1
|
Chou CL, Lin CW, Li WS, Chen TJ, Lee SW, Tian YF, Kuo YH, Tsai HH, Wu LC, Yeh CF, Shiue YL, Lai HY, Yang CC. High Intelectin-1 Expression Associated with Aggressive Tumor Behavior and Worse Survival in Rectal Cancer. Onco Targets Ther 2025; 18:15-26. [PMID: 39830923 PMCID: PMC11740546 DOI: 10.2147/ott.s488608] [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: 07/25/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Background Multimodal treatment involving preoperative chemoradiotherapy (CRT) followed by surgery is the current standard of care for rectal cancer. Despite advancements, the risk of recurrence, metastasis, and decreased survival remains high. This study aims to evaluate potential biomarkers to stratify prognosis in patients with rectal cancer undergoing preoperative CRT and surgery. Methods Through data mining of receptor-binding pathways in a published transcriptome for rectal cancer cases, ITLN1 was identified as the most relevant gene associated with poor response to chemoradiation (GO:0005102). Rectal cancer specimens (n = 343) collected between 1998 and 2017 were analyzed for ITLN1 expression using immunohistochemistry. The association between ITLN1 protein expression and clinicopathological features was assessed using Pearson's chi-square test. Survival outcomes based on ITLN1 expression were evaluated using the Kaplan-Meier method and compared with Log rank tests. Results ITLN1 immunoreactivity was significantly elevated in rectal tumor tissues. High ITLN1 expression was strongly associated with adverse clinicopathological features, including advanced post-treatment tumor status (T3-4; p = 0.001), post-treatment nodal status (N1-2; p < 0.001), vascular invasion (p = 0.017), perineural invasion (p = 0.001), and a lower degree of tumor regression (p = 0.009). Uni- and multivariable analyses revealed that high ITLN1 expression correlated with poorer disease-specific survival, local recurrence-free survival, and distant metastasis-free survival compared to low ITLN1 expression. Conclusion Elevated ITLN1 expression is significantly associated with aggressive tumor behavior and unfavorable survival outcomes in rectal cancer. These findings highlight ITLN1 as a potential prognostic biomarker and provide a foundation for future research into its role in rectal cancer progression and treatment response.
Collapse
Affiliation(s)
- Chia-Lin Chou
- Division of Colon & Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Shan Li
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Tzu-Ju Chen
- Department of Pathology, Tainan Municipal Hospital, Tainan, Taiwan
| | - Sung-Wei Lee
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Taiwan
| | - Yu-Feng Tian
- Division of Colon & Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Yu-Hsuan Kuo
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
| | - Hsin-Hwa Tsai
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Trans-Omic Laboratory for Precision Medicine, Precision Medicine Center, Chi Mei Medical Center, Tainan, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Li-Ching Wu
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Trans-Omic Laboratory for Precision Medicine, Precision Medicine Center, Chi Mei Medical Center, Tainan, Taiwan
| | - Cheng-Fa Yeh
- Division of General Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Environment Engineering and Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yow-Ling Shiue
- Institute of Precision Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hong-Yue Lai
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Trans-Omic Laboratory for Precision Medicine, Precision Medicine Center, Chi Mei Medical Center, Tainan, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Chieh Yang
- Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Ye H, Zhang R, Zhang C, Xia Y, Jin L. Advances in hyaluronic acid: Bioactivity, complexed biomaterials and biological application: A review. Asian J Surg 2024:S1015-9584(24)01841-4. [PMID: 39217010 DOI: 10.1016/j.asjsur.2024.08.100] [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: 04/01/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Hyaluronic acid (HA) is a natural glycosaminoglycan found in the human body, particularly in the extracellular matrix of body fluids and tissues. It plays a critical role in cellular processes of living organisms by maintaining tissue hydration, cell proliferation, differentiation, and inflammatory response. HA exhibits significant biological activity in skin care, aesthetic anti-aging, medical orthopedic repair, gynecological cancer monitoring, and other pathological conditions. Due to its exceptional biocompatibility, biodegradability, lack of toxicity, non-immunogenicity, and its capacity to bond with other substances, various HA-based biomedical products like hydrogels, microneedles, and microspheres have been developed. These innovations have also been applied in various medical and health fields, such as bone and tissue regeneration, gels for medical aesthetic fillers, and gynecology-related cancer treatment, utilizing the HA drug delivery pathway. The interest in HA and its products is increasing due to their biological functions. Therefore, this review aimed to summarize the biological properties of HA and to focus on its applications in the bone tissue engineering and healthcare, for HA has some practical applications of HA-based complexes in biomedical materials, tissue repair, medical aesthetics, and gynecology. Through this review, we seek to offer theoretical research assistance for the development of HA-based bioproducts in the healthcare domain and provide innovative insights for human health.
Collapse
Affiliation(s)
- Huijun Ye
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318 Chaowang Road, Hangzhou, 310005, Zhejiang, China
| | - Ruijuan Zhang
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China
| | - Chunye Zhang
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China
| | - Yujie Xia
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China.
| | - Lihua Jin
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318 Chaowang Road, Hangzhou, 310005, Zhejiang, China.
| |
Collapse
|
3
|
Wu H, Fu M, Wu M, Cao Z, Zhang Q, Liu Z. Emerging mechanisms and promising approaches in pancreatic cancer metabolism. Cell Death Dis 2024; 15:553. [PMID: 39090116 PMCID: PMC11294586 DOI: 10.1038/s41419-024-06930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Pancreatic cancer is an aggressive cancer with a poor prognosis. Metabolic abnormalities are one of the hallmarks of pancreatic cancer, and pancreatic cancer cells can adapt to biosynthesis, energy intake, and redox needs through metabolic reprogramming to tolerate nutrient deficiency and hypoxic microenvironments. Pancreatic cancer cells can use glucose, amino acids, and lipids as energy to maintain malignant growth. Moreover, they also metabolically interact with cells in the tumour microenvironment to change cell fate, promote tumour progression, and even affect immune responses. Importantly, metabolic changes at the body level deserve more attention. Basic research and clinical trials based on targeted metabolic therapy or in combination with other treatments are in full swing. A more comprehensive and in-depth understanding of the metabolic regulation of pancreatic cancer cells will not only enrich the understanding of the mechanisms of disease progression but also provide inspiration for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengdi Fu
- Department of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhen Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiyao Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
4
|
Zhu Y, Liang X, Zhang G, Li F, Xu J, Ma R, Chen X, Ma M, Wang Y, Chen C, Tang H, Li L, Li Z. Microbiota and metabolite alterations in pancreatic head and body/tail cancer patients. Cancer Sci 2024; 115:2738-2750. [PMID: 38888048 PMCID: PMC11309928 DOI: 10.1111/cas.16238] [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: 03/15/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Pancreatic head cancer (PHC) and pancreatic body/tail cancer (PBTC) have distinct clinical and biological behaviors. The microbial and metabolic differences in PHC and PBTC have not been studied. The pancreatic microbiota and metabolome of 15 PHC and 8 PBTC tissues and their matched nontumor tissues were characterized using 16S rRNA amplicon sequencing and untargeted metabolomics. At the genus level, Bradyrhizobium was increased while Corynebacterium and Ruminococcus were decreased in the PHC tissues (Head T) compared with the matched nontumor tissues (Head N) significantly. Shuttleworthia, Bacillus, and Bifidobacterium were significantly decreased in the PBTC tissues (Body/Tail T) compared with the matched nontumor tissues (Body/Tail N). Significantly, Ileibacterium was increased whereas Pseudoxanthomonas was decreased in Head T and Body/Tail T, and Lactobacillus was increased in Head T but decreased in Body/Tail T. A total of 102 discriminative metabolites were identified between Head T and Head N, which were scattered through linoleic acid metabolism and purine metabolism pathways. However, there were only four discriminative metabolites between Body/Tail T and Body/Tail N, which were related to glycerophospholipid metabolism and autophagy pathways. The differential metabolites in PHC and PBTC were commonly enriched in alpha-linolenic acid metabolism and choline metabolism in cancer pathways. Eubacterium decreased in Head T was positively correlated with decreased linoleic acid while negatively correlated with increased arachidyl carnitine and stearoylcarnitine. Bacillus decreased in Body/Tail T was negatively correlated with increased L-carnitine. These microbiota and metabolites deserve further investigations to reveal their roles in the pathogenesis of PHC and PBTC, providing clues for future treatments.
Collapse
|
5
|
Kim K. The Role of Endocrine Disruption Chemical-Regulated Aryl Hydrocarbon Receptor Activity in the Pathogenesis of Pancreatic Diseases and Cancer. Int J Mol Sci 2024; 25:3818. [PMID: 38612627 PMCID: PMC11012155 DOI: 10.3390/ijms25073818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The aryl hydrocarbon receptor (AHR) serves as a ligand-activated transcription factor crucial for regulating fundamental cellular and molecular processes, such as xenobiotic metabolism, immune responses, and cancer development. Notably, a spectrum of endocrine-disrupting chemicals (EDCs) act as agonists or antagonists of AHR, leading to the dysregulation of pivotal cellular and molecular processes and endocrine system disruption. Accumulating evidence suggests a correlation between EDC exposure and the onset of diverse pancreatic diseases, including diabetes, pancreatitis, and pancreatic cancer. Despite this association, the mechanistic role of AHR as a linchpin molecule in EDC exposure-related pathogenesis of pancreatic diseases and cancer remains unexplored. This review comprehensively examines the involvement of AHR in EDC exposure-mediated regulation of pancreatic pathogenesis, emphasizing AHR as a potential therapeutic target for the pathogenesis of pancreatic diseases and cancer.
Collapse
Affiliation(s)
- Kyounghyun Kim
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas Medical Sciences, Little Rock, AR 72225, USA
| |
Collapse
|
6
|
Xiao C, Comer L, Pan X, Everaert N, Schroyen M, Song Z. Zinc glycinate alleviates LPS-induced inflammation and intestinal barrier disruption in chicken embryos by regulating zinc homeostasis and TLR4/NF-κB pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116111. [PMID: 38350216 DOI: 10.1016/j.ecoenv.2024.116111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/26/2024] [Accepted: 02/11/2024] [Indexed: 02/15/2024]
Abstract
The effect of an immune challenge induced by a lipopolysaccharide (LPS) exposure on systemic zinc homeostasis and the modulation of zinc glycinate (Zn-Gly) was investigated using a chicken embryo model. 160 Arbor Acres broiler fertilized eggs were randomly divided into 4 groups: CON (control group, injected with saline), LPS (LPS group, injected with 32 µg of LPS saline solution), Zn-Gly (zinc glycinate group, injected with 80 µg of zinc glycinate saline solution) and Zn-Gly+LPS (zinc glycinate and LPS group, injected with the same content of zinc glycinate and LPS saline solution). Each treatment consisted of eight replicates of five eggs each. An in ovo feeding procedure was performed at 17.5 embryonic day and samples were collected after 12 hours. The results showed that Zn-Gly attenuated the effects of LPS challenge-induced upregulation of pro-inflammatory factor interleukin 1β (IL-1β) level (P =0.003). The LPS challenge mediated zinc transporter proteins and metallothionein (MT) to regulate systemic zinc homeostasis, with increased expression of the jejunum zinc export gene zinc transporter protein 1 (ZnT-1) and elevated expression of the import genes divalent metal transporter 1 (DMT1), Zrt- and Irt-like protein 3 (Zip3), Zip8 and Zip14 (P < 0.05). A similar trend could be observed for the zinc transporter genes in the liver, which for ZnT-1 mitigated by Zn-Gly supplementation (P =0.01). Liver MT gene expression was downregulated in response to the LPS challenge (P =0.004). These alterations caused by LPS resulted in decreased serum and liver zinc levels and increased small intestinal, muscle and tibial zinc levels. Zn-Gly reversed the elevated expression of the liver zinc finger protein A20 induced by the LPS challenge (P =0.025), while Zn-Gly reduced the gene expression of the pro-inflammatory factors IL-1β and IL-6, decreased toll-like receptor 4 (TLR4) and nuclear factor kappa-B p65 (NF-κB p65) (P < 0.05). Zn-Gly also alleviated the LPS-induced downregulation of the intestinal barrier gene Claudin-1. Thus, LPS exposure prompted the mobilization of zinc transporter proteins and MT to perform the remodeling of systemic zinc homeostasis, Zn-Gly participated in the regulation of zinc homeostasis and inhibited the production of pro-inflammatory factors through the TLR4/NF-κB pathway, attenuating the inflammatory response and intestinal barrier damage caused by an immune challenge.
Collapse
Affiliation(s)
- Chuanpi Xiao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Luke Comer
- Nutrition and Animal Microbiota Ecosystems lab, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Nadia Everaert
- Nutrition and Animal Microbiota Ecosystems lab, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
7
|
Shapera E, Ross S, Sucandy I, Touadi M, Pattilachan T, Christodoulou M, Rosemurgy A. The weight of BMI in impacting postoperative and oncologic outcomes in pancreaticoduodenectomy is attenuated by a robotic approach. J Robot Surg 2024; 18:77. [PMID: 38353858 DOI: 10.1007/s11701-024-01833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/14/2024] [Indexed: 02/16/2024]
Abstract
This study was undertaken to observe the effect of body mass index (BMI) on perioperative outcomes and survival when comparing robotic vs 'open' pancreaticoduodenectomy. With IRB approval, we prospectively followed 505 consecutive patients who underwent either robotic or 'open' pancreaticoduodenectomy from 2012 to 2021. For illustrative purposes, patients were separated based on the Center for Disease Control and Prevention BMI table but regression analysis was utilized to identify significant relationships involving BMI. Data are presented as median (mean ± SD). Significance was determined at p ≤ 0.05. 205 and 300 patients underwent 'open' and robotic pancreaticoduodenectomy, respectively. Neither sex nor age correlated with BMI in patients undergoing 'open' nor robotic operation. Operative duration correlated with increasing BMI in each operational approach, which was statistically significant for those receiving the 'open' operation (p = 0.02). There were statistically significantly fewer lymph nodes harvested with rising BMI in patients that had an 'open' operation (p = 0.01), but no such difference was found in patients undergoing the robotic approach. Length of stay (LOS) and in-hospital mortality were statistically significantly associated with rising BMI when an 'open' operation was undertaken (p = 0.02 and p = 0.0002, respectively) but not when the robotic platform was utilized. Patients with higher BMI had significantly longer operative duration, smaller lymph node harvest, greater LOS, and increased in-hospital mortality rate when undergoing 'open' pancreaticoduodenectomy, but not robotic pancreaticoduodenectomy. Thus, the robotic platform may attenuate the increased technical and oncologic difficulties associated with a greater BMI in patients undergoing pancreaticoduodenectomy.
Collapse
Affiliation(s)
- Emanuel Shapera
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Sharona Ross
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA.
| | - Iswanto Sucandy
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Melissa Touadi
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Tara Pattilachan
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Maria Christodoulou
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Alexander Rosemurgy
- Digestive Health Institute, AdventHealth, 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| |
Collapse
|
8
|
Kordshouli SO, Tahmasebi A, Moghadam A, Ramezani A, Niazi A. A comprehensive meta-analysis of transcriptome data to identify signature genes associated with pancreatic ductal adenocarcinoma. PLoS One 2024; 19:e0289561. [PMID: 38324544 PMCID: PMC10849254 DOI: 10.1371/journal.pone.0289561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 07/20/2023] [Indexed: 02/09/2024] Open
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) has a five-year survival rate of less than 5%. Absence of symptoms at primary tumor stages, as well as high aggressiveness of the tumor can lead to high mortality in cancer patients. Most patients are recognized at the advanced or metastatic stage without surgical symptom, because of the lack of reliable early diagnostic biomarkers. The objective of this work was to identify potential cancer biomarkers by integrating transcriptome data. METHODS Several transcriptomic datasets comprising of 11 microarrays were retrieved from the GEO database. After pre-processing, a meta-analysis was applied to identify differentially expressed genes (DEGs) between tumor and nontumor samples for datasets. Next, co-expression analysis, functional enrichment and survival analyses were used to determine the functional properties of DEGs and identify potential prognostic biomarkers. In addition, some regulatory factors involved in PDAC including transcription factors (TFs), protein kinases (PKs), and miRNAs were identified. RESULTS After applying meta-analysis, 1074 DEGs including 539 down- and 535 up-regulated genes were identified. Pathway enrichment analyzes using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that DEGs were significantly enriched in the HIF-1 signaling pathway and focal adhesion. The results also showed that some of the DEGs were assigned to TFs that belonged to 23 conserved families. Sixty-four PKs were identified among the DEGs that showed the CAMK family was the most abundant group. Moreover, investigation of corresponding upstream regions of DEGs identified 11 conserved sequence motifs. Furthermore, weighted gene co-expression network analysis (WGCNA) identified 8 modules, more of them were significantly enriched in Ras signaling, p53 signaling, MAPK signaling pathways. In addition, several hubs in modules were identified, including EMP1, EVL, ELP5, DEF8, MTERF4, GLUP1, CAPN1, IGF1R, HSD17B14, TOM1L2 and RAB11FIP3. According to survival analysis, it was identified that the expression levels of two genes, EMP1 and RAB11FIP3 are related to prognosis. CONCLUSION We identified several genes critical for PDAC based on meta-analysis and system biology approach. These genes may serve as potential targets for the treatment and prognosis of PDAC.
Collapse
Affiliation(s)
| | | | - Ali Moghadam
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
| | - Amin Ramezani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Niazi
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
| |
Collapse
|
9
|
Tanaka S, Tsujimae M, Masuda A, Inoue J, Inomata N, Uemura H, Kohashi S, Nagao K, Masuda S, Abe S, Gonda M, Yamakawa K, Ashina S, Nakano R, Tanaka T, Yamada Y, Sakai A, Kobayashi T, Shiomi H, Fujita K, Anami T, Fujita T, Watanabe A, Kodama Y. Metabolic Syndrome Accelerates the Age-Related Increase of Intraductal Papillary Mucinous Neoplasm of the Pancreas. Pancreas 2024; 53:e9-e15. [PMID: 37890158 DOI: 10.1097/mpa.0000000000002267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
OBJECTIVES Aging is associated with a high prevalence of pancreatic cysts and intraductal papillary mucinous neoplasms (IPMNs). Metabolic syndrome (MS) may increase the risk of neoplasms, including those that develop in the pancreas. However, the influence of factors associated with MS on the development of IPMN remains unclear. METHODS A total of 9363 patients who underwent abdominal ultrasound examinations between April 2012 and May 2013 were included in this study. Multivariate logistic regression analysis was performed to identify factors associated with the presence of IPMN by age. RESULTS Pancreatic cysts were detected in 198 of 9363 patients, of whom 129 were found to have IPMNs. The presence of IPMN significantly correlated with age (10-year increments; odds ratio, 2.73; 95% CI, 2.28-3.29; P < 0.001). High body mass index, history of smoking, hyperlipidemia, hypertension, and MS were associated with a higher prevalence of IPMN with advancing age. In multivariate analysis, the presence of IPMN was more frequent in elderly patients with MS (odds ratio, 3.14; 95% CI, 3.14-6.72; P = 0.003). CONCLUSIONS The present study suggests that the incidence of IPMN increases with age and is accelerated in the presence of MS.
Collapse
Affiliation(s)
- Shunta Tanaka
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Masahiro Tsujimae
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Atsuhiro Masuda
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Jun Inoue
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Noriko Inomata
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Hisahiro Uemura
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Shinya Kohashi
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Kae Nagao
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Shigeto Masuda
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Shohei Abe
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Masanori Gonda
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Kohei Yamakawa
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Shigeto Ashina
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Ryota Nakano
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Takeshi Tanaka
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Yasutaka Yamada
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Arata Sakai
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Takashi Kobayashi
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Hideyuki Shiomi
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| | - Koichi Fujita
- Department of Gastroenterology, Yodogawa Christian Hospital, Osaka, Japan
| | - Takahiro Anami
- Department of Gastroenterology, Yodogawa Christian Hospital, Osaka, Japan
| | - Tsuyoshi Fujita
- Department of Gastroenterology, Yodogawa Christian Hospital, Osaka, Japan
| | - Akihiko Watanabe
- Department of Gastroenterology, Yodogawa Christian Hospital, Osaka, Japan
| | - Yuzo Kodama
- From the Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo
| |
Collapse
|
10
|
Olowosoke CB, Gbemisola O, Alaba AA, Adepoju OH, Okorie B, Odjegba PI, Ogunsanmi AO, Oke GA, Akinlolu O, Olubena TL, Bello RO, Adegboyega BB. Multi-regulator of EZH2-PPARs Therapeutic Targets: A Hallmark for Prospective Restoration of Pancreatic Insulin Production and Cancer Dysregulation. Appl Biochem Biotechnol 2023; 195:7520-7552. [PMID: 37010741 DOI: 10.1007/s12010-023-04433-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/04/2023]
Abstract
The unexpected rise in cancer and diabetes statistics has been a significant global threat, inciting ongoing research into various biomarkers that can act as innovative therapeutic targets for their management. The recent discovery of how EZH2-PPARs' regulatory function affects the metabolic and signalling pathways contributing to this disease has posed a significant breakthrough, with the synergistic combination of inhibitors like GSK-126 and bezafibrate for treating these diseases. Nonetheless, no findings on other protein biomarkers involved in the associated side effects have been reported. As a result of this virtual study, we identified the gene-disease association, protein interaction networks between EZH2-PPARs and other protein biomarkers regulating pancreatic cancer and diabetes pathology, ADME/Toxicity profiling, docking simulation and density functional theory of some natural products. The results indicated a correlation between obesity and hypertensive disease for the investigated biomarkers. At the same time, the predicted protein network validates the link to cancer and diabetes, and nine natural products were screened to have versatile binding capacity against the targets. Among all natural products, phytocassane A outperforms the standard drugs' (GSK-126 and bezafibrate) in silico validation for drug-likeness profiles. Hence, these natural products were conclusively proposed for additional experimental screening to complement the results on their utility in drug development for diabetes and cancer therapy against the EZH2-PPARs' new target.
Collapse
Affiliation(s)
- Christopher Busayo Olowosoke
- Department of Biotechnology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria.
- Department of Biotechnology (College of Natural and Applied Sciences), Chrisland University, Km 5, Ajebo Road, Abeokuta, Ogun State, Nigeria.
- Research Development Unit, Institute of Bioinformatics and Molecular Therapeutics, Osogbo, Osun State, Nigeria.
| | - Otitoola Gbemisola
- Department of Human Nutrition and Dietetics (College of Medicine), University of Ibadan, P.M.B 5017, Ibadan, Oyo State, Nigeria
| | - Adebola Abosede Alaba
- Department of Microbiology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
| | - Oluwadamilola Hope Adepoju
- Department of Biotechnology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
- Department of Biochemistry (College of Biosciences), Federal University of Agriculture, Abeokuta, P.M.B 2240, Abeokuta, Ogun State, Nigeria
| | - Benson Okorie
- Department of Biotechnology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
| | - Peace Ifeoma Odjegba
- Department of Microbiology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
| | - Ayomide Oluwaseyi Ogunsanmi
- Department of Biochemistry (Faculty of Pure and Applied Science), Kwara State University, P.M.B. 1530, Ilorin, Kwara State, Nigeria
| | - Grace Ayomide Oke
- Department of Food Science and Technology (School of Agriculture and Agricultural Technology), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
| | - Oluwatoyin Akinlolu
- Department of Microbiology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
| | - Tomiwa Lois Olubena
- Department of Biotechnology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
| | - Ridwan Opeyemi Bello
- Department of Biotechnology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
- Computer-Aided Therapeutic Discovery and Design Group, FUTA, Akure, Ondo State, Nigeria
| | - Benjamin Babatunde Adegboyega
- Department of Biotechnology (School of Life Sciences), Federal University of Technology, Akure, P.M.B 704, Akure, Ondo State, Nigeria
- Department of Biotechnology (College of Natural and Applied Sciences), Chrisland University, Km 5, Ajebo Road, Abeokuta, Ogun State, Nigeria
| |
Collapse
|
11
|
Srilatha M, Malla R, Adem MP, Foote JB, Nagaraju GP. Obesity associated pancreatic ductal adenocarcinoma: Therapeutic challenges. Semin Cancer Biol 2023; 97:12-20. [PMID: 37926347 DOI: 10.1016/j.semcancer.2023.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
Obesity is a prominent health issue worldwide and directly impacts pancreatic health, with obese individuals exhibiting a significant risk for increasing pancreatic ductal adenocarcinoma (PDAC). Several factors potentially explain the increased risk for the development of PDAC, including obesity-induced chronic inflammation within and outside of the pancreas, development of insulin resistance and metabolic dysfunction, promotion of immune suppression within the pancreas during inflammation, pre- and malignant stages, variations in hormones levels (adiponectin, ghrelin, and leptin) produced from the adipose tissue, and acquisition of somatic mutations in tumor once- and suppressor proteins critical for pancreatic tumorigenesis. In this manuscript, we will explore the broad impact of these obesity-induced risk factors on the development and progression of PDAC, focusing on changes within the tumor microenvironment (TME) as they pertain to prevention, current therapeutic strategies, and future directions for targeting obesity management as they relate to the prevention of pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | - Ramarao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India
| | - Megha Priya Adem
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women's University), Tirupati, Andhra Pradesh 517502, India
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | |
Collapse
|
12
|
Hamdy Gad E. Introductory Chapter: Pancreatic Cancer – How to Prevent, Screen, and Detect? PANCREATIC CANCER- UPDATES IN PATHOGENESIS, DIAGNOSIS AND THERAPIES 2023. [DOI: 10.5772/intechopen.111726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
|
13
|
Goswami S, Zhang Q, Celik CE, Reich EM, Yilmaz ÖH. Dietary fat and lipid metabolism in the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2023; 1878:188984. [PMID: 37722512 PMCID: PMC10937091 DOI: 10.1016/j.bbcan.2023.188984] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/20/2023]
Abstract
Metabolic reprogramming has been considered a core hallmark of cancer, in which excessive accumulation of lipids promote cancer initiation, progression and metastasis. Lipid metabolism often includes the digestion and absorption of dietary fat, and the ways in which cancer cells utilize lipids are often influenced by the complex interactions within the tumor microenvironment. Among multiple cancer risk factors, obesity has a positive association with multiple cancer types, while diets like calorie restriction and fasting improve health and delay cancer. Impact of these diets on tumorigenesis or cancer prevention are generally studied on cancer cells, despite heterogeneity of the tumor microenvironment. Cancer cells regularly interact with these heterogeneous microenvironmental components, including immune and stromal cells, to promote cancer progression and metastasis, and there is an intricate metabolic crosstalk between these compartments. Here, we focus on discussing fat metabolism and response to dietary fat in the tumor microenvironment, focusing on both immune and stromal components and shedding light on therapeutic strategies surrounding lipid metabolic and signaling pathways.
Collapse
Affiliation(s)
- Swagata Goswami
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Qiming Zhang
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Cigdem Elif Celik
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Hacettepe Univ, Canc Inst, Department Basic Oncol, Ankara TR-06100, Turkiye
| | - Ethan M Reich
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ömer H Yilmaz
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Massachusetts General Hospital and Beth Israel Deaconness Medical Center and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
14
|
Pandey P, Khan F. Gut microbiome in cancer immunotherapy: Current trends, translational challenges and future possibilities. Biochim Biophys Acta Gen Subj 2023; 1867:130401. [PMID: 37307905 DOI: 10.1016/j.bbagen.2023.130401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Gut microbiota is regarded as a crucial regulator of the immune system. Healthy gut microbiota plays a specialized role in host xenobiotics, nutrition, drug metabolism, regulation of the structural integrity of the gut mucosal barrier, defense against infections, and immunomodulation. It is now understood that any imbalance in gut microbiota composition from that present in a healthy state is linked to genetic susceptibility to a number of metabolic disorders, including diabetes, autoimmunity, and cancer. Recent research has suggested that immunotherapy can treat many different cancer types with fewer side effects and better ability to eradicate tumors than conventional chemotherapy or radiotherapy. However, a significant number of patients eventually develop immunotherapy resistance. A strong correlation was observed between the composition of the gut microbiome and the effectiveness of treatment by examining the variations between populations that responded to immunotherapy and those that did not. Therefore, we suggest that modulating the microbiome could be a potential adjuvant therapy for cancer immunotherapy and that the architecture of the gut microbiota may be helpful in explaining the variation in treatment response. Herein, we focus on recent research on the interactions among the gut microbiome, host immunity, and cancer immunotherapy. In addition, we highlighted the clinical manifestations, future opportunities, and limitations of microbiome manipulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India.
| |
Collapse
|
15
|
Hamdy Gad E. Pancreatic Cancer: Updates in Pathogenesis and Therapies. PANCREATIC CANCER- UPDATES IN PATHOGENESIS, DIAGNOSIS AND THERAPIES [WORKING TITLE] 2023. [DOI: 10.5772/intechopen.112675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Despite the progress in pancreatic cancer (PC) chemo/radiotherapies, immunotherapies, and novel targeted therapies and the improvement in its peri-operative management policies, it still has a dismal catastrophic prognosis due to delayed detection, early neural and vascular invasions, early micro-metastatic spread, tumour heterogeneities, drug resistance either intrinsic or acquired, unique desmoplastic stroma, and tumour microenvironment (TME). Understanding tumour pathogenesis at the detailed genetic/epigenetic/metabolic/molecular levels as well as studying the tumour risk factors and its known precancerous lesions aggressively is required for getting a more successful therapy for this challenging tumour. For a better outcome of this catastrophic tumour, it should be diagnosed early and treated through multidisciplinary teams of surgeons, gastroenterologists/interventional upper endoscopists, medical/radiation oncologists, diagnostic/intervention radiologists, and pathologists at high-volume centres. Moreover, surgical resection with a negative margin (R0) is the only cure for it. In this chapter; we discuss the recently updated knowledge of PC pathogenesis, risk factors, and precancerous lesions as well as its different management tools (i.e. surgery, chemo/radiotherapies, immunotherapies, novel targeted therapies, local ablative therapies, etc.).
Collapse
|
16
|
Cuttica CM, Briata IM, DeCensi A. Novel Treatments for Obesity: Implications for Cancer Prevention and Treatment. Nutrients 2023; 15:3737. [PMID: 37686769 PMCID: PMC10490004 DOI: 10.3390/nu15173737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
It is now established that obesity is related to a higher incidence of cancer during a lifespan. The effective treatment of obesity opens up new perspectives in the treatment of a relevant modifiable cancer risk factor. The present narrative review summarizes the correlations between weight loss in obesity and cancer. The current knowledge between obesity treatment and cancer was explored, highlighting the greatest potential for its use in the treatment of cancer in the clinical setting. Evidence for the effects of obesity therapy on proliferation, apoptosis, and response to chemotherapy is summarized. While more studies, including large, long-term clinical trials, are needed to adequately evaluate the relationship and durability between anti-obesity treatment and cancer, collaboration between oncologists and obesity treatment experts is increasingly important.
Collapse
Affiliation(s)
| | - Irene Maria Briata
- Division of Medical Oncology, E.O. Ospedali Galliera, 16128 Genoa, Italy; (I.M.B.); (A.D.)
| | - Andrea DeCensi
- Division of Medical Oncology, E.O. Ospedali Galliera, 16128 Genoa, Italy; (I.M.B.); (A.D.)
- Wolfson Institute of Population Health, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
| |
Collapse
|
17
|
Giansante V, Stati G, Sancilio S, Guerra E, Alberti S, Di Pietro R. The Dual Role of Necroptosis in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2023; 24:12633. [PMID: 37628814 PMCID: PMC10454309 DOI: 10.3390/ijms241612633] [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: 07/09/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer-related death. PC incidence has continued to increase by about 1% each year in both men and women. Although the 5-year relative survival rate of PC has increased from 3% to 12%, it is still the lowest among cancers. Hence, novel therapeutic strategies are urgently needed. Challenges in PC-targeted therapeutic strategies stem from the high PC heterogeneity and from the poorly understood interplay between cancer cells and the surrounding microenvironment. Signaling pathways that drive PC cell growth have been the subject of intense scrutiny and interest has been attracted by necroptosis, a distinct type of programmed cell death. In this review, we provide a historical background on necroptosis and a detailed analysis of the ongoing debate on the role of necroptosis in PC malignant progression.
Collapse
Affiliation(s)
- Valentina Giansante
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Gianmarco Stati
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Silvia Sancilio
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Emanuela Guerra
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technologies (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Saverio Alberti
- Unit of Medical Genetics, Department of Biomedical Sciences, University of Messina, 98122 Messina, Italy
| | - Roberta Di Pietro
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
18
|
Dec P, Poniewierska-Baran A, Modrzejewski A, Pawlik A. The Role of Omentin-1 in Cancers Development and Progression. Cancers (Basel) 2023; 15:3797. [PMID: 37568613 PMCID: PMC10417146 DOI: 10.3390/cancers15153797] [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: 06/16/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Adipose tissue serves as an energy store and is also an active endocrine organ, exerting activity that influences obesity-related processes through the production of regulatory proteins called adipokines or adipocytokines. Adipokines play important direct and indirect roles in the pathogenesis of insulin resistance, the regulation of local and systemic inflammatory processes, and related metabolic complications. There have been an increasing number of studies showing the relationship between some adipokines and carcinogenesis. This work reviews the current literature concerning the effects of omentin-1 on carcinogenesis.
Collapse
Affiliation(s)
- Paweł Dec
- Department of Plastic and Reconstructive Surgery, 109 Military Hospital, 71-422 Szczecin, Poland;
| | - Agata Poniewierska-Baran
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
19
|
Binang HB, Perera CJ, Apte MV. Role of Pancreatic Tumour-Derived Exosomes and Their Cargo in Pancreatic Cancer-Related Diabetes. Int J Mol Sci 2023; 24:10203. [PMID: 37373351 DOI: 10.3390/ijms241210203] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
One of the most common and deadly types of pancreatic cancer (PC) is pancreatic ductal adenocarcinoma (PDAC), with most patients succumbing to the disease within one year of diagnosis. Current detection strategies do not address asymptomatic PC; therefore, patients are diagnosed at an advanced stage when curative treatment is often no longer possible. In order to detect PC in asymptomatic patients earlier, the risk factors that could serve as reliable markers need to be examined. Diabetic mellitus (DM) is a significant risk factor for this malignancy and can be both a cause and consequence of PC. Typically, DM caused by PC is known as new-onset, pancreatogenic, pancreoprivic, or pancreatic cancer-related diabetes (PCRD). Although PCRD is quite distinct from type 2 DM (T2DM), there are currently no biomarkers that differentiate PCRD from T2DM. To identify such biomarkers, a better understanding of the mechanisms mediating PCRD is essential. To this end, there has been a growing research interest in recent years to elucidate the role of tumour-derived exosomes and their cargo in the pathogenesis of PCRD. Exosomes derived from tumours can be recognized for their specificity because they reflect the characteristics of their parent cells and are important in intercellular communication. Their cargo consists of proteins, lipids, and nucleic acids, which can be transferred to and alter the behaviour of recipient cells. This review provides a concise overview of current knowledge regarding tumour-derived exosomes and their cargo in PCRD and discusses the potential areas worthy of further study.
Collapse
Affiliation(s)
- Helen B Binang
- Pancreatic Research Group, South Western Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia
| | - Chamini J Perera
- Pancreatic Research Group, South Western Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia
| |
Collapse
|
20
|
Farahmand Y, Tehrany PM, Nazari A, Nava ZH, Alsaffar MF, Yazdani O, Adili A, Esbati R, Ghafouri K. A comprehensive survey into the role of exosomes in pancreatic cancer; from the origin of cancer to the progress and possibility of diagnosis and treatment. Pathol Res Pract 2023; 245:154465. [PMID: 37119731 DOI: 10.1016/j.prp.2023.154465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
Pancreatic cancer is the fourth most common malignant tumor in the world, which has a high mortality rate due to high invasiveness, early metastases, lack of specific symptoms, and high invasiveness. Recent studies have shown that exosomes can be essential sources of biomarkers in pancreatic cancer. Over the past ten years, exosomes have been implicated in multiple trials to prevent the growth and metastasis of many cancers, including pancreatic cancer. Exosomes also play essential roles in immune evasion, invasion, metastasis, proliferation, apoptosis, drug resistance, and cancer stemness. Exosomes help cells communicate by carrying proteins and genetic material, such as non-coding RNAs, including mRNAs and microRNAs. This review examines the biological significance of exosomes in pancreatic cancer and their functions in tumor invasion, metastasis, treatment resistance, proliferation, stemness, and immune evasion. We also emphasize recent advances in our understanding of the main functions of exosomes in diagnosing and treating pancreatic cancer.
Collapse
Affiliation(s)
- Yalda Farahmand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooya M Tehrany
- Faculty of Medicine, National University of Malaysia, Bani, Malaysia
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | | | - Marwa Fadhil Alsaffar
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, 51001 Hillah, Babil, Iraq
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Ali Adili
- Tabriz University of Medical Sciences, Tabriz, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran.
| | - Kimia Ghafouri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Twohig PA, Butt MU, Gardner TB, Chahal P, Sandhu DS. Racial and Gender Disparities Among Obese Patients With Pancreatic Cancer: A Trend Analysis in the United States. J Clin Gastroenterol 2023; 57:410-416. [PMID: 35324480 DOI: 10.1097/mcg.0000000000001688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/04/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is the third leading cause of cancer death. Obesity can increase the risk of PC by up to 50%. Studies have shown racial and gender disparities in PC, however, there is a paucity of such information in obese PC patients. AIM The aim of this study was to: (1) evaluate the incidence and prevalence of obesity among PC patients in the United States over the last 15 years, and (2) determine if variation exists in the demographic of obese PC patients over the last 15 years. It is hoped that this information could be used to assist in primary prevention and early detection of PC. METHODS A population-based retrospective analysis in IBM Explorys, a pooled, national, deidentified database of 63 million patients from 300 hospitals in the United States. Patient populations were identified using SNOMED and ICD codes. Cochrane-Armitage testing was performed to analyze trends in obesity among PC. Subgroup analysis for gender, age, race, and mortality rate were assessed. RESULTS The percentage of obese patients with PC increased over the 15-year period (2.5% to 8.5%, P <0.0001). Rates of obesity among PC patients increased among females ( P =0.0004), individuals under age 65 years ( P =0.0002), and all races, but especially for African Americans ( P =0.0007) and those in minority groups. CONCLUSION Awareness of disparities in PC and applying targeted care to those at increased risk are essential to improve future outcomes, including increased health care access and recruitment in research studies for minority groups.
Collapse
Affiliation(s)
- Patrick A Twohig
- Department of Gastroenterology & Hepatology, University of Nebraska Medical Center, Omaha, NE
- Departments of Internal Medicine
| | - Muhammad U Butt
- Department of Cardiac Electrophysiology, New York University Langone Hospital, New York, NY
| | - Timothy B Gardner
- Department of Gastroenterology & Hepatology, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Prabhleen Chahal
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Dalbir S Sandhu
- Gastroenterology & Hepatology, MetroHealth Medical Center, Case Western Reserve University
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH
| |
Collapse
|
22
|
Li P, Hu Y, Scelo G, Myrskylä M, Martikainen P. Pre-existing psychological disorders, diabetes, and pancreatic cancer: A population-based study of 38,952 Finns. Cancer Epidemiol 2023; 82:102307. [PMID: 36459909 DOI: 10.1016/j.canep.2022.102307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND It remains unclear how pre-existing depression, anxiety, and diabetes of different durations are associated with the risk of pancreatic cancer, its clinical characteristics, treatment modalities, and subsequent survival. METHODS From a register-based random sample of Finns residing in Finland at the end of the period 1987-2007, 6492 patients diagnosed with primary pancreatic cancer in 2000-2014, and 32 460 controls matched for birth cohort and sex, were identified. Pre-existing depression, anxiety, and diabetes were ascertained from the records of prescribed medication purchases. Information on pancreatic cancer outcomes was obtained from the Finnish cancer register. Data were analyzed using logistic and Cox regressions. RESULTS The risk of developing pancreatic cancer was found to be associated with long-term anxiety (treatment started 36 + months before the cancer diagnosis) (odds ratio (OR): 1.13, 95% confidence interval (95%CI): 1.04-1.22) and long-term diabetes (OR 1.72, 95%CI 1.55-1.90), as well as with new-onset (treatment started 0-24 months before the cancer diagnosis) depression (OR 1.59, 95%CI 1.34-1.88), anxiety (OR 1.76, 95%CI 1.50-2.07), and diabetes (OR 3.92, 95%CI 3.44-4.48). However, the effects of these new-onset conditions were driven by cases that began treatment within 3 months before the cancer diagnosis (concomitant period). Patients with long-term depression, anxiety and diabetes and those with new-onset anxiety had a higher risk of not receiving standard treatments. Lower survival was found for pancreatic cancer patients with new-onset depression (hazards ratio (HR) 1.38, 95%CI 1.16-1.64). Survival was not associated with pre-existing anxiety or diabetes. CONCLUSIONS The associations between pancreatic cancer risk and pre-existing depression and anxiety were mostly driven by concomitant effects. Individuals with diabetes, regardless of duration, should be closely monitored for pancreatic cancer. Pancreatic cancer patients with new-onset depression should be targeted to improve their survival.
Collapse
Affiliation(s)
- Peng Li
- Max Planck Institute for Demographic Research, Rostock, Germany
| | - Yaoyue Hu
- School of Public Health, Chongqing Medical University, Chongqing, China.
| | - Ghislaine Scelo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Mikko Myrskylä
- Max Planck Institute for Demographic Research, Rostock, Germany; Population Research Unit, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland
| | - Pekka Martikainen
- Max Planck Institute for Demographic Research, Rostock, Germany; Population Research Unit, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Modanwal S, Mishra N. Identification of common genes in obesity and cancer through network interaction and targeting those genes by virtual screening approach. J Biomol Struct Dyn 2023; 41:1109-1127. [PMID: 34961392 DOI: 10.1080/07391102.2021.2020169] [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] [Indexed: 01/11/2023]
Abstract
Obesity may have an effect on cancer outcomes, resulting in global inequalities in cancer survival and death. Microarray data analysis was done to identify differentially expressed genes (DEGs) in obese and cancer patients. Total 1977 differentially expressed genes among obesity and gastric cancer, breast cancer, pancreatic cancer, and colorectal cancer were used to build a gene interaction network, which was then analyzed by using Cytoscape software. It has been identified that JUN, CXCL12, and LEP genes show a higher degree and stress, and play an important role in obesity and cancer progression. Further, CXCL12 and LEP were taken for virtual screening study with coumarin and its derivatives to develop a drug against obesity and cancer. The interactions of CXCL12 and LEP with coumarins were studied by molecular docking and it shows good interaction as well as docking score as compared to the standard one. The ADME properties were predicted to check the drug-likeness activity of coumarins and the most of the drug-likeness activities are in admire range. The Binding free energy of the docked complex was calculated by performing MM-GBSA. The molecular docking, ADME properties prediction, and MM-GBSA was performed on Maestro 12.6. The top docked score compounds were further subjected to molecular dynamic simulation to check the stability by using GROMACS. The MM-PBSA study was performed to calculate the binding energy components as well as the energy contributions of specific amino acids. The resultant compounds could be a potent anti-obesity and anti-cancer drug.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shristi Modanwal
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, U.P, India
| | - Nidhi Mishra
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, U.P, India
| |
Collapse
|
24
|
Pathophysiology of obesity and its associated diseases. Acta Pharm Sin B 2023. [DOI: 10.1016/j.apsb.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
25
|
Tsuchiya T, Saisho Y, Inaishi J, Sasaki H, Sato M, Nishikawa M, Masugi Y, Yamada T, Itoh H. Increased alpha cell to beta cell ratio in patients with pancreatic cancer. Endocr J 2022; 69:1407-1414. [PMID: 35934795 DOI: 10.1507/endocrj.ej22-0170] [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] [Indexed: 12/30/2022] Open
Abstract
The development of pancreatic cancer (PC) is associated with worsening of glucose tolerance. However, there is limited information about the effects of PC on islet morphology. The aim of this study was to elucidate changes in alpha and beta cell mass in patients with PC. We enrolled 30 autopsy cases with death due to PC (9 with diabetes; DM) and 31 age- and BMI-matched autopsy cases without PC (controls, 12 with DM). Tumor-free pancreatic sections were stained for insulin and glucagon, and fractional beta cell (BCA) and alpha cell area (ACA) were quantified. In addition, expression of de-differentiation markers, i.e., ALDH1A3 and UCN3, was qualitatively evaluated. The pancreas of subjects with PC showed atrophic and fibrotic changes. There was no significant difference in BCA in subjects with PC compared to controls (1.53 ± 1.26% vs. 0.95 ± 0.42%, p = 0.07). However, ACA and ACA to BCA ratio were significantly higher in subjects with PC compared to controls (2.48 ± 2.39% vs. 0.53 ± 0.26% and 1.94 ± 1.93 vs. 0.59 ± 0.26, respectively, both p < 0.001). Increased ACA to BCA ratio was observed in subjects with PC irrespective of the presence of DM. Qualitative evaluation of ALDH1A3 and UCN3 expression showed no significant difference between the groups. In conclusion, in subjects with PC, alpha to beta cell mass ratio is increased, which may contribute to the increased risk of worsening glucose metabolism. Further studies are warranted to elucidate the mechanisms of increased alpha to beta cell mass in patients with PC.
Collapse
Affiliation(s)
- Tami Tsuchiya
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yoshifumi Saisho
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Saisho Diabetes Clinic, Tokyo 164-0001, Japan
| | - Jun Inaishi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hironobu Sasaki
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Midori Sato
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masaru Nishikawa
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Taketo Yamada
- Department of Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Pathology, Saitama Medical University, Saitama 350-0495, Japan
| | - Hiroshi Itoh
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
26
|
Gianfredi V, Ferrara P, Dinu M, Nardi M, Nucci D. Diets, Dietary Patterns, Single Foods and Pancreatic Cancer Risk: An Umbrella Review of Meta-Analyses. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14787. [PMID: 36429506 PMCID: PMC9691178 DOI: 10.3390/ijerph192214787] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Pancreatic cancer (PC) represents the third leading cause of cancer death in 2020. Despite the fact that, in 2018, the World Cancer Research Fund report concluded that there is still a lack of evidence on the role of foods or diets and risk for PC, a flourishing body of evidence has been published and needs to be analyzed. For this reason, we conducted an umbrella review on the association between different dietary patterns/food components and PC. Data sources PubMed/MEDLINE, Scopus, Web of Science, EMBASE, and the Cochrane Collaboration were searched. The Joanna Briggs Institute Umbrella Review Methodology was used. The protocol was registered in PROSPERO. A total of 23 articles were included, covering a wide range of dietary patterns/food components: healthy/prudent dietary patterns (n = 4), Mediterranean diets (MedDiet) (n = 1), plant-based diets (n = 2), the Dietary Inflammatory Index (DII) (n = 2), western diets (n = 2), and, lastly, unhealthy diets (n = 2). Regarding dietary components, the following were assessed: total fruit (n = 2), citrus fruit (n = 1), total vegetables (n = 2), cruciferous vegetables (n = 1), red meat (n = 6), processed meat (n = 4), poultry (n = 2), eggs (n = 1), fish (n = 5), whole grain (n = 2), potato (n = 1), and nuts (n = 2). The methodological quality of the included meta-analyses was generally low or critically low. Although the strength of evidence was generally weak, convincing or suggestive evidence was found for a healthy/prudent, plant-based diet, fruit and vegetables, and lower risk of PC, whereas a high intake of red meat was associated with a higher risk of PC at a convincing level of evidence. Further studies are needed to confirm the role of the other dietary patterns/food components and the risk of PC.
Collapse
Affiliation(s)
- Vincenza Gianfredi
- Department of Biomedical Sciences for Health, University of Milan, Via Pascal, 36, 20133 Milan, Italy
- CAPHRI Care and Public Health Research Institute, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Pietro Ferrara
- Center for Public Health Research, University of Milan-Bicocca, 20900 Monza, Italy
- IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Monica Dinu
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Mariateresa Nardi
- Nutritional Support Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy
| | - Daniele Nucci
- Nutritional Support Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy
| |
Collapse
|
27
|
Fukui H, Onishi H, Nakamoto A, Tsuboyama T, Ota T, Yano K, Enchi Y, Yamada D, Takeda Y, Kobayashi S, Fukuda Y, Eguchi H, Matsui T, Tatsumi M, Tomiyama N. Pancreatic fibrosis by extracellular volume fraction using Contrast-enhanced computed tomography and relationship with pancreatic cancer. Eur J Radiol 2022; 156:110522. [PMID: 36113381 DOI: 10.1016/j.ejrad.2022.110522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE This study aimed to assess the relationship between pancreatic fibrosis measured by the extracellular volume fraction (ECV) using contrast-enhanced computed tomography (CT) and the histologic pancreatic fibrosis fraction and investigate the relationship between pancreatic fibrosis and pancreatic cancer. METHOD The study included 88 consecutive patients (48 males, 40 females; median age, 69 years; range, 17-89 years); 47 had pancreatic cancer, and 41 had other diseases. Fifty-two cases were evaluated pathologically for pancreatic fibrosis. The histologic pancreatic fibrosis fraction was quantified using image analysis software in nontumorous pancreatic tissue at the resection stump using 2-µm-thick Azan-stained slides. Two board-certified radiologists measured ECV in the pancreatic parenchyma at an estimated transection line. The correlation between histologic pancreatic fibrosis fraction and ECV was investigated, and whether the ECV value could be used as a biomarker for pancreatic cancer was investigated. RESULTS The histologic pancreatic fibrosis fraction was significantly correlated with the ECV (r = 0.64, P < 0.01). Pancreatic fibrosis evaluated by ECV was higher in pancreatic cancer patients than in other patients (P < 0.01). On receiver-operating characteristic curve analysis, the ECV had good diagnostic accuracy for the development of pancreatic cancer (cut-off value 32.8%; sensitivity 61.0%, specificity 85.1%). ECV was identified on multivariate analysis as an independent risk factor for pancreatic cancer (odds ratio 1.16; P < 0.01). CONCLUSIONS Extracellular volume fraction was strongly related to the histologic pancreatic fibrosis fraction, which was independently associated with pancreatic cancer. Thus, extracellular volume fraction is an imaging biomarker that reflects the progression of pancreatic fibrosis and may potentially help predict the development of pancreatic cancer, although further investigation will be needed.
Collapse
Affiliation(s)
- Hideyuki Fukui
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan.
| | - Hiromitsu Onishi
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Atsushi Nakamoto
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Takahiro Tsuboyama
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Takashi Ota
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Keigo Yano
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Yukihiro Enchi
- Division of Radiology, Department of Medical Technology, Osaka University Hospital, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Yu Takeda
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Yasunari Fukuda
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan; Department of Surgery, Kinan Hospital, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Takahiro Matsui
- Department of Pathology, Osaka University Graduate School of Medicine, Japan
| | - Mitsuaki Tatsumi
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Noriyuki Tomiyama
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
28
|
Kfoury S, Michl P, Roth L. Modeling Obesity-Driven Pancreatic Carcinogenesis-A Review of Current In Vivo and In Vitro Models of Obesity and Pancreatic Carcinogenesis. Cells 2022; 11:3170. [PMID: 36231132 PMCID: PMC9563584 DOI: 10.3390/cells11193170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy with a 5-year survival rate below 10%, thereby exhibiting the worst prognosis of all solid tumors. Increasing incidence together with a continued lack of targeted treatment options will cause PDAC to be the second leading cause of cancer-related deaths in the western world by 2030. Obesity belongs to the predominant risk factors for pancreatic cancer. To improve our understanding of the impact of obesity on pancreatic cancer development and progression, novel laboratory techniques have been developed. In this review, we summarize current in vitro and in vivo models of PDAC and obesity as well as an overview of a variety of models to investigate obesity-driven pancreatic carcinogenesis. We start by giving an overview on different methods to cultivate adipocytes in vitro as well as various in vivo mouse models of obesity. Moreover, established murine and human PDAC cell lines as well as organoids are summarized and the genetically engineered models of PCAC compared to xenograft models are introduced. Finally, we review published in vitro and in vivo models studying the impact of obesity on PDAC, enabling us to decipher the molecular basis of obesity-driven pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Sally Kfoury
- Department of Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
| | - Patrick Michl
- Department of Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
- Department of Medicine, Internal Medicine IV, University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120 Heidelberg, Germany
| | - Laura Roth
- Department of Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
29
|
George S, Jean-Baptiste W, Yusuf Ali A, Inyang B, Koshy FS, George K, Poudel P, Chalasani R, Goonathilake MR, Waqar S, Mohammed L. The Role of Type 2 Diabetes in Pancreatic Cancer. Cureus 2022; 14:e26288. [PMID: 35898377 PMCID: PMC9308974 DOI: 10.7759/cureus.26288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/24/2022] [Indexed: 11/05/2022] Open
Abstract
The incidence of type 2 diabetes mellitus (T2DM) and its potential complications, such as cancers, are increasing worldwide at an astounding rate. There are many factors such as obesity, diabetes, alcohol consumption, and the adoption of sedentary lifestyles that are driving pancreatic cancer (PC) to become one of the leading causes of cancer mortality in the United States. PC is notorious for its generic symptoms and late-stage presentation with rapid metastasis. The connection between T2DM and the risk of PC development is multifaceted and complex. Some of the proposed theories reveal that chronic inflammation, insulin resistance, hyperinsulinemia, hyperglycemia, and abnormalities in the insulin and insulin-like growth factor axis (IGF) contribute to the disease association between these two conditions. This literature review aims to highlight relevant studies and explore the molecular mechanisms involved in the etiology of diabetes and its impact on PC development, as well as the role of anti-diabetic agents on PC. Despite extensive studies, the exact interaction between T2DM and PC remains obscure and will need further investigation. According to current knowledge, there is a substantial link between diabetes, obesity, and dietary patterns in the development and progression of PC. Consequently, focusing our efforts on preventive measures by reducing modifiable risk factors remains the most effective strategy to reduce the risk of PC at this time. Antidiabetic drugs can have various effects on the occurrence and prognosis of PC with metformin offering a clear benefit of inhibiting PC and insulin increasing the risk of PC. The development of future novel therapies will require a deeper knowledge of the triggering mechanisms and interplay between these two disease states.
Collapse
|
30
|
Shinoda S, Nakamura N, Roach B, Bernlohr DA, Ikramuddin S, Yamamoto M. Obesity and Pancreatic Cancer: Recent Progress in Epidemiology, Mechanisms and Bariatric Surgery. Biomedicines 2022; 10:1284. [PMID: 35740306 PMCID: PMC9220099 DOI: 10.3390/biomedicines10061284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 12/24/2022] Open
Abstract
More than 30% of people in the United States (US) are classified as obese, and over 50% are considered significantly overweight. Importantly, obesity is a risk factor not only for the development of metabolic syndrome but also for many cancers, including pancreatic ductal adenocarcinoma (PDAC). PDAC is the third leading cause of cancer-related death, and 5-year survival of PDAC remains around 9% in the U.S. Obesity is a known risk factor for PDAC. Metabolic control and bariatric surgery, which is an effective treatment for severe obesity and allows massive weight loss, have been shown to reduce the risk of PDAC. It is therefore clear that elucidating the connection between obesity and PDAC is important for the identification of a novel marker and/or intervention point for obesity-related PDAC risk. In this review, we discussed recent progress in obesity-related PDAC in epidemiology, mechanisms, and potential cancer prevention effects of interventions, including bariatric surgery with preclinical and clinical studies.
Collapse
Affiliation(s)
- Shuhei Shinoda
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.S.); (N.N.); (B.R.); (S.I.)
| | - Naohiko Nakamura
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.S.); (N.N.); (B.R.); (S.I.)
| | - Brett Roach
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.S.); (N.N.); (B.R.); (S.I.)
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Sayeed Ikramuddin
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.S.); (N.N.); (B.R.); (S.I.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.S.); (N.N.); (B.R.); (S.I.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
31
|
Zeng J, Shen F, Zou ZY, Yang RX, Jin Q, Yang J, Chen GY, Fan JG. Association of maternal obesity and gestational diabetes mellitus with overweight/obesity and fatty liver risk in offspring. World J Gastroenterol 2022; 28:1681-1691. [PMID: 35581961 PMCID: PMC9048784 DOI: 10.3748/wjg.v28.i16.1681] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/30/2021] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Childhood obesity and fatty liver are associated with adverse outcomes such as diabetes, metabolic syndrome, and cardiovascular diseases in adulthood. It is very important to identify relevant risk factors and intervene as early as possible. At present, the relationship between maternal and offspring metabolic factors is conflicting.
AIM To estimate the association of maternal obesity and gestational diabetes mellitus (GDM) with overweight/obesity and fatty liver risk in offspring at 8 years of age.
METHODS The prospective study included mothers who all had a 75-g oral glucose tolerance test at 24-28 wk of gestation and whose offspring completed follow-up at 8 years of age. Offspring birth weight, sex, height, weight, and body mass index (BMI) were measured and calculated. FibroScan-502 examination with an M probe (Echosens, Paris, France) was prospectively conducted in offspring aged 8 years from the Shanghai Prenatal Cohort Study.
RESULTS A total of 430 mother-child pairs were included in the analysis. A total of 62 (14.2%) mothers were classified as obese, and 48 (11.1%) were classified as having GDM. The mean age of the offspring at follow-up was 8 years old. Thirty-seven (8.6%) offspring were overweight, 14 (3.3%) had obesity, and 60 (14.0%) had fatty liver. The prevalence of overweight, obesity and fatty liver in offspring increased significantly across maternal BMI quartiles (all P < 0.05). Among offspring of mothers with GDM, 12 (25.0%) were overweight, 4 (8.3%) were obese, and 12 (25.0%) had fatty liver vs. 25 (6.5%), 10 (2.6%) and 48 (12.6%), respectively, for offspring of mothers without GDM (all P < 0.05). In multiple logistic regression, after adjustment for variables, the OR for fatty liver in offspring was 8.26 (95%CI: 2.38-28.75) for maternal obesity and GDM.
CONCLUSION This study showed that maternal obesity can increase the odds of overweight/obesity and fatty liver in offspring, and GDM status also increases the odds of overweight/obesity in offspring. Weight management and glycemic control before and during pregnancy need to be highlighted in primary prevention of pediatric obesity and fatty liver.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zi-Yuan Zou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Rui-Xu Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian Jin
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guang-Yu Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| |
Collapse
|
32
|
Stott MC, Oldfield L, Hale J, Costello E, Halloran CM. Recent advances in understanding pancreatic cancer. Fac Rev 2022; 11:9. [PMID: 35509672 PMCID: PMC9022729 DOI: 10.12703/r/11-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an intractable cancer and a leading cause of cancer deaths worldwide. Over 90% of patients die within 1 year of diagnosis. Deaths from PDAC are increasing and it remains a cancer of substantial unmet need. A number of factors contribute to its poor prognosis: namely, late presentation, early metastases and limited systemic therapy options because of chemoresistance. A variety of research approaches underway are aimed at improving patient survival. Here, we review high-risk groups and efforts for early detection. We examine recent developments in the understanding of complex molecular and metabolic alterations which accompany PDAC. We explore artificial intelligence and biological targets for therapy and examine the role of tumour stroma and the immune microenvironment. We also review recent developments with respect to the PDAC microbiome. It is hoped that current research efforts will translate into earlier diagnosis, improvements in treatment and better outcomes for patients.
Collapse
Affiliation(s)
- Martyn C Stott
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Lucy Oldfield
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Jessica Hale
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Eithne Costello
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Christopher M Halloran
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| |
Collapse
|
33
|
Fatty Pancreas-Centered Metabolic Basis of Pancreatic Adenocarcinoma: From Obesity, Diabetes and Pancreatitis to Oncogenesis. Biomedicines 2022; 10:biomedicines10030692. [PMID: 35327494 PMCID: PMC8945032 DOI: 10.3390/biomedicines10030692] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest types of cancer, and it is currently the third most common cause of cancer death in the U.S.A. Progress in the fight against PDAC has been hampered by an inability to detect it early in the overwhelming majority of patients, and also by the reduced oxygen levels and nutrient perfusion caused by new matrix formation through the activation of stromal cells in the context of desmoplasia. One harbinger of PDAC is excess intrapancreatic fat deposition, namely, fatty pancreas, which specifically affects the tumor macro- and microenvironment in the organ. Over half of PDAC patients have diabetes mellitus (DM) at the time of diagnosis, and fatty pancreas is associated with subsequent DM development. Moreover, there is a strong association between fatty pancreas and fatty liver through obesity, and a higher intrapancreatic fat percentage has been noted in acute pancreatitis patients with DM than in those without DM. All these findings suggest that the link between fatty pancreas and PDAC might occur through metabolic alterations, either DM-related or non-DM-related. Based on clinical, in vivo and in vitro evidence, the current review highlights the etiologies of fatty pancreas (including fatty infiltration and replacement) and the fatty pancreas-associated metabolic alterations involved in oncogenesis to provide crucial targets to prevent, detect, and/or effectively treat PDAC.
Collapse
|
34
|
Dehghanian F, Azhir Z, Khalilian S, Grüning B. Non-coding RNAs underlying the pathophysiological links between type 2 diabetes and pancreatic cancer: A systematic review. J Diabetes Investig 2022; 13:405-428. [PMID: 34859606 PMCID: PMC8902405 DOI: 10.1111/jdi.13727] [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: 08/02/2021] [Revised: 11/11/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes is known as a risk factor for pancreatic cancer (PC). Various genetic and environmental factors cause both these global chronic diseases. The mechanisms that define their relationships are complex and poorly understood. Recent studies have implicated that metabolic abnormalities, including hyperglycemia and hyperinsulinemia, could lead to cell damage responses, cell transformation, and increased cancer risk. Hence, these kinds of abnormalities following molecular events could be essential to develop our understanding of this complicated link. Among different molecular events, focusing on shared signaling pathways including metabolic (PI3K/Akt/mTOR) and mitogenic (MAPK) pathways in addition to regulatory mechanisms of gene expression such as those involved in non-coding RNAs (miRNAs, circRNAs, and lncRNAs) could be considered as powerful tools to describe this association. A better understanding of the molecular mechanisms involved in the development of type 2 diabetes and pancreatic cancer would help us to find a new research area for developing therapeutic and preventive strategies. For this purpose, in this review, we focused on the shared molecular events resulting in type 2 diabetes and pancreatic cancer. First, a comprehensive literature review was performed to determine similar molecular pathways and non-coding RNAs; then, the final results were discussed in more detail.
Collapse
Affiliation(s)
- Fariba Dehghanian
- Department of Cell and Molecular Biology and MicrobiologyFaculty of Biological Science and TechnologyUniversity of IsfahanIsfahanIran
| | - Zahra Azhir
- Department of Cell and Molecular Biology and MicrobiologyFaculty of Biological Science and TechnologyUniversity of IsfahanIsfahanIran
| | - Sheyda Khalilian
- Department of Cell and Molecular Biology and MicrobiologyFaculty of Biological Science and TechnologyUniversity of IsfahanIsfahanIran
| | - Björn Grüning
- Department of Computer ScienceBioinformatics GroupUniversity of FreiburgFreiburgGermany
| |
Collapse
|
35
|
Jiang J, Mei J, Ma Y, Jiang S, Zhang J, Yi S, Feng C, Liu Y, Liu Y. Tumor hijacks macrophages and microbiota through extracellular vesicles. EXPLORATION (BEIJING, CHINA) 2022; 2:20210144. [PMID: 37324578 PMCID: PMC10190998 DOI: 10.1002/exp.20210144] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/16/2021] [Indexed: 06/17/2023]
Abstract
The tumor microenvironment (TME) is a biological system with sophisticated constituents. In addition to tumor cells, tumor-associated macrophages (TAMs) and microbiota are also dominant components. The phenotypic and functional changes of TAMs are widely considered to be related to most tumor progressions. The chronic colonization of pathogenic microbes and opportunistic pathogens accounts for the generation and development of tumors. As messengers of cell-to-cell communication, tumor-derived extracellular vesicles (TDEVs) can transfer various malignant factors, regulating physiological and pathological changes in the recipients and affecting TAMs and microbes in the TME. Despite the new insights into tumorigenesis and progress brought by the above factors, the crosstalk among tumor cells, macrophages, and microbiota remain elusive, and few studies have focused on how TDEVs act as an intermediary. We reviewed how tumor cells recruit and domesticate macrophages and microbes through extracellular vehicles and how hijacked macrophages and microbiota interact with tumor-promoting feedback, achieving a reciprocal coexistence under the TME and working together to facilitate tumor progression. It is significant to seek evidence to clarify those specific interactions and reveal therapeutic targets to curb tumor progression and improve prognosis.
Collapse
Affiliation(s)
- Jipeng Jiang
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing P. R. China
- University of Chinese Academy of Science Beijing P. R. China
| | - Yongfu Ma
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Shasha Jiang
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Jian Zhang
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Shaoqiong Yi
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Changjiang Feng
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Yang Liu
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing P. R. China
- GBA National Institute for Nanotechnology Innovation Guangdong P. R. China
| |
Collapse
|
36
|
Kesh K, Mendez R, Mateo-Victoriano B, Garrido VT, Durden B, Gupta VK, Oliveras Reyes A, Merchant N, Datta J, Banerjee S, Banerjee S. Obesity enriches for tumor protective microbial metabolites and treatment refractory cells to confer therapy resistance in PDAC. Gut Microbes 2022; 14:2096328. [PMID: 35816618 PMCID: PMC9275504 DOI: 10.1080/19490976.2022.2096328] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity causes chronic inflammation and changes in gut microbiome. However, how this contributes to poor survival and therapy resistance in patients with pancreatic cancer remain undetermined. Our current study shows that high fat diet-fed obese pancreatic tumor bearing mice do not respond to standard of care therapy with gemcitabine and paclitaxel when compared to corresponding control diet-fed mice. C57BL6 mice were put on control and high fat diet for 1 month following with pancreatic tumors were implanted in both groups. Microbiome of lean (control) and obese (high fat diet fed) mice was analyzed. Fecal matter transplant from control mice to obese mice sensitized tumors to chemotherapy and demonstrated extensive cell death. Analysis of gut microbiome showed an enrichment of queuosine (Q) producing bacteria in obese mice and an enrichment of S-adenosyl methionine (SAM) producing bacteria in control diet-fed mice. Further, supplementation of obese animals with SAM sensitized pancreatic tumors to chemotherapy. Treatment of pancreatic cancer cells with Q increased PRDX1 involved in oxidative stress protection. In parallel, tumors in obese mice showed increase in CD133+ treatment refractory tumor populations compared to control animals. These observations indicated that microbial metabolite Q accumulation in high fat diet-fed mice protected tumors from chemotherapy induced oxidative stress by upregulating PRDX1. This protection could be reversed by treatment with SAM. We conclude that relative concentration of SAM and queuosine in fecal samples of pancreatic cancer patients can be developed as a potential biomarker and therapeutic target in chemotherapy refractory pancreatic cancer.
Collapse
Affiliation(s)
- Kousik Kesh
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Roberto Mendez
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Vanessa T Garrido
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Brittany Durden
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Vineet K Gupta
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Nipun Merchant
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Jashodeep Datta
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Santanu Banerjee
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA
| | - Sulagna Banerjee
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| |
Collapse
|
37
|
Wirth K, Shinoda S, Sato-Dahlman M, Dickey DM, Bernlohr DA, Ikramuddin S, Yamamoto M. Fatty acid binding protein 4 regulates pancreatic cancer cell proliferation via activation of nuclear factor E2-related factor 2. Surg Obes Relat Dis 2021; 18:485-493. [PMID: 34998697 DOI: 10.1016/j.soard.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Obesity and diabetes are associated with an increased incidence of pancreatic cancer. Fatty acid binding protein 4 (FABP4), noted to be higher in patients with severe obesity, is linked to the development and progression of several cancers, and its level in the circulation decreases after bariatric surgery. OBJECTIVE In this paper, we evaluate the role of FABP4 in pancreatic cancer progression. SETTING University Hospital and Laboratories, United States. METHODS AND RESULTS When Panc-1 (human) and Pan02 (mouse) pancreatic cancer cells were treated with FABP4 or the-single-point mutant FABP4 (R126Q, fatty acid binding site mutant), only FABP4 stimulated cellular proliferation. The transcriptional activity of nuclear factor E2-related factor 2 (NRF2) was increased in response to FABP4 but not the R126Q. FABP4 treatment also led to downregulation of reactive oxygen species (ROS) activity. Consistent with induced cell propagation by FABP4, the growth of Pan02 tumor was decreased in FABP4-null animals compared with C57BL/6J controls. CONCLUSION These results suggest that FABP4 increases pancreatic cancer proliferation via activation of NRF2 and downregulation of ROS activity.
Collapse
Affiliation(s)
- Keith Wirth
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Shuhei Shinoda
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Mizuho Sato-Dahlman
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Deborah M Dickey
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Sayeed Ikramuddin
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
38
|
Luu HN, Paragomi P, Jin A, Wang R, Neelakantan N, van Dam RM, Brand RE, Koh WP, Yuan JM. Quality Diet Index and Risk of Pancreatic Cancer: Findings from the Singapore Chinese Health Study. Cancer Epidemiol Biomarkers Prev 2021; 30:2068-2078. [PMID: 34446471 PMCID: PMC8568638 DOI: 10.1158/1055-9965.epi-21-0033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/28/2021] [Accepted: 08/19/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Limited research has been conducted on the effect of quality diet index (QDI), which represents a comprehensive assessment of healthy diet quality and quantity, on pancreatic cancer risk in Asian populations. METHODS Using data from the Singapore Chinese Health Study, a prospective cohort of 63,257 middle-aged or older Chinese men and women, four QDI scores: the Alternative Health Eating Index-2010 (AHEI-2010), the alternate Mediterranean Diet (aMED), the Dietary Approaches to Stop Hypertension (DASH), and the Heathy Diet Indicator (HDI), at baseline were calculated. After 25 years of follow-up, 311 cohort participants developed pancreatic cancer. Cox proportional hazard regression method was used to estimate HR and 95% confidence interval (CI) for pancreatic cancer associated with higher QDI scores. RESULTS Higher scores of AHEI-2010, aMED, and DASH were significantly associated with lower pancreatic cancer risk (all P trend < 0.05). Compared with the lowest quartile, HRs (95% CIs) of pancreatic cancer for the highest quartiles of AHEI-2010, aMED, and DASH scores were 0.65 (0.46-0.90), 0.57 (0.38-0.85), and 0.66 (0.46-0.95), respectively. These associations were more apparent among men. Overall, there was no statistically significant difference in the QDI-pancreatic cancer risk association between subgroups stratified by levels of body mass index, history of diabetes, and smoking status. CONCLUSIONS Higher QDI scores were significantly associated with reduced risk of pancreatic cancer. IMPACT The consistent results across multiple QDIs shows that adherence to a healthy diet may lower pancreatic cancer risk, suggesting that dietary modification may be a promising approach for primary prevention of pancreatic cancer.
Collapse
Affiliation(s)
- Hung N Luu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania.
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | | | - Aizhen Jin
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore
| | - Renwei Wang
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Nithya Neelakantan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Randall E Brand
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Woon-Puay Koh
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jian-Min Yuan
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Yuan Q, Ren J, Wang Z, Ji L, Deng D, Shang D. Identification of the Real Hub Gene and Construction of a Novel Prognostic Signature for Pancreatic Adenocarcinoma Based on the Weighted Gene Co-expression Network Analysis and Least Absolute Shrinkage and Selection Operator Algorithms. Front Genet 2021; 12:692953. [PMID: 34490033 PMCID: PMC8417717 DOI: 10.3389/fgene.2021.692953] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Pancreatic adenocarcinoma (PAAD) has a considerably bad prognosis, and its pathophysiologic mechanism remains unclear. Thus, we aimed to identify real hub genes to better explore the pathophysiology of PAAD and construct a prognostic panel to better predict the prognosis of PAAD using the weighted gene co-expression network analysis (WGCNA) and the least absolute shrinkage and selection operator (LASSO) algorithms. Methods: WGCNA identified the modules most closely related to the PAAD stage and grade based on the Gene Expression Omnibus. The module genes significantly associated with PAAD progression and prognosis were considered as the real hub genes. Eligible genes in the most significant module were selected for construction and validation of a multigene prognostic signature based on the LASSO-Cox regression analysis in The Cancer Genome Atlas and the International Cancer Genome Consortium databases, respectively. Results: The brown module identified by WGCNA was most closely associated with the clinical characteristics of PAAD. Scaffold attachment factor B (SAFB) was significantly associated with PAAD progression and prognosis, and was identified as the real hub gene of PAAD. Moreover, both transcriptional and translational levels of SAFB were significantly lower in PAAD tissues compared with normal pancreatic tissues. In addition, a novel multigene-independent prognostic signature consisting of SAFB, SP1, and SERTAD3 was identified and verified. The predictive accuracy of our signature was superior to that of previous studies, especially for predicting 3- and 5-year survival probabilities. Furthermore, a prognostic nomogram based on independent prognostic variables was developed and validated using calibration curves. The predictive ability of this nomogram was also superior to the well-established AJCC stage and histological grade. The potential mechanisms of different prognoses between the high- and low-risk subgroups were also investigated using functional enrichment analysis, GSEA, ssGSEA, immune checkpoint analysis, and mutation profile analysis. Conclusion: SAFB was identified as the real hub gene of PAAD. A novel multigene-independent prognostic signature was successfully identified and validated to better predict PAAD prognosis. An accurate nomogram was also developed and verified to aid in the accurate treatment of tumors, as well as in early intervention.
Collapse
Affiliation(s)
- Qihang Yuan
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Ren
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhizhou Wang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li Ji
- Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dawei Deng
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
40
|
Wang B, Wang S, Wang W, Liu E, Guo S, Zhao C, Niu J, Zhang Z. Hyperglycemia Promotes Liver Metastasis of Colorectal Cancer via Upregulation of Integrin αvβ6. Med Sci Monit 2021; 27:e930921. [PMID: 34408123 PMCID: PMC8383819 DOI: 10.12659/msm.930921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Diabetes is related to higher risk of multiple cancers. This study aimed to explore the effect and mechanism of diabetes on liver metastasis of CRC. Material/Methods Overall and liver metastasis-free survival in diabetic and non-diabetic CRC patients were compared by Kaplan-Meier analysis. Expression of αvβ6 was detected by immunohistochemistry in clinical specimens. Effects of hyperglycemia on αvβ6 expression in colon cancer cells were assessed by western blot, real-time PCR, and flowcytometry. Effects of hyperglycemia on migration and invasion were demonstrated by Transwell assay. Expression and activity of MMP-9 and MMP-2 were determined by real-time PCR and gelatin zymography. Liver metastatic nodules were counted and β6 expression was detected by western blot in a liver metastasis mouse model. Results CRC patients with diabetes had poorer overall and liver metastasis-free survival, and diabetes was associated with higher αvβ6 expression in CRC specimens. Hyperglycemia promoted the invasion and migration of colon cancer cells, and upregulated the expression and activity of MMP-9, which were attenuated by inhibition of αvβ6. Hyperglycemia upregulated the expression of β6 and cell surface expression of αvβ6, which was reduced by ERK inhibitor. The in vitro results were confirmed in vivo in the mouse model. Conclusions Our study demonstrated the enhancing effect of hyperglycemia on liver metastasis of CRC, and showed that αvβ6 was involved in this process, suggesting that control of glucose levels and inhibition of αvβ6 can reduce the risk of liver metastasis in diabetic CRC patients.
Collapse
Affiliation(s)
- Ben Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Shanjie Wang
- Department of General Surgery, People's Hospital, Zhangqiu District, Jinan, Shandong, China (mainland)
| | - Wenke Wang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Enyu Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Sen Guo
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Chuanzong Zhao
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Jun Niu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
41
|
Duan X, Wang W, Pan Q, Guo L. Type 2 Diabetes Mellitus Intersects With Pancreatic Cancer Diagnosis and Development. Front Oncol 2021; 11:730038. [PMID: 34485159 PMCID: PMC8415500 DOI: 10.3389/fonc.2021.730038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
The relationship between type 2 diabetes mellitus (T2DM) and pancreatic cancer (PC) is complex. Diabetes is a known risk factor for PC, and new-onset diabetes (NOD) could be an early manifestation of PC that may be facilitate the early diagnosis of PC. Metformin offers a clear benefit of inhibiting PC, whereas insulin therapy may increase the risk of PC development. No evidence has shown that novel hypoglycemic drugs help or prevent PC. In this review, the effects of T2DM on PC development are summarized, and novel strategies for the prevention and treatment of T2DM and PC are discussed.
Collapse
Affiliation(s)
- Xiaoye Duan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Weihao Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Pan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Hu JX, Zhao CF, Chen WB, Liu QC, Li QW, Lin YY, Gao F. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J Gastroenterol 2021; 27:4298-4321. [PMID: 34366606 PMCID: PMC8316912 DOI: 10.3748/wjg.v27.i27.4298] [Citation(s) in RCA: 247] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/18/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Despite rapid advances in modern medical technology and significant improvements in survival rates of many cancers, pancreatic cancer is still a highly lethal gastrointestinal cancer with a low 5-year survival rate and difficulty in early detection. At present, the incidence and mortality of pancreatic cancer are increasing year by year worldwide, no matter in the United States, Europe, Japan, or China. Globally, the incidence of pancreatic cancer is projected to increase to 18.6 per 100000 in 2050, with the average annual growth of 1.1%, meaning that pancreatic cancer will pose a significant public health burden. Due to the special anatomical location of the pancreas, the development of pancreatic cancer is usually diagnosed at a late stage with obvious clinical symptoms. Therefore, a comprehensive understanding of the risk factors for pancreatic cancer is of great clinical significance for effective prevention of pancreatic cancer. In this paper, the epidemiological characteristics, developmental trends, and risk factors of pancreatic cancer are reviewed and analyzed in detail.
Collapse
Affiliation(s)
- Jian-Xiong Hu
- Intensive Care Unit (ICU), Affiliated Hospital of Putian University, Putian 351100, Fujian Province, China
| | - Cheng-Fei Zhao
- School of Pharmacy and Medical Technology, Putian University, Putian 351100, Fujian Province, China
- Key Laboratory of Pharmaceutical Analysis and Laboratory Medicine in University of Fujian Province, Putian University, Putian 351100, Fujian Province, China
| | - Wen-Biao Chen
- Department of Basic Medicine, Quanzhou Medical College, Quanzhou 362011, Fujian Province, China
| | - Qi-Cai Liu
- Department of Reproductive Medicine Centre, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Qu-Wen Li
- Department of Priority Laboratory for Zoonoses Research, Fujian Center for Disease Control and Prevention, Fuzhou 350001, Fujian Province, China
| | - Yan-Ya Lin
- Intensive Care Unit (ICU), Affiliated Hospital of Putian University, Putian 351100, Fujian Province, China
| | - Feng Gao
- Department of Pathology, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
43
|
Yuan Q, Ren J, Li L, Li S, Xiang K, Shang D. Development and validation of a novel N6-methyladenosine (m6A)-related multi- long non-coding RNA (lncRNA) prognostic signature in pancreatic adenocarcinoma. Bioengineered 2021; 12:2432-2448. [PMID: 34233576 PMCID: PMC8806915 DOI: 10.1080/21655979.2021.1933868] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence has unveiled the pivotal roles of N6-methyladenosine (m6A) in pancreatic adenocarcinoma (PAAD). However, there are not many researches to predict the prognosis of PAAD using m6A-related long non-coding RNAs (lncRNAs). Raw data from The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC), and the Genotype-Tissue Expression project (GTEx) were utilized to comprehensively analyze the expression and prognostic performances of 145 m6A-related lncRNAs in PAAD and to develop and validate a novel m6A-related multi-lncRNA prognostic signature (m6A-LPS) for PAAD patients. In total, 57 differentially expressed m6A-related lncRNAs with prognostic values were identified. Based on LASSO-Cox regression analysis, m6A-LPS was constructed and verified by using five-lncRNA expression profiles for TCGA and ICGC cohorts. PAAD patients were then divided into high- and low-risKBIE_A_1933868k subgroups with different clinical outcomes according to the median risk score; this was further verified by time-dependent receiver operating characteristic curves. Risk scores were significantly associated with clinical parameters such as histological grade and cancer status among PAAD patients. A nomogram consisting of risk score, grade, and cancer status was generated to predict the survival probability of PAAD patients, as also demonstrated by calibration curves. Discrepancies in cellular processes, signaling pathways, and immune status between the high- and low-risk subgroups were investigated by functional and single-sample gene set enrichment analyses. In conclusion, the novel m6A-LPS for PAAD patients was developed and validated, which might provide new insight into clinical decision-making and precision medicine.
Collapse
Affiliation(s)
- Qihang Yuan
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Ren
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lunxu Li
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuang Li
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kailai Xiang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
44
|
Nikmaneshi MR, Firoozabadi B, Mozafari A. Chemo-mechanistic multi-scale model of a three-dimensional tumor microenvironment to quantify the chemotherapy response of cancer. Biotechnol Bioeng 2021; 118:3871-3887. [PMID: 34133020 DOI: 10.1002/bit.27863] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/03/2023]
Abstract
Exploring efficient chemotherapy would benefit from a deeper understanding of the tumor microenvironment (TME) and its role in tumor progression. As in vivo experimental methods are unable to isolate or control individual factors of the TME, and in vitro models often cannot include all the contributing factors, some questions are best addressed with mathematical models of systems biology. In this study, we establish a multi-scale mathematical model of the TME to simulate three-dimensional tumor growth and angiogenesis and then implement the model for an array of chemotherapy approaches to elucidate the effect of TME conditions and drug scheduling on controlling tumor progression. The hyperglycemic condition as the most common disorder for cancer patients is considered to evaluate its impact on cancer response to chemotherapy. We show that combining antiangiogenic and anticancer drugs improves the outcome of treatment and can decrease accumulation of the drug in normal tissue and enhance drug delivery to the tumor. Our results demonstrate that although both concurrent and neoadjuvant combination therapies can increase intratumoral drug exposure and therapeutic accuracy, neoadjuvant therapy surpasses this, especially against hyperglycemia. Our model provides mechanistic explanations for clinical observations of tumor progression and response to treatment and establishes a computational framework for exploring better treatment strategies.
Collapse
Affiliation(s)
| | - Bahar Firoozabadi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Aliasghar Mozafari
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
45
|
Cancer cachexia: molecular mechanism and pharmacological management. Biochem J 2021; 478:1663-1688. [PMID: 33970218 DOI: 10.1042/bcj20201009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022]
Abstract
Cancer cachexia often occurs in malignant tumors and is a multifactorial and complex symptom characterized by wasting of skeletal muscle and adipose tissue, resulting in weight loss, poor life quality and shorter survival. The pathogenic mechanism of cancer cachexia is complex, involving a variety of molecular substrates and signal pathways. Advancements in understanding the molecular mechanisms of cancer cachexia have provided a platform for the development of new targeted therapies. Although recent outcomes of early-phase trials have showed that several drugs presented an ideal curative effect, monotherapy cannot be entirely satisfactory in the treatment of cachexia-associated symptoms due to its complex and multifactorial pathogenesis. Therefore, the lack of definitive therapeutic strategies for cancer cachexia emphasizes the need to develop a better understanding of the underlying mechanisms. Increasing evidences show that the progression of cachexia is associated with metabolic alternations, which mainly include excessive energy expenditure, increased proteolysis and mitochondrial dysfunction. In this review, we provided an overview of the key mechanisms of cancer cachexia, with a major focus on muscle atrophy, adipose tissue wasting, anorexia and fatigue and updated the latest progress of pharmacological management of cancer cachexia, thereby further advancing the interventions that can counteract cancer cachexia.
Collapse
|
46
|
Petrov MS. Post-pancreatitis diabetes mellitus and excess intra-pancreatic fat deposition as harbingers of pancreatic cancer. World J Gastroenterol 2021; 27:1936-1942. [PMID: 34007131 PMCID: PMC8108030 DOI: 10.3748/wjg.v27.i17.1936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/02/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
Progress in the fight against pancreatic cancer has been hampered by many factors. One of them is the inability to detect the disease early in overwhelming majority of patients. The present paper outlines a novel way in which progress could be accelerated. This includes a focus on two harbingers—post-pancreatitis diabetes mellitus and excess intra-pancreatic fat deposition—that converge at affecting the tumor macroenvironment and microenvironment specifically in the pancreas, not other organs. The two entities have the potential to be incorporated into future screening strategies with a view to early detecting of pancreatic cancer.
Collapse
Affiliation(s)
- Maxim S Petrov
- School of Medicine, The University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
47
|
Hu X, Xiao Y, Sun J, Ji B, Luo S, Wu B, Zheng C, Wang P, Xu F, Cheng K, Hua H, Li D. New possible silver lining for pancreatic cancer therapy: Hydrogen sulfide and its donors. Acta Pharm Sin B 2021; 11:1148-1157. [PMID: 34094825 PMCID: PMC8144891 DOI: 10.1016/j.apsb.2020.10.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/30/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
As one of the most lethal diseases, pancreatic cancer shows a dismal overall prognosis and high resistance to most treatment modalities. Furthermore, pancreatic cancer escapes early detection during the curable period because early symptoms rarely emerge and specific markers for this disease have not been found. Although combinations of new drugs, multimodal therapies, and adjuvants prolong survival, most patients still relapse after surgery and eventually die. Consequently, the search for more effective treatments for pancreatic cancer is highly relevant and justified. As a newly re-discovered mediator of gasotransmission, hydrogen sulfide (H2S) undertakes essential functions, encompassing various signaling complexes that occupy key processes in human biology. Accumulating evidence indicates that H2S exhibits bimodal modulation of cancer development. Thus, endogenous or low levels of exogenous H2S are thought to promote cancer, whereas high doses of exogenous H2S suppress tumor proliferation. Similarly, inhibition of endogenous H2S production also suppresses tumor proliferation. Accordingly, H2S biosynthesis inhibitors and H2S supplementation (H2S donors) are two distinct strategies for the treatment of cancer. Unfortunately, modulation of endogenous H2S on pancreatic cancer has not been studied so far. However, H2S donors and their derivatives have been extensively studied as potential therapeutic agents for pancreatic cancer therapy by inhibiting cell proliferation, inducing apoptosis, arresting cell cycle, and suppressing invasion and migration through exploiting multiple signaling pathways. As far as we know, there is no review of the effects of H2S donors on pancreatic cancer. Based on these concerns, the therapeutic effects of some H2S donors and NO–H2S dual donors on pancreatic cancer were summarized in this paper. Exogenous H2S donors may be promising compounds for pancreatic cancer treatment.
Collapse
Key Words
- 3-MST, 3-mercaptopyruvate sulfurtransferase
- AMPK, adenosine 5′-monophosphate-activated protein kinase
- Antitumor effect
- BCL-2, B-cell lymphoma-2
- BITC, benzyl isothiocyanate
- BRCA2, breast cancer 2
- CAT, cysteine aminotransferase
- CBS, cystathionine-β-synthase
- CDC25B, cell division cycle 25B
- CDK1, cyclin-dependent kinase 1
- CHK2, checkpoint kinase 2
- CSE, cystathionine-γ-lyase
- Cell proliferation
- DATS, diallyl trisulfide
- DR4, death receptor
- EMT, epithelial–mesenchymal transition
- ERK1/2, extracellular signal-regulated kinase
- ERU, erucin
- FOXM1, forkhead box protein M1
- GLUTs, glucose transporters
- H2S, hydrogen sulfide
- HDAC, histone deacetylase
- HEATR1, human HEAT repeat-containing protein 1
- HIF-1α, hypoxia inducible factor
- Hydrogen sulfide donor
- ITCs, isothiocyanates
- JNK, c-Jun N-terminal kinase
- KEAP1‒NRF2‒ARE, the recombinant protein 1-nuclear factor erythroid-2 related factor 2-antioxidant response element
- KRAS, kirsten rat sarcoma viral oncogene
- NF-κB, nuclear factor kappa B
- NO, nitric oxide
- OCT-4, octamer-binding transcription factor 4
- P16, multiple tumor suppressor 1
- PARP, poly(ADP-ribose)-polymerase
- PDGFRα, platelet-derived growth factor receptor
- PEITC, phenethyl isothiocyanate
- PI3K/AKT, phosphoinositide 3-kinase/v-AKT murine thymoma viral oncogene
- Pancreatic cancer
- RASAL2, RAS protein activator like 2
- ROS, reactive oxygen species
- RPL10, human ribosomal protein L10
- SFN, sulforaphane
- SHH, sonic hedgehog
- SMAD4, mothers against decapentaplegic homolog 4
- STAT-3, signal transducer and activator of transcription 3
- Signaling pathway
- Sulfur-containing compound
- TRAIL, The human tumor necrosis factor-related apoptosis-inducing ligand
- VEGF, vascular endothelial growth factor
- XIAP, X-linked inhibitor of apoptosis protein
- ZEB1, zinc finger E box-binding protein-1
- iNOS, inducible nitric oxide synthase
Collapse
Affiliation(s)
- Xu Hu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yan Xiao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianan Sun
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bao Ji
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shanshan Luo
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Bo Wu
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA 02129, USA
| | - Chao Zheng
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors. Tel./fax: +86 24 23986465.
| | - Keguang Cheng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources; School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors. Tel./fax: +86 24 23986465.
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors. Tel./fax: +86 24 23986465.
| |
Collapse
|
48
|
Talib WH, Mahmod AI, Abuarab SF, Hasen E, Munaim AA, Haif SK, Ayyash AM, Khater S, AL-Yasari IH, Kury LTA. Diabetes and Cancer: Metabolic Association, Therapeutic Challenges, and the Role of Natural Products. Molecules 2021; 26:2179. [PMID: 33920079 PMCID: PMC8070467 DOI: 10.3390/molecules26082179] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is considered the second leading cause of death worldwide and in 2018 it was responsible for approximately 9.6 million deaths. Globally, about one in six deaths are caused by cancer. A strong correlation was found between diabetes mellitus and carcinogenesis with the most evident correlation was with type 2 diabetes mellitus (T2DM). Research has proven that elevated blood glucose levels take part in cell proliferation and cancer cell progression. However, limited studies were conducted to evaluate the efficiency of conventional therapies in diabetic cancer patients. In this review, the correlation between cancer and diabetes will be discussed and the mechanisms by which the two diseases interact with each other, as well as the therapeutics challenges in treating patients with diabetes and cancer with possible solutions to overcome these challenges. Natural products targeting both diseases were discussed with detailed mechanisms of action. This review will provide a solid base for researchers and physicians to test natural products as adjuvant alternative therapies to treat cancer in diabetic patients.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Sara Feras. Abuarab
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Eliza Hasen
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Amer A. Munaim
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Shatha Khaled Haif
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Amani Marwan Ayyash
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Samar Khater
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (A.I.M.); (S.F.A.); (E.H.); (A.A.M.); (S.K.H.); (A.M.A.); (S.K.)
| | - Intisar Hadi AL-Yasari
- Department of Genetic Engineering, College of Biotechnology, Al-Qasim Green University, Babylon 00964, Iraq;
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates;
| |
Collapse
|
49
|
Jiang W, Qiao L, Han Y, Zhang A, An H, Xiao J, Ren L. Pancreatic stellate cells regulate branched-chain amino acid metabolism in pancreatic cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:417. [PMID: 33842638 PMCID: PMC8033345 DOI: 10.21037/atm-21-761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is the most lethal malignancy: it has a 5-year survival rate of less than 9%. Although surgical resection is an effective treatment for PDAC, only a small number of patients can have their tumors surgically removed. Thus, an urgent need to find new therapeutic targets for PDAC exists. Understanding the molecular mechanism of PDAC development is essential for the treatment of this malignancy. This research aimed to study the mechanisms of pancreatic stellate cells (PSCs), which regulate branched-chain amino acid (BCAA) metabolism in PDAC. Methods Differentially expressed proteins were detected via nanoliquid chromatography coupled to mass spectrometry (nano-LC-MS/MS). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment methods were used to find the valine-leucine-isoleucine (BCAA) degradation pathway. The levels of BCAAs in the sera and tissues of patients with PDAC were measured by using nuclear magnetic resonance (NMR). The functions of BCAA concentrations and the effects of activated pancreatic stellate cells (aPSCs) were also evaluated by performing Cell Counting Kit-8, colony formation, and wound healing assays. Results A total of 1,519 proteins with significantly differential expression were discovered in PDAC and adjacent tissues by using nano-LC-MS/MS. KEGG pathway enrichment analysis identified the BCAA degradation pathway. The content of BCAA in PDAC clinical samples was up-regulated. However, the addition of different concentrations of BCAA to PDAC cell culture medium failed to promote the proliferation and migration of PDAC cells. Given that analysis based on The Cancer Genome Atlas database showed that the number of aPSCs gradually increased with the progression of PDAC, the effects of aPSCs on PDAC cells were explored. After coculture with aPSCs, PDAC cell proliferation showed a significant increase, and the proteins involved in the BCAA degradation pathway in PDAC cells had also changed. Conclusions aPSCs could regulate BCAA metabolism to enhance the progression of PDAC, indicating that the regulation of BCAA metabolism may serve as a new therapeutic direction for PDAC.
Collapse
Affiliation(s)
- Wenna Jiang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Immune Microenvironment and Disease (Tianjin Medical University), Ministry of Education
| | - Lu Qiao
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yawei Han
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Immune Microenvironment and Disease (Tianjin Medical University), Ministry of Education
| | - Aimin Zhang
- Department of Clinical Laboratory, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Haohua An
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jiawei Xiao
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Li Ren
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
50
|
Suhail Y, Afzal J. Evolved Resistance to Placental Invasion Secondarily Confers Increased Survival in Melanoma Patients. J Clin Med 2021; 10:jcm10040595. [PMID: 33562461 PMCID: PMC7915120 DOI: 10.3390/jcm10040595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/25/2022] Open
Abstract
Mammals exhibit large differences in rates of cancer malignancy, even though the tumor formation rates may be similar. In placental mammals, rates of malignancy correlate with the extent of placental invasion. Our Evolved Levels of Invasibility (ELI) framework links these two phenomena identifying genes that potentially confer resistance in stromal fibroblasts to limit invasion, from trophoblasts in the endometrium, and from disseminating melanoma in the skin. Herein, using patient data from The Cancer Genome Atlas (TCGA), we report that these anti-invasive genes may be crucial in melanoma progression in human patients, and that their loss is correlated with increased cancer spread and lowered survival. Our results suggest that, surprisingly, these anti-invasive genes, which have lower expression in humans compared to species with non-invasive placentation, may potentially prevent stromal invasion, while a further reduction in their levels increases the malignancy and lethality of melanoma. Our work links evolution, comparative biology, and cancer progression across tissues, indicating new avenues for using evolutionary medicine to prognosticate and treat human cancers.
Collapse
Affiliation(s)
- Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA;
- Cancer Systems Biology (CaSB@Yale), Yale West Campus, West Haven, CT 06477, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Junaid Afzal
- Department of Cardiology, University of California, San Francisco, CA 94143, USA;
| |
Collapse
|