR protected against CRC disease in humans by inhibiting Wnt/β-catenin signalling to control cancer cell invasion and apoptosis, providing new evidence to explore VDR biomarkers or agonists for CRC patient diagnosis and treatment.
Collapse
Affiliation(s)
- Jie Yu
- Department of Pathology, The People's Hospital of Suzhou New District, No. 95, Huashan Road, High Tech Zone, Suzhou, Jiangsu Prov, China
| | - Qi Sun
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yi Hui
- Department of Pathology, The People's Hospital of Suzhou New District, No. 95, Huashan Road, High Tech Zone, Suzhou, Jiangsu Prov, China
| | - Jinping Xu
- Department of Pathology, The People's Hospital of Suzhou New District, No. 95, Huashan Road, High Tech Zone, Suzhou, Jiangsu Prov, China
| | - Pancheng Shi
- Department of Pathology, The People's Hospital of Suzhou New District, No. 95, Huashan Road, High Tech Zone, Suzhou, Jiangsu Prov, China
| | - Yu Chen
- Department of Pathology, The People's Hospital of Suzhou New District, No. 95, Huashan Road, High Tech Zone, Suzhou, Jiangsu Prov, China
| | - Yunzhao Chen
- Department of Pathology, The People's Hospital of Suzhou New District, No. 95, Huashan Road, High Tech Zone, Suzhou, Jiangsu Prov, China.
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, China.
| |
Collapse
|
37
|
Zou Y, Yaguchi T. Programmed cell death-1 blockade therapy in melanoma: Resistance mechanisms and combination strategies. Exp Dermatol 2023; 32:264-275. [PMID: 36645031 DOI: 10.1111/exd.14750] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023]
Abstract
Melanoma is a highly aggressive tumor derived from melanocytes. In recent years, the incidence and mortality of melanoma have gradually increased, seriously threatening human health. Classic treatments like surgery, chemotherapy, and radiotherapy show very limited efficacy. Due to the high immunogenicity of melanoma cells, immune checkpoint inhibitors have received considerable attention as melanoma treatments. One such therapy is blockade of programmed cell death-1 (PD-1), which is one of the most important negative immune regulators and is mainly expressed on activated T cells. Disruption of the interactions between PD-1 and its ligands, programmed death-ligand 1 (PD-L1) or programmed death-ligand 2 (PD-L2) rejuvenates exhausted T cells and enhances antitumor immunity. Although PD-1 blockade therapy is widely used in melanoma, a substantial proportion of patients still show no response or short durations of remission. Recent researches have focused on revealing the underlying mechanisms for resistance to this treatment and improving its efficacy through combination therapy. Here, we will introduce the resistance mechanisms associated with PD-1 blockade therapy in melanoma and review the combination therapies available.
Collapse
Affiliation(s)
- Yixin Zou
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomonori Yaguchi
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
38
|
Żmijewski MA. Nongenomic Activities of Vitamin D. Nutrients 2022; 14:nu14235104. [PMID: 36501134 PMCID: PMC9737885 DOI: 10.3390/nu14235104] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/04/2022] Open
Abstract
Vitamin D shows a variety of pleiotropic activities which cannot be fully explained by the stimulation of classic pathway- and vitamin D receptor (VDR)-dependent transcriptional modulation. Thus, existence of rapid and nongenomic responses to vitamin D was suggested. An active form of vitamin D (calcitriol, 1,25(OH)2D3) is an essential regulator of calcium-phosphate homeostasis, and this process is tightly regulated by VDR genomic activity. However, it seems that early in evolution, the production of secosteroids (vitamin-D-like steroids) and their subsequent photodegradation served as a protective mechanism against ultraviolet radiation and oxidative stress. Consequently, direct cell-protective activities of vitamin D were proven. Furthermore, calcitriol triggers rapid calcium influx through epithelia and its uptake by a variety of cells. Subsequently, protein disulfide-isomerase A3 (PDIA3) was described as a membrane vitamin D receptor responsible for rapid nongenomic responses. Vitamin D was also found to stimulate a release of secondary massagers and modulate several intracellular processes-including cell cycle, proliferation, or immune responses-through wingless (WNT), sonic hedgehog (SSH), STAT1-3, or NF-kappaB pathways. Megalin and its coreceptor, cubilin, facilitate the import of vitamin D complex with vitamin-D-binding protein (DBP), and its involvement in rapid membrane responses was suggested. Vitamin D also directly and indirectly influences mitochondrial function, including fusion-fission, energy production, mitochondrial membrane potential, activity of ion channels, and apoptosis. Although mechanisms of the nongenomic responses to vitamin D are still not fully understood, in this review, their impact on physiology, pathology, and potential clinical applications will be discussed.
Collapse
Affiliation(s)
- Michał A Żmijewski
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, PL-80211 Gdańsk, Poland
| |
Collapse
|
39
|
Bueno AC, More CB, Marrero-Gutiérrez J, de Almeida E Silva DC, Leal LF, Montaldi AP, Ramalho FS, Vêncio RZN, de Castro M, Antonini SRR. Vitamin D receptor activation is a feasible therapeutic target to impair adrenocortical tumorigenesis. Mol Cell Endocrinol 2022; 558:111757. [PMID: 36049598 DOI: 10.1016/j.mce.2022.111757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To evaluate the therapeutic potential of vitamin D receptor (VDR) signaling in adrenocortical carcinoma (ACC) cells. METHODS We evaluated VDR's methylation pattern in H295R ACC cells, and investigated the effects of calcitriol and seocalcitol treatments on adrenocortical tumorigenesis. RESULTS VDR was hypermethylated and underexpressed in basal H295R cells. Treatments with calcitriol and seocalcitol restored VDR signaling, resulted in antiproliferative effects, and impaired Wnt/B-catenin signaling. RNAseq of treated cells demonstrated VDR activation on steroid hormones biosynthesis and Rap1 signaling, among others. In vivo, seocalcitol constrained the growth of H295R xenografts and reduced autonomous tumor steroid secretion without hypercalcemia-associated side effects. CONCLUSIONS H295R cells present VDR hypermethylation, which can be responsible for its underexpression and signaling inactivation under basal conditions. VDR signaling promoted antiproliferative effects in vitro and in vivo, suggesting that it may be a potential therapeutic target for ACC and a valuable tool for patient's clinical management.
Collapse
Affiliation(s)
- Ana Carolina Bueno
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Candy Bellido More
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Junier Marrero-Gutiérrez
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Danillo C de Almeida E Silva
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Leticia Ferro Leal
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ana Paula Montaldi
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Fernando Silva Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ricardo Zorzetto Nicoliello Vêncio
- Department of Computation and Mathematics, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Margaret de Castro
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sonir Roberto R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
40
|
Portales-Castillo I, Simic P. PTH, FGF-23, Klotho and Vitamin D as regulators of calcium and phosphorus: Genetics, epigenetics and beyond. Front Endocrinol (Lausanne) 2022; 13:992666. [PMID: 36246903 PMCID: PMC9558279 DOI: 10.3389/fendo.2022.992666] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
The actions of several bone-mineral ion regulators, namely PTH, FGF23, Klotho and 1,25(OH)2 vitamin D (1,25(OH)2D), control calcium and phosphate metabolism, and each of these molecules has additional biological effects related to cell signaling, metabolism and ultimately survival. Therefore, these factors are tightly regulated at various levels - genetic, epigenetic, protein secretion and cleavage. We review the main determinants of mineral homeostasis including well-established genetic and post-translational regulators and bring attention to the epigenetic mechanisms that affect the function of PTH, FGF23/Klotho and 1,25(OH)2D. Clinically relevant epigenetic mechanisms include methylation of cytosine at CpG-rich islands, histone deacetylation and micro-RNA interference. For example, sporadic pseudohypoparathyroidism type 1B (PHP1B), a disease characterized by resistance to PTH actions due to blunted intracellular cAMP signaling at the PTH/PTHrP receptor, is associated with abnormal methylation at the GNAS locus, thereby leading to reduced expression of the stimulatory G protein α-subunit (Gsα). Post-translational regulation is critical for the function of FGF-23 and such modifications include glycosylation and phosphorylation, which regulate the cleavage of FGF-23 and hence the proportion of available FGF-23 that is biologically active. While there is extensive data on how 1,25(OH)2D and the vitamin D receptor (VDR) regulate other genes, much more needs to be learned about their regulation. Reduced VDR expression or VDR mutations are the cause of rickets and are thought to contribute to different disorders. Epigenetic changes, such as increased methylation of the VDR resulting in decreased expression are associated with several cancers and infections. Genetic and epigenetic determinants play crucial roles in the function of mineral factors and their disorders lead to different diseases related to bone and beyond.
Collapse
Affiliation(s)
- Ignacio Portales-Castillo
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Petra Simic
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
41
|
Al-kuraishy HM, Batiha GES, Faidah H, Al-Gareeb AI, Saad HM, Simal-Gandara J. Pirfenidone and post-Covid-19 pulmonary fibrosis: invoked again for realistic goals. Inflammopharmacology 2022; 30:2017-2026. [PMID: 36044102 PMCID: PMC9430017 DOI: 10.1007/s10787-022-01027-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/20/2022] [Indexed: 01/10/2023]
Abstract
Pirfenidone (PFN) is an anti-fibrotic drug with significant anti-inflammatory property used for treatment of fibrotic conditions such as idiopathic pulmonary fibrosis (IPF). In the coronavirus disease 2019 (Covid-19) era, severe acute respiratory syndrome 2 (SARS-CoV-2) could initially lead to acute lung injury (ALI) and in severe cases may cause acute respiratory distress syndrome (ARDS) which is usually resolved with normal lung function. However, some cases of ALI and ARDS are progressed to the more severe critical stage of pulmonary fibrosis commonly named post-Covid-19 pulmonary fibrosis which needs an urgent address and proper management. Therefore, the objective of the present study was to highlight the potential role of PFN in the management of post-Covid-19 pulmonary fibrosis. The precise mechanism of post-Covid-19 pulmonary fibrosis is related to the activation of transforming growth factor beta (TGF-β1), which activates the release of extracellular proteins, fibroblast proliferation, fibroblast migration and myofibroblast conversion. PFN inhibits accumulation and recruitment of inflammatory cells, fibroblast proliferation, deposition of extracellular matrix in response to TGFβ1 and other pro-inflammatory cytokines. In addition, PFN suppresses furin (TGFβ1 convertase activator) a protein effector involved in the entry of SARS-CoV-2 and activation of TGFβ1, and thus PFN reduces the pathogenesis of SARS-CoV-2. Besides, PFN modulates signaling pathways such as Wingless/Int (Wnt/β-catenin), Yes-Associated Protein (YAP)/Transcription Co-Activator PDZ Binding Motif (TAZ) and Hippo Signaling Pathways that are involved in the pathogenesis of post-Covid-19 pulmonary fibrosis. In conclusion, the anti-inflammatory and anti-fibrotic properties of PFN may attenuate post-Covid-19 pulmonary fibrosis.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, M.B.Ch.B, FRCP, Baghdad, Iraq
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| | - Hani Faidah
- Microbiolgy Department Faculty of Medicine, Umm Al Qura University, Mecca, Saudi Arabia
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, M.B.Ch.B, FRCP, Baghdad, Iraq
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744 Matrouh Egypt
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade de Vigo, E-32004 Ourense, Spain
| |
Collapse
|
42
|
Liu T, Huang T, Shang M, Han G. CircRNA ITCH: Insight Into Its Role and Clinical Application Prospect in Tumor and Non-Tumor Diseases. Front Genet 2022; 13:927541. [PMID: 35910224 PMCID: PMC9335290 DOI: 10.3389/fgene.2022.927541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
CircRNA E3 ubiquitin protein ligase (ITCH) (circRNA ITCH, circ-ITCH), a stable closed-loop RNA derived from the 20q11.22 region of chromosome 20, is a new circRNA discovered in the cytoplasm in recent decades. Studies have shown that it does not encode proteins, but regulates proteins expression at different levels. It is down-regulated in tumor diseases and is involved in a number of biological activities, including inhibiting cell proliferation, migration, invasion, and promoting apoptosis. It can also alter disease progression in non-tumor disease by affecting the cell cycle, inflammatory response, and critical proteins. Circ-ITCH also holds a lot of promise in terms of tumor and non-tumor clinical diagnosis, prognosis, and targeted therapy. As a result, in order to aid clinical research in the hunt for a new strategy for diagnosing and treating human diseases, this study describes the mechanism of circ-ITCH as well as its clinical implications.
Collapse
Affiliation(s)
- Tong Liu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tao Huang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Mei Shang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Gang Han
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Hutchinson PE, Pringle JH. Consideration of possible effects of vitamin D on established cancer, with reference to malignant melanoma. Pigment Cell Melanoma Res 2022; 35:408-424. [PMID: 35445563 PMCID: PMC9322395 DOI: 10.1111/pcmr.13040] [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: 11/16/2021] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 12/11/2022]
Abstract
Epidemiological studies indicate that Vitamin D has a beneficial, inhibitory effect on cancer development and subsequent progression, including melanoma (MM), and favourable MM outcome has been reported as directly related to vitamin D3 status, assessed by serum 25-hydroxyvitamin D3 (25[OH]D3 ) levels taken at diagnosis. It has been recommended that MM patients with deficient levels of 25(OH)D3 be given vitamin D3 . We examine possible beneficial or detrimental effects of treating established cancer with vitamin D3 . We consider the likely biological determinants of cancer outcome, the reported effects of vitamin D3 on these in both cancerous and non-cancerous settings, and how the effect of vitamin D3 might change depending on the integrity of tumour vitamin D receptor (VDR) signalling. We would argue that the effect of defective tumour VDR signalling could result in loss of suppression of growth, reduction of anti-tumour immunity, with potential antagonism of the elimination phase and enhancement of the escape phase of tumour immunoediting, possibly increased angiogenesis but continued suppression of inflammation. In animal models, having defective VDR signalling, vitamin D3 administration decreased survival and increased metastases. Comparable studies in man are lacking but in advanced disease, a likely marker of defective VDR signalling, studies have shown modest or no improvement in outcome with some evidence of worsening. Work is needed in assessing the integrity of tumour VDR signalling and the safety of vitamin D3 supplementation when defective.
Collapse
Affiliation(s)
| | - James H. Pringle
- Leicester Cancer Research CentreUniversity of LeicesterLeicesterUK
| |
Collapse
|
44
|
Conway K, Tsai YS, Edmiston SN, Parker JS, Parrish EA, Hao H, Kuan PF, Scott GA, Frank JS, Googe P, Ollila DW, Thomas NE. Characterization of the CpG Island Hypermethylated Phenotype Subclass in Primary Melanomas. J Invest Dermatol 2022; 142:1869-1881.e10. [PMID: 34843679 PMCID: PMC9135958 DOI: 10.1016/j.jid.2021.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022]
Abstract
Cutaneous melanoma can be lethal even if detected at an early stage. Epigenetic profiling may facilitate the identification of aggressive primary melanomas with unfavorable outcomes. We performed clustering of whole-genome methylation data to identify subclasses that were then assessed for survival, clinical features, methylation patterns, and biological pathways. Among 89 cutaneous primary invasive melanomas, we identified three methylation subclasses exhibiting low methylation, intermediate methylation, or hypermethylation of CpG islands, known as the CpG island methylator phenotype (CIMP). CIMP melanomas occurred as early as tumor stage 1b and, compared with low-methylation melanomas, were associated with age at diagnosis ≥65 years, lentigo maligna melanoma histologic subtype, presence of ulceration, higher American Joint Committee on Cancer stage and tumor stage, and lower tumor-infiltrating lymphocyte grade (all P < 0.05). Patients with CIMP melanomas had worse melanoma-specific survival (hazard ratio = 11.84; confidence interval = 4.65‒30.20) than those with low-methylation melanomas, adjusted for age, sex, American Joint Committee on Cancer stage, and tumor-infiltrating lymphocyte grade. Genes hypermethylated in CIMP compared with those in low-methylation melanomas included PTEN, VDR, PD-L1, TET2, and gene sets related to development/differentiation, the extracellular matrix, and immunity. CIMP melanomas exhibited hypermethylation of genes important in melanoma progression and tumor immunity, and although present in some early melanomas, CIMP was associated with worse survival independent of known prognostic factors.
Collapse
Affiliation(s)
- Kathleen Conway
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Dermatology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | - Yihsuan S Tsai
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sharon N Edmiston
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Genetics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eloise A Parrish
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Honglin Hao
- Department of Genetics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Pei Fen Kuan
- Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York, USA
| | - Glynis A Scott
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Jill S Frank
- Department of Surgery, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paul Googe
- Department of Dermatology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pathology and Lab Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David W Ollila
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Surgery, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nancy E Thomas
- Department of Dermatology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
45
|
Reichrath J, Biersack F, Wagenpfeil S, Schöpe J, Pföhler C, Saternus R, Vogt T. Low Vitamin D Status Predicts Poor Clinical Outcome in Advanced Melanoma Treated With Immune Checkpoint or BRAF/MEK Inhibitors: A Prospective Non-Interventional Side-by-Side Analysis. Front Oncol 2022; 12:839816. [PMID: 35669434 PMCID: PMC9166268 DOI: 10.3389/fonc.2022.839816] [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: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 12/04/2022] Open
Abstract
In melanoma and other malignancies, low vitamin D status is associated with increased risk and poor prognosis. However, there are limited data of the impact of 25(OH)D serum concentration (s.c.) on clinical outcome in advanced melanoma. We tested the hypothesis that vitamin D status is predictive of efficacy and safety in patients treated for metastasized melanoma with B-rapidly accelerated fibrosarcoma (BRAF), mitogen-activated protein kinase kinase (MEK), cytotoxic T lymphocyte-associated protein-4 (CTLA-4), and/or programmed cell death protein-1 (PD-1) inhibitors. Severe vitamin D deficiency [defined as 25(OH)D s.c. <10 ng/ml] was associated with markedly reduced overall (OS) and progress-free (PFS) survival, with increased tumor load [TL; measured as s.c. of S100 protein or lactate dehydrogenase (LDH)], and with a trend for higher frequency of adverse events (AEs). An increase in average 25(OH)D s.c. of 1 ng/ml was associated with a 3.9% reduced risk for progressive disease [hazard ratio (HR) = 0.961, p = 0.044], with a reduction of LDH s.c. of 3.86 U/l (p = 0.034, indicating a reduction of TL), and with a trend for reduced frequency of AEs (AE ratio -0.005; p = 0.295). Patients with average 25(OH)D s.c. ≥10 ng/ml and BRAF-mutant melanoma showed a trend for a higher frequency of AEs as compared to individuals with BRAF wild-type melanomas. Our data indicate that vitamin D deficiency is associated with poor clinical outcome in patients treated for metastasized melanoma with BRAF/MEK inhibitors or immunotherapy. Although it needs to be proven in future interventional trials whether optimizing serum 25(OH)D improves clinical outcome in these patients, we recommend that 25(OH)D s.c. should be analyzed and vitamin D deficiency treated in all patients with advanced melanoma.
Collapse
Affiliation(s)
- Jörg Reichrath
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany
| | - Florian Biersack
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany
| | - Stefan Wagenpfeil
- Institute of Medical Biometry, Epidemiology and Medical Informatics, Saarland University Medical Center, Homburg, Germany
| | - Jakob Schöpe
- Institute of Medical Biometry, Epidemiology and Medical Informatics, Saarland University Medical Center, Homburg, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany
| | - Roman Saternus
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany
| | - Thomas Vogt
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
46
|
Bernicke B, Engelbogen N, Klein K, Franzenburg J, Borzikowsky C, Peters C, Janssen O, Junker R, Serrano R, Kabelitz D. Analysis of the Seasonal Fluctuation of γδ T Cells and Its Potential Relation with Vitamin D 3. Cells 2022; 11:1460. [PMID: 35563767 PMCID: PMC9099506 DOI: 10.3390/cells11091460] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023] Open
Abstract
In addition to its role in bone metabolism, vitamin D3 exerts immunomodulatory effects and has been proposed to contribute to seasonal variation of immune cells. This might be linked to higher vitamin D3 levels in summer than in winter due to differential sun exposure. γδ T cells comprise a numerically small subset of T cells in the blood, which contribute to anti-infective and antitumor immunity. We studied the seasonal fluctuation of γδ T cells, the possible influence of vitamin D3, and the effect of the active metabolite 1α,25(OH)2D3 on the in vitro activation of human γδ T cells. In a retrospective analysis with 2625 samples of random blood donors, we observed higher proportions of γδ T cells in winter when compared with summer. In a prospective study over one year with a small cohort of healthy adults who did or did not take oral vitamin D3 supplementation, higher proportions of γδ T cells were present in donors without oral vitamin D3 uptake, particularly in spring. However, γδ T cell frequency in blood did not directly correlate with serum levels of 25(OH)D3. The active metabolite 1α,25(OH)2D3 inhibited the in vitro activation of γδ T cells at the level of proliferation, cytotoxicity, and interferon-γ production. Our study reveals novel insights into the seasonal fluctuation of γδ T cells and the immunomodulatory effects of vitamin D3.
Collapse
Affiliation(s)
- Birthe Bernicke
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (B.B.); (K.K.); (C.P.); (O.J.)
| | - Nils Engelbogen
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (N.E.); (J.F.); (R.J.)
| | - Katharina Klein
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (B.B.); (K.K.); (C.P.); (O.J.)
| | - Jeanette Franzenburg
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (N.E.); (J.F.); (R.J.)
| | - Christoph Borzikowsky
- Institute of Bioinformatics and Statistics, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany;
| | - Christian Peters
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (B.B.); (K.K.); (C.P.); (O.J.)
| | - Ottmar Janssen
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (B.B.); (K.K.); (C.P.); (O.J.)
| | - Ralf Junker
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (N.E.); (J.F.); (R.J.)
| | - Ruben Serrano
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (B.B.); (K.K.); (C.P.); (O.J.)
| | - Dieter Kabelitz
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany; (B.B.); (K.K.); (C.P.); (O.J.)
| |
Collapse
|
47
|
Bueno AC, Stecchini MF, Marrero-Gutiérrez J, More CB, Leal LF, Gomes DC, de Lima Neto DF, Brandalise SR, Cardinalli IA, Yunes JA, Junqueira T, Scrideli CA, Molina CAF, Ramalho FS, Tucci S, Coeli-Lacchini FB, Moreira AC, Ramalho L, Vêncio RZN, De Castro M, Antonini SRR. Vitamin D receptor hypermethylation as a biomarker for pediatric adrenocortical tumors. Eur J Endocrinol 2022; 186:573-585. [PMID: 35290212 DOI: 10.1530/eje-21-0879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/14/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Pediatric adrenocortical tumors (pACT) display complex genomic backgrounds, lacking robust prognostic markers and targeted therapeutic options. Vitamin D3 receptor (VDR) promoter hypermethylation and underexpression were reported in adrenocortical carcinomas from adult patients. In this study, we aimed to investigate VDR expression levels and methylation status in pACT and their clinical and prognostic significance. DESIGN Retrospective cross-sectional study enrolling pediatric patients with ACT from two tertiary referral institutions. METHODS We evaluated clinicopathological features, VDR mRNA (qPCR) and protein (immunohistochemistry) expression, and VDR-wide methylation of ACT samples from 108 pediatric patients. Fourteen pediatric and 32 fetal and postnatal normal adrenals were used as controls. RESULTS Unlike in pre- and post-natal normal adrenals, most pACT lacked nuclear VDR expression and had reduced mRNA levels, especially the carcinomas. Unsupervised analysis of VDR methylation data revealed two groups of pACT with distinct disease features and outcomes. Tumors with high VDR methylation presented lower mRNA levels, and the respective patients presented advanced disease and reduced disease-free and overall survival. CONCLUSIONS VDR has a role in normal adrenocortical development and homeostasis, which is impaired during tumorigenesis. VDR hypermethylation and underexpression may be both predictive and prognostic biomarkers for pACT.
Collapse
Affiliation(s)
- Ana Carolina Bueno
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Mônica F Stecchini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Junier Marrero-Gutiérrez
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Candy Bellido More
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Leticia Ferro Leal
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Daniel Ferreira de Lima Neto
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | | | - José Andres Yunes
- Boldrini Children's Center, State University of Campinas, Campinas, Sao Paulo, Brazil
| | - Thais Junqueira
- Boldrini Children's Center, State University of Campinas, Campinas, Sao Paulo, Brazil
| | - Carlos Alberto Scrideli
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Carlos Augusto Fernandes Molina
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Silva Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Silvio Tucci
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Ayrton Custodio Moreira
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Leandra Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Ricardo Zorzetto Nicoliello Vêncio
- Department of Computation and Mathematics, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Margaret De Castro
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Sonir Roberto R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
48
|
Rapid Nontranscriptional Effects of Calcifediol and Calcitriol. Nutrients 2022; 14:nu14061291. [PMID: 35334948 PMCID: PMC8951353 DOI: 10.3390/nu14061291] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 01/28/2023] Open
Abstract
Classically, a secosteroid hormone, vitamin D, has been implicated in calcium and phosphate homeostasis and has been associated with the pathogenesis of rickets and osteomalacia in patients with severe nutritional vitamin D deficiency. The spectrum of known vitamin D-mediated effects has been expanded in recent years. However, the mechanisms of how exactly this hormone elicits its biological function are still not fully understood. The interaction of this metabolite with the vitamin D receptor (VDR) and, subsequently, with the vitamin D-responsive element in the region of specific target genes leading to the transcription of genes whose protein products are involved in the traditional function of calcitriol (known as genomic actions). Moreover, in addition to these transcription-dependent mechanisms, it has been recognized that the biologically active form of vitamin D3, as well as its immediate precursor metabolite, calcifediol, initiate rapid, non-genomic actions through the membrane receptors that are bound as described for other steroid hormones. So far, among the best candidates responsible for mediating rapid membrane response to vitamin D metabolites are membrane-associated VDR (VDRm) and protein disulfide isomerase family A member 3 (Pdia3). The purpose of this paper is to provide an overview of the rapid, non-genomic effects of calcifediol and calcitriol, whose elucidation could improve the understanding of the vitamin D3 endocrine system. This will contribute to a better recognition of the physiological acute functions of vitamin D3, and it could lead to the identification of novel therapeutic targets able to modulate these actions.
Collapse
|
49
|
Ye Z, Ren C, Wang Q, Wang Y. A novel role of TCAIM: suppressing renal carcinoma growth and enhancing its sensitivity to sunitinib. FEBS J 2022; 289:5259-5278. [PMID: 35238167 DOI: 10.1111/febs.16417] [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: 09/26/2021] [Revised: 01/18/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
Renal carcinoma is a common malignancy for which the underlying molecular mechanisms warrant further investigation. T cell activation inhibitor mitochondrial (TCAIM), a mitochondrial protein, was found to be expressed to a low extent in renal carcinoma specimens. However, its role in carcinomas remains unclear. In the present study, the role of TCAIM in renal carcinoma was explored through loss-of-function and gain-of-function experiments. Here, we showed that TCAIM delayed the growth of renal carcinoma cells both in vitro and in vivo by modulating their cell cycle progress. Additionally, TCAIM also enhanced their sensitivity to sunitinib by aggravating apoptosis. Furthermore, TCAIM also decreased the adhesion and migration of renal carcinoma cells. Moreover, the transcription of TCAIM was regulated by vitamin D receptor, which acts as a transcriptional factor. To identify the pathways regulated by TCAIM, 425 unique proteins bound to TCAIM were pulled down through co-immunoprecipitation and analyzed by mass spectrometry followed by Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes analysis. In summary, the present study reveals a tumor-suppressor role of TCAIM in renal carcinoma cells, as well as its upstream regulation and downstream potential mechanisms. Our study provides theoretical bases and novel insights with respect to the development of new therapeutic strategies for the treatment of renal carcinoma.
Collapse
Affiliation(s)
- Zhuo Ye
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, China
| | - Chuanchuan Ren
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, China
| | - Qingwei Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, China
| | - Yan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, China
| |
Collapse
|
50
|
Davies J, Muralidhar S, Randerson-Moor J, Harland M, O'Shea S, Diaz J, Walker C, Nsengimana J, Laye J, Mell T, Chan M, Appleton L, Birkeälv S, Adams DJ, Cook GP, Ball G, Bishop DT, Newton-Bishop JA. Ulcerated melanoma: Systems biology evidence of inflammatory imbalance towards pro-tumourigenicity. Pigment Cell Melanoma Res 2022; 35:252-267. [PMID: 34826184 DOI: 10.1111/pcmr.13023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/03/2021] [Accepted: 11/23/2021] [Indexed: 01/05/2023]
Abstract
Microscopic ulceration is an independent predictor of melanoma death. Here, we used systems biology to query the role of host and tumour-specific processes in defining the phenotype. Albumin level as a measure of systemic inflammation was predictive of fewer tumour-infiltrating lymphocytes and poorer survival in the Leeds Melanoma Cohort. Ulcerated melanomas were thicker and more mitotically active (with corresponding transcriptomic upregulated cell cycle pathways). Sequencing identified tumoural p53 and APC mutations, and TUBB2B amplification as associated with the phenotype. Ulcerated tumours had perturbed expression of cytokine genes, consistent with protumourigenic inflammation and histological and transcriptomic evidence for reduced adaptive immune cell infiltration. Pathway/network analysis of multiomic data using neural networks highlighted a role for the β-catenin pathway in the ulceration, linking genomic changes in the tumour to immunosuppression and cell proliferation. In summary, the data suggest that ulceration is in part associated with genomic changes but that host factors also predict melanoma death with evidence of reduced immune responses to the tumour.
Collapse
Affiliation(s)
- John Davies
- Leeds Institute of Data Analytics, University of Leeds, Leeds, UK
| | - Sathya Muralidhar
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, UK
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | - Mark Harland
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Sally O'Shea
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Dermatology Department, South Infirmary-Victoria University Hospital Cork and University College Cork, Cork, Ireland
| | - Joey Diaz
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Christy Walker
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Jérémie Nsengimana
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Population Health Sciences Institute, University of Newcastle, Newcastle upon Tyne, UK
| | - Jon Laye
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Tracey Mell
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - May Chan
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Lizzie Appleton
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Sofia Birkeälv
- Experimental Cancer Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Graham P Cook
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Graham Ball
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - David T Bishop
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | |
Collapse
|