1
|
Delle Cave D. Emerging Therapeutic Options in Pancreatic Cancer Management. Int J Mol Sci 2024; 25:1929. [PMID: 38339207 PMCID: PMC10855952 DOI: 10.3390/ijms25031929] [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: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a 5-year survival rate of <8% [...].
Collapse
Affiliation(s)
- Donatella Delle Cave
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, 80131 Naples, Italy
| |
Collapse
|
2
|
Li YM, Jia W, Xin T, Fang YQ. Case report: Heterozygous mutation in HTRA1 causing typical cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy. Front Genet 2023; 14:1235650. [PMID: 37799144 PMCID: PMC10547585 DOI: 10.3389/fgene.2023.1235650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
Background: Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is an autosomal recessive disorder characterized by baldness, recurrent ischemic stroke, lumbago, headache, and dementia which is closely related to homozygous mutations of the high-temperature requirement serine peptidase A1 (HTRA1) gene. Heterozygous mutations of HTRA1 are usually considered to be non-pathogenic. Although it has been revealed that only a few patients with heterozygous mutations could present some manifestations, their clinical symptoms were atypical, milder, and always with a lower frequency of extra-neurological features. Here, a rare patient with heterozygous mutation of HTRA1 who had all typical features of CARASIL as well as severe clinical symptoms and rapid progression was initially reported in our study. Case presentation: A 43-year-old female patient presented with a gradual onset of headache and cognitive decline. As time progressed, her headache intensified and symptoms of dementia began to manifest gradually. During her early years, she had thinning hair and subsequently experienced two occurrences of ischemic strokes in her thirties. Furthermore, she also had a history of lumbago and urinary retention before visiting our hospital. The patient's magnetic resonance imaging revealed the presence of widespread white matter lesions, infarctions, and microbleeds, in addition to lumbar disc herniation and degenerative lesions. The observed clinical characteristics had a strong correlation with CARASIL, and the patient was diagnosed with a heterozygous missense mutation of 905G>A (Arg302Gln) in the HTRA1 gene. The patient has been under continuous follow-up for a duration exceeding 3 years subsequent to her release from the hospital. She underwent cystostomy, and symptoms of bulbar paralysis developed in a progressive way. Currently, there has been a notable decrease in motor function and activities of daily living, resulting in the individual being confined to bed for a duration exceeding 1 year. Conclusion: This case suggests that patients carrying a heterozygous mutation in G905A may also have typical clinical features of CARASIL, which allows us to have a more comprehensive understanding of CARASIL.
Collapse
Affiliation(s)
- Yu-Ming Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Wei Jia
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Neurosurgery, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- Post-Doctoral Scientific Research Station, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yu-Qing Fang
- Post-Doctoral Scientific Research Station, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
3
|
Chuanfen L, Xiaoling W, Wen J, Bingzhen C, Min W. HtrA1L364P leads to cognitive dysfunction and vascular destruction through TGF-β/Smad signaling pathway in CARASIL model mice. Brain Behav 2022; 12:e2691. [PMID: 35841197 PMCID: PMC9392535 DOI: 10.1002/brb3.2691] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
AIMS Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is a life-threatening, inherited, nonhypertensive arteriole disease of the brain. Therapeutic strategy for CARASIL is limited because its pathogenesis is not clear. We previously reported the first family with CARASIL in China, which involves a high-temperature requirement serine protease gene mutation (HtrA1L364P ). Based on this previous study, we constructed a CARASIL mouse model (Mut-hHtrA1L364P mouse, hereinafter referred to as Mut). This paper aimed to systematically study the behavior, pathology, and molecular biology of Mut mice and explore the pathogenesis and possible therapeutic strategies of CARASIL. METHODS Food maze and water maze experiments were used in the behavioral studies. Pathological studies were carried out by arteriole labeling staining and electron microscopy. The mRNA and protein expression levels of the key factors of TGF-β/Smad signaling pathway (TGF-β, Smad2, Smad3, and Smad4) in the brain of the model mice were detected by immunohistochemistry, real-time quantitative polymerase chain reaction (RT-PCR), and Western blot assay. RESULTS The food maze and water maze experiment data showed significant differences between the Mut and wild-type (WT) mice in the first time to find food, the time to contact the escape table for the first time, and the number of times to travel in the escape table quadrant (p < 0.001). The results of vascular labeling staining showed that some small arteries in the brain of Mut mice lost normal structure. The results of electron microscopy showed that the cell morphologies in the cortex and hippocampus of Mut mice were abnormal; the number of synapses was reduced; the walls of capillaries, venules, and arterioles thickened; lumen stenosis and other abnormal phenomenon occurred; and lipofuscin deposition and autophagosomes were found in the hippocampus. Immunohistochemistry, RT-PCR, and Western Blot results showed that the mRNA and protein expression levels of TGF-β, Smad2, and Smad3 in the brain of Mut mice increased to different degrees. CONCLUSIONS The most significant innovation of this study is the first study on the pathogenesis of CARASIL disease using model animals. The Mut mice can well simulate the pathogenesis of CARASIL in behavioral and pathological aspects. The TGF-β/Smad signaling pathway, which is involved in the pathogenesis of CARASIL, is abnormally upregulated in the brain of Mut mice.
Collapse
Affiliation(s)
- Li Chuanfen
- Shandong Normal University, College of Physical Education Sports Human Science LaboratoryJinanShandongChina
| | - Wang Xiaoling
- Neurology DepartmentPLA 960th HospitalJinanShandongChina
| | - Jing Wen
- Shandong Normal University, College of Physical Education Sports Human Science LaboratoryJinanShandongChina
| | - Cao Bingzhen
- Neurology DepartmentPLA 960th HospitalJinanShandongChina
| | - Wang Min
- Shandong Normal University, College of Physical Education Sports Human Science LaboratoryJinanShandongChina
| |
Collapse
|
4
|
Anti-Inflammatory and Antioxidant Effects of Carvacrol on N-Methyl-N′-Nitro-N-Nitrosoguanidine (MNNG) Induced Gastric Carcinogenesis in Wistar Rats. Nutrients 2022; 14:nu14142848. [PMID: 35889805 PMCID: PMC9323991 DOI: 10.3390/nu14142848] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022] Open
Abstract
Carvacrol is a dietary polyphenol from Lamiaceae plants that has been shown to possess a wide range of biological activities including antioxidant and antitumor effects. This study aimed to investigate its anti-inflammatory and antioxidant effects on N-methyl-N′-nitro-N-nitrosoguanidine (MNNG) induced gastric carcinogenesis in Wistar rats. Forty-nine rats were randomly assigned to four treatment and three control groups. Over 60 days, MNNG (200 mg/kg BW) was orally applied to animals of groups 1–5 while the rats in groups 2–5 also received different doses of carvacrol (10, 25, 50, and 100 mg/kg BW, respectively) until the end of the experiment. Group 6 rats were treated with 100 mg/kg BW carvacrol and no MNNG whereas group 7 was the control group without any treatment. After the euthanasia of all rats, the inflammatory cytokines and oxidative stress parameters were assessed in the blood and tissues. The expression of caspase 9, Bax, and Bcl-2 proteins in the stomach tissues were investigated through histopathological examinations. Statistically significant differences were observed in the body weight, oxidative stress, and inflammation parameters of groups 1 to 6 compared to group 7 (p ≤ 0.001). Animals in MNNG groups 2 and 3 treated with the low dose carvacrol (10 and 25 mg/kg BW) showed significantly reduced oxidative stress, inflammation, and apoptotic effect compared to animals of the MNNG groups receiving increased doses of carvacrol (50 and 100 mg/kg BW) or no carvacrol. Rats exposed to MNNG exhibited gastric cancer cells in several areas. In the MNNG group receiving 100 mg/kg BW carvacrol, the inflammatory cell infiltration was observed in gastric mucosal and submucosal areas whereas MNNG rats supplemented with 10 and 25 mg/kg BW carvacrol showed no pathological alterations of the gastric cells. The results of this study indicate that significant antioxidant and anti-inflammatory effects induced by carvacrol at doses of 10 and 25 mg/kg BW interfered with gastric carcinogenesis induced by MNNG in Wistar rats as well as provide hepatoprotection. However, high doses of carvacrol (50 and 100 mg/kg BW) increased oxidative stress, inflammation, and apoptosis.
Collapse
|
5
|
Shang D, Li G, Zhang C, Liu Y. Synergistic Inhibitory Effects of 5-Aza-2'-Deoxycytidine and Cisplatin on Urothelial Carcinoma Growth via Suppressing TGFBI-MAPK Signaling Pathways. Biochem Cell Biol 2021; 100:115-124. [PMID: 34890285 DOI: 10.1139/bcb-2021-0277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study is to reveal the gene transcriptional alteration, possible molecular mechanism, and pathways involved in the synergy of 5-aza-2'-deoxycytidine (DAC) and CDDP in UC. Two UC cell lines, 5637 and T24, were used in the study. A cDNA microarray was carried out to identify critical genes in the synergistic mechanism of both agents against UC cells. The results showed that several key regulatory genes, such as interleukin 24(IL24), fibroblast growth factor 1(FGF1), and transforming growth factor beta-induced (TGFBI), were identified and may play critical roles in the synergy of DAC and CDDP in UC. Pathway enrichment suggested that many carcinogenesis-related pathways, such as ECM-receptor interaction and MAPK signaling pathways, may participate in the synergy of both agents. Our results suggested that TGF-β1 stimulates the phosphorylation levels of ERK1/2 and p38 via increasing TGFBI expression, TGFBI-MAPK signaling pathway plays an important role in the synergy of DAC and CDDP against UC. Therefore, we revealed the synergistic mechanism of DAC and CDDP in UC, several key regulatory genes play critical roles in the synergy of combined treatment, and TGFBI-MAPK signaling pathway may be an important potential target of these two agents.
Collapse
Affiliation(s)
- Donghao Shang
- Capital Medical University, 12517, Department of Urology, Beijing, China;
| | - Gang Li
- Cancer Hospital of China Medical University, 74665, Department of Urology, Shenyang, China;
| | - Caixing Zhang
- Capital Medical University, 12517, Department of Urology, Beijing, China;
| | - Yuting Liu
- Capital Medical University, 12517, Department of Pathology, Beijing, China;
| |
Collapse
|
6
|
Sato R, Imamura K, Semba T, Tomita Y, Saeki S, Ikeda K, Komohara Y, Suzuki M, Sakagami T, Saya H, Arima Y. TGFβ Signaling Activated by Cancer-Associated Fibroblasts Determines the Histological Signature of Lung Adenocarcinoma. Cancer Res 2021; 81:4751-4765. [PMID: 34289987 PMCID: PMC9397619 DOI: 10.1158/0008-5472.can-20-3941] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 05/03/2021] [Accepted: 07/20/2021] [Indexed: 01/07/2023]
Abstract
Invasive lung adenocarcinoma (LADC) can be classified histologically as lepidic, acinar, papillary, micropapillary, or solid. Most LADC tumors manifest several of these histological subtypes, with heterogeneity being related to therapeutic resistance. We report here that in immunodeficient mice, human LADC cells form tumors with distinct histological features, MUC5AC-expressing solid-type or cytokeratin 7 (CK7)-expressing acinar-type tumors, depending on the site of development, and that a solid-to-acinar transition (SAT) could be induced by the tumor microenvironment. The TGFβ-Smad signaling pathway was activated in both tumor and stromal cells of acinar-type tumors. Immortalized cancer-associated fibroblasts (CAF) derived from acinar-type tumors induced SAT in 3D cocultures with LADC cells. Exogenous TGFβ1 or overexpression of an active form of TGFβ1 increased CK7 expression and reduced MUC5AC expression in LADC cells, and knockdown of Tgfb1 mRNA in CAFs attenuated SAT induction. RNA-sequencing analysis suggested that angiogenesis and neutrophil recruitment are associated with SAT in vivo. Our data indicate that CAF-mediated paracrine TGFβ signaling induces remodeling of tumor tissue and determines the histological pattern of LADC, thereby contributing to tumor heterogeneity. SIGNIFICANCE: CAFs secrete TGFβ to induce a solid-to-acinar transition in lung cancer cells, demonstrating how the tumor microenvironment influences histological patterns and tumor heterogeneity in lung adenocarcinoma.
Collapse
Affiliation(s)
- Ryo Sato
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kosuke Imamura
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Semba
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University Hospital, Kumamoto, Japan
| | - Yusuke Tomita
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Sho Saeki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koei Ikeda
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University Hospital, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University Hospital, Kumamoto, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University Hospital, Kumamoto, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yoshimi Arima
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Corresponding Author: Yoshimi Arima, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160–8582, Japan. Phone: 81–3-5363–3983. Fax: 81–3-5363–3983. E-mail:
| |
Collapse
|
7
|
Recent advances in the role of Th17/Treg cells in tumor immunity and tumor therapy. Immunol Res 2021; 69:398-414. [PMID: 34302619 DOI: 10.1007/s12026-021-09211-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 06/30/2021] [Indexed: 12/12/2022]
Abstract
Th17 and Treg cells play an important role in regulating tissue inflammation and maintaining the stability of the immune system. They regulate inflammatory responses, participate in the occurrence and development of autoimmune diseases and tumors, and determine the disease progress. Malignant tumor is one of the diseases with the highest mortality rate in the world. However, the efficacy of traditional treatment is limited, so it is necessary to find safe and efficient treatment methods. Studies have shown that the balance of Th17/Treg cells plays a critical role in tumor progression. In this paper, we review the antitumor and tumor-suppressing effects of Th17/Treg cells, and new strategies for tumor therapy, combined with new research hotspots such as immune checkpoint therapy, miRNA-related gene therapy, and metabolic pathway regulation of Th17/Treg cell differentiation and tumor generation. The synergistic therapy is expected to be widely used in the future clinical practice, providing a new choice for the prevention and treatment of malignant tumors.
Collapse
|
8
|
Kim HJ, Ahn D, Park TI, Jeong JY. TGFBI Expression Predicts the Survival of Patients With Oropharyngeal Squamous Cell Carcinoma. In Vivo 2021; 34:3005-3012. [PMID: 32871844 DOI: 10.21873/invivo.12132] [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: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM This study was conducted to investigate transforming growth factor beta-induced protein (TGFBI) expression and analyze the clinical and prognostic significance of TGFBI in oropharyngeal squamous cell carcinoma (OPSCC). PATIENTS AND METHODS We evaluated TGFBI expression by immunohistochemistry in 94 patients with OPSCC. For comprehensive analysis, TGFBI expression was subdivided into tumor cell score (T), stroma score (S), and the sum of two scores (TS) calculated using H-score. Clinicopathological features and survival outcomes were compared between groups of high expression and low expression of TGFBI in each area. RESULTS Overall, 12 patients (12.8%) showed high T score, and 41 patients (43.6%) revealed high S score. Although T score showed no significant difference both in overall survival (OS) (p=0.080) and recurrence free survival (RFS) (p=0.272), high S score patients had significantly worse OS (p=0.003) and worse RFS (p=0.043). High TS score also showed significant association with worse OS (p=0.011) and worse RFS (p=0.021). High S score was an independent prognostic factor predicting shorter OS (HR=6.352, 95%CI=1.206-40.050, p=0.029) and RFS (HR=18.843, 95%CI=1.030-344.799, p=0.048) in the multivariate analysis. CONCLUSION High S score of TGFBI was a significant predictor of poor prognosis in OPSCC. TGFBI could be a useful new predictive and prognostic biomarker in OPSCC.
Collapse
Affiliation(s)
- Ha-Jeong Kim
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dongbin Ahn
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-In Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea .,Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| |
Collapse
|
9
|
Azimi M, Totonchi M, Rahimi M, Firouzi J, Sahranavard P, Emami Razavi A, Memari F, Kamali F, Ebrahimi M. An integrated analysis to predict micro-RNAs targeting both stemness and metastasis in human gastric cancer. J Gastroenterol Hepatol 2021; 36:436-445. [PMID: 32633423 DOI: 10.1111/jgh.15176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/06/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM Cancer stem cells (CSCs), a subpopulation of tumor cells, assess the capacity of self-renewal, metastasis, and therapeutic resistance. Regulation of CSCs and their epithelial to mesenchymal transition (EMT) potential is one of the promising strategies to eliminate cancer or to inhibit metastasis. Micro-RNAs (miRNAs) as regulators of several cell properties, such as self-renewal, metastasis, and resistance to the drug, could be proper targets in cancer diagnosis and therapy. The aim of the present study is to select common miRNAs targeting both self-renewal and metastasis in gastric cancer. METHODS Stemness-related and EMT-related genes were selected by literature mining. The common miRNAs targeting genes were chosen using different databases and r programming language. The expression pattern of selected miRNAs and genes was evaluated in gastrospheres-as a gastric CSC model-and gastric tumor biopsies. RESULTS Based on the integrated analysis, six miRNAs common to both stemness and metastasis were identified. miR-200c-3p and miR-520c-3p overexpressed in MKN-45 gastrospheres and grade III tumors. In AGS spheres, however, miR-520c-3p and miR-200c-3p upregulation and miR-34a-5p downregulation were similar to grade II tumors. Interestingly, miR-200c-3p and miR-520c-3p indicated a positive correlation with OCT4 and NOTCH1 expression in grade III tumors and MKN-45 spheres. Protein-protein network revealed that the EMT acquisition can be induced by stemness activation through intermediated core-regulatory genes, including CTNNB1, CTNND1, MAML1, KAT2A, and MAML3. CONCLUSION The upregulation of mir-200c-3p and mir-520c-3p could effect on stemness and metastasis in gastric cancer as well as gastric CSCs. Therefore, they can be used as diagnosis and prognostic factors.
Collapse
Affiliation(s)
- Mahnaz Azimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedical Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahsa Rahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Javad Firouzi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Parisa Sahranavard
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amirnader Emami Razavi
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereidoon Memari
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Kamali
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
10
|
Chiavarina B, Costanza B, Ronca R, Blomme A, Rezzola S, Chiodelli P, Giguelay A, Belthier G, Doumont G, Van Simaeys G, Lacroix S, Yokobori T, Erkhem-Ochir B, Balaguer P, Cavailles V, Fabbrizio E, Di Valentin E, Gofflot S, Detry O, Jerusalem G, Goldman S, Delvenne P, Bellahcène A, Pannequin J, Castronovo V, Turtoi A. Metastatic colorectal cancer cells maintain the TGFβ program and use TGFBI to fuel angiogenesis. Theranostics 2021; 11:1626-1640. [PMID: 33408771 PMCID: PMC7778592 DOI: 10.7150/thno.51507] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/04/2020] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) cells are traditionally considered unresponsive to TGFβ due to mutations in the receptors and/or downstream signaling molecules. TGFβ influences CRC cells only indirectly via stromal cells, such as cancer-associated fibroblasts. However, CRC cell ability to directly respond to TGFβ currently remains unexplored. This represents a missed opportunity for diagnostic and therapeutic interventions. Methods: We examined whether cancer cells from primary CRC and liver metastases respond to TGFβ by inducing TGFβ-induced protein ig-h3 (TGFBI) expression, and the contribution of canonical and non-canonical TGFβ signaling pathways to this effect. We then investigated in vitro and in vivo TGFBI impact on metastasis formation and angiogenesis. Using patient serum samples and an orthotopic mouse model of CRC liver metastases we assessed the diagnostic/tumor targeting value of novel antibodies against TGFBI. Results: Metastatic CRC cells, such as circulating tumor cells, directly respond to TGFβ. These cells were characterized by the absence of TGFβ receptor mutations and the frequent presence of p53 mutations. The pro-tumorigenic program orchestrated by TGFβ in CRC cells was mediated through TGFBI, the expression of which was positively regulated by non-canonical TGFβ signaling cascades. TGFBI inhibition was sufficient to significantly reduce liver metastasis formation in vivo. Moreover, TGFBI pro-tumorigenic function was linked to its ability to stimulate angiogenesis. TGFBI levels were higher in serum samples from untreated patients with CRC than in patients who were receiving chemotherapy. A radiolabeled anti-TGFBI antibody selectively targeted metastatic lesions in vivo, underscoring its diagnostic and therapeutic potential. Conclusions: TGFβ signaling in CRC cells directly contributes to their metastatic potential and stromal cell-independence. Proteins downstream of activated TGFβ, such as TGFBI, represent novel diagnostic and therapeutic targets for more specific anti-metastatic therapies.
Collapse
Affiliation(s)
- Barbara Chiavarina
- Cancer Research Institute of Montpellier, Tumor Microenvironment and Resistance to Treatment Laboratory, INSERM U1194, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Institut du Cancer de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Brunella Costanza
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Roberto Ronca
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Arnaud Blomme
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Sara Rezzola
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Paola Chiodelli
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Ambre Giguelay
- Cancer Research Institute of Montpellier, Tumor Microenvironment and Resistance to Treatment Laboratory, INSERM U1194, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Institut du Cancer de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
- Cancer Research Institute of Montpellier, Cancer Bioinformatics and Systems Biology Team, INSERM U1194, Montpellier, France
| | - Guillame Belthier
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut de Génomique Fonctionnelle, Montpellier, France
- Centre National de la Recherche Scientifique, Montpellier, France
| | - Gilles Doumont
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), rue Adrienne Bolland 8, B-6041 Charleroi (Gosselies), Belgium
| | - Gaetan Van Simaeys
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), rue Adrienne Bolland 8, B-6041 Charleroi (Gosselies), Belgium
- Nuclear Medicine department, ULB Hôpital Érasme, route de Lennik 808, B-1070 Brussels, Belgium
| | - Simon Lacroix
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), rue Adrienne Bolland 8, B-6041 Charleroi (Gosselies), Belgium
- Nuclear Medicine department, ULB Hôpital Érasme, route de Lennik 808, B-1070 Brussels, Belgium
| | - Takehiko Yokobori
- Gunma University Initiative for Advanced Research, International Open Laboratory, Universities of Liege and Montpellier Laboratory, Gunma University, Gunma, Japan
| | - Bilguun Erkhem-Ochir
- Gunma University Initiative for Advanced Research, International Open Laboratory, Universities of Liege and Montpellier Laboratory, Gunma University, Gunma, Japan
| | - Patrick Balaguer
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Institut du Cancer de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
- Cancer Research Institute of Montpellier, Hormone Signaling and Cancer Laboratory, Montpellier, France
| | - Vincent Cavailles
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Institut du Cancer de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
- Cancer Research Institute of Montpellier, Hormone Signaling and Cancer Laboratory, Montpellier, France
| | - Eric Fabbrizio
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Institut du Cancer de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
- Cancer Research Institute of Montpellier, Oncogenic Pathways in Cancer Laboratory, INSERM U1194, Montpellier, France
| | | | | | - Olivier Detry
- Department of Abdominal Surgery, University Hospital, University of Liège, Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital, University of Liège, Liège, Belgium
| | - Serge Goldman
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), rue Adrienne Bolland 8, B-6041 Charleroi (Gosselies), Belgium
- Nuclear Medicine department, ULB Hôpital Érasme, route de Lennik 808, B-1070 Brussels, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Julie Pannequin
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut de Génomique Fonctionnelle, Montpellier, France
- Centre National de la Recherche Scientifique, Montpellier, France
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Andrei Turtoi
- Cancer Research Institute of Montpellier, Tumor Microenvironment and Resistance to Treatment Laboratory, INSERM U1194, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Institut du Cancer de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
- Gunma University Initiative for Advanced Research, International Open Laboratory, Universities of Liege and Montpellier Laboratory, Gunma University, Gunma, Japan
| |
Collapse
|
11
|
Cheng LC, Chao YJ, Wang CY, Phan NN, Chen YL, Wang TW, Hsu HP, Lin YJ, Shan YS, Lai MD. Cancer-Derived Transforming Growth Factor-β Modulates Tumor-Associated Macrophages in Ampullary Cancer. Onco Targets Ther 2020; 13:7503-7516. [PMID: 32821120 PMCID: PMC7423398 DOI: 10.2147/ott.s246714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Tumor-associated macrophages (TAMs) originate from monocytes and differentiate into mature macrophages. The interaction between cancer cells and TAMs promotes tumor growth and suppresses immunosurveillance. However, this phenomenon has seldom been observed in ampullary cancer. Patients and Methods TAMs in ampullary cancer were investigated using immunohistochemical (IHC) staining of cancer tissues. Bioinformatic analysis of data from the Gene Expression Omnibus (GEO) database revealed transforming growth factor-beta (TGF-β) signaling in ampullary cancer. The complementary DNA microarray of cancer was compared with adjacent normal duodenum and enzyme-linked immunosorbent assay of serum was used to verify TGF-β signaling in patients. The THP-1 cell line was activated in vitro to imitate M2 TAMs. ClueGo and CluePedia software were operated to simulate TGF-β-related networks in ampullary cancer. Results The IHC study revealed that the majority of TAMs inside ampullary cancer were cluster of differentiation (CD)163+ cells and that the expression of mature CD68+ macrophages was correlated with advanced cancer stage. Bioinformatics analysis revealed that TGF-β and its downstream signaling were significantly upregulated. To verify our bioinformatics-derived predictions, we performed several experiments and demonstrated that increased TGF-β expression was detected in the cDNA microarray. Higher serum levels of TGF-β were correlated with fewer CD68+ and more inducible nitric oxide synthase macrophages in ampullary cancer. Treatment with TGF-β induced modulation of THP-1-derived macrophages. Conclusion The present study demonstrates that TGF-β modulates macrophage activity in ampullary cancer. Targeting TGF-β could be an approach to activating immunosurveillance.
Collapse
Affiliation(s)
- Li-Chin Cheng
- Division of Colorectal Surgery, Department of Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Ying-Jui Chao
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Nam Nhut Phan
- NTT Institute of Hi-Technology, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Yi-Ling Chen
- Senior Citizen Service Management, Chia-Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Tzu-Wen Wang
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yih-Jyh Lin
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
12
|
Sakai A, Ando M, Fukusumi T, Ren S, Liu C, Qualliotine J, Haft S, Sadat S, Saito Y, Guo TW, Xu G, Sasik R, Fisch KM, Gutkind JS, Fertig EJ, Molinolo AA, Califano JA. Aberrant expression of CPSF1 promotes head and neck squamous cell carcinoma via regulating alternative splicing. PLoS One 2020; 15:e0233380. [PMID: 32437477 PMCID: PMC7241804 DOI: 10.1371/journal.pone.0233380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/03/2020] [Indexed: 12/22/2022] Open
Abstract
Alternative mRNA splicing increases protein diversity, and alternative splicing events (ASEs) drive oncogenesis in multiple tumor types. However, the driving alterations that underlie the broad dysregulation of ASEs are incompletely defined. Using head and neck squamous cell carcinoma (HNSCC) as a model, we hypothesized that the genomic alteration of genes associated with the spliceosome may broadly induce ASEs across a broad range of target genes, driving an oncogenic phenotype. We identified 319 spliceosome genes and employed a discovery pipeline to identify 13 candidate spliceosome genes altered in HNSCC using The Cancer Genome Atlas (TCGA) HNSCC data. Phenotypic screens identified amplified and overexpressed CPSF1 as a target gene alteration that was validated in proliferation, colony formation, and apoptosis assays in cell line and xenograft systems as well as in primary HNSCC. We employed knockdown and overexpression assays followed by identification of ASEs regulated by CPSF1 overexpression to identify changes in ASEs, and the expression of these ASEs was validated using RNA from cell line models. Alterations in expression of spliceosome genes, including CPSF1, may contribute to HNSCC by mediating aberrant ASE expression.
Collapse
Affiliation(s)
- Akihiro Sakai
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
- Department of Otolaryngology, Head and Neck Surgery, Tokai University, Isehara, Japan
| | - Mizuo Ando
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
| | - Takahito Fukusumi
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
| | - Shuling Ren
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao Liu
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jesse Qualliotine
- Division of Otolaryngology, Head and Neck Surgery, Department of Surgery, University of California San Diego, San Diego, CA, United States of America
| | - Sunny Haft
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
| | - Sayed Sadat
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
| | - Yuki Saito
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
| | - Theresa W. Guo
- Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Guorong Xu
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, San Diego, CA, United States of America
| | - Roman Sasik
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, San Diego, CA, United States of America
| | - Kathleen M. Fisch
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, San Diego, CA, United States of America
| | - J. Silvio Gutkind
- Department of Pharmacology, University of California San Diego, San Diego, CA, United States of America
| | - Elana J. Fertig
- Division of Oncology Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, Baltimore, MD, United States of America
| | - Alfredo A. Molinolo
- Department of Pathology, University of California San Diego, San Diego, California, United States of America
| | - Joseph A. Califano
- Moores Cancer Center, University of California San Diego, San Diego, California, United States of America
- Division of Otolaryngology, Head and Neck Surgery, Department of Surgery, University of California San Diego, San Diego, CA, United States of America
| |
Collapse
|
13
|
Analysis of hiPSCs differentiation toward hepatocyte-like cells upon extended exposition to oncostatin. Differentiation 2020; 114:36-48. [PMID: 32563741 DOI: 10.1016/j.diff.2020.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
The capability to produce and maintain functional human adult hepatocytes remains one of the major challenges for the use of in-vitro models toward liver cell therapy and industrial drug-screening applications. Among the suggested strategies to solve this issue, the use of human-induced pluripotent stem cells (hiPSCs), differentiated toward hepatocyte-like cells (HLCs) is promising. In this work, we propose a 31-day long protocol, that includes a final 14-day long phase of oncostatin treatment, as opposed to a 7-day treatment which led to the formation of a hepatic tissue functional for CYP1A2, CYP2B6, CYP2C8, CYP2D6, and CYP3A4. The production of albumin, as well as bile acid metabolism and transport, were also detected. Transcriptome profile comparisons and liver transcription factors (TFs) motif dynamics revealed increased expression of typical hepatic markers such as HNF1A and of important metabolic markers like PPARA. The performed analysis has allowed for the extraction of potential targets and pathways which would allow enhanced hepatic maturation in-vitro. From this investigation, NRF1 and SP3 appeared as transcription factors of importance. Complex epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) patterns were also observed during the differentiation process. Moreover, whole transcriptome analysis highlighted a response typical of the one observed in liver regeneration and hepatocyte proliferation. While a complete maturation of hepatocytes was yet to be obtained, the results presented in this work provide new insights into the process of liver development and highlight potential targets aimed to improve in-vitro liver regeneration.
Collapse
|
14
|
Zhang H, Sun X, Lu Y, Wu J, Feng J. DNA-methylated gene markers for colorectal cancer in TCGA database. Exp Ther Med 2020; 19:3042-3050. [PMID: 32256791 PMCID: PMC7086203 DOI: 10.3892/etm.2020.8565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer (CRC) is characterized by the accumulation of genetic and epigenetic alterations in neoplastic processes. DNA methylation, as an important epigenetic process, contributes to the development of CRC. In the present study, the epigenetic landscape of genes in CRC was characterized by analyzing the dataset from The Cancer Genome Atlas database and 177 DNA-methylated genes were screened based on the criterion of the Pearson correlation (R) between expression and methylation levels being >0.4. Pathway enrichment analysis revealed prominent pathways, including transcription and metabolism, further implying their significant role in tumorigenesis. Among the methylated genes, only zinc finger protein (ZNF)726 with aberrant expression was determined to affect overall survival (OS) as well as disease-free survival of patients with CRC. In addition, ZNF726 was identified as an independent prognostic risk factor for OS in patients with CRC. The methylation-based regulation of ZNF726 expression in CRC cells was further assessed using the Cancer Cell Line Encyclopedia database. Finally, the CpG island methylation of the ZNF726 promoter was evaluated to further elucidate its role in the development of CRC. In conclusion, the epigenetic landscape of genes in terms of promoter methylation in CRC was characterized, revealing that aberrant expression of ZNF726 may be an independent prognostic risk factor for OS in patients with CRC.
Collapse
Affiliation(s)
- Hui Zhang
- Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Xun Sun
- Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Ya Lu
- Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Jianzhong Wu
- Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Jifeng Feng
- Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
15
|
Correlation of TNF-α and TGF-β polymorphisms with protein levels in pancreatic ductal adenocarcinoma and colorectal cancer. Contemp Oncol (Pozn) 2019; 23:214-219. [PMID: 31992953 PMCID: PMC6978761 DOI: 10.5114/wo.2019.91537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/22/2019] [Indexed: 01/19/2023] Open
Abstract
Aim of the study To determine the correlation of protein serum levels of two cytokines and their polymorphisms, which have an influence on their expression. Material and methods The study group consisted of 65 patients (33 men, 31 women) who met the criteria for inclusion and exclusion of pancreatic cancer, and 41 patients (25 men, 16 women) with colorectal cancer. The control group consisted of 100 healthy volunteers (63 men, 37 women). Detection of polymorphisms was performed using TaqMan probes, and concentration of proteins by ELISA method. Results The mean TNF-α concentration in patients with colorectal cancer was significantly higher compared to the control group, p< 0.0001. A statistically significant difference was noted when comparing both study groups, p = 0.0009. The analyses show that the occurrence of the polymorphic genotype -308AA of the TNF-α gene was not correlated with the increased concentration of the examined protein in patients with both pancreatic and colorectal cancer. It was also noted that the concentration of TGF-β protein was significantly higher in patients with colorectal cancer than in patients with pancreatic cancer. These results also proved to be statistically significant, p = 0.0353. Conclusions The only statistically significant effects were the correlations between patients belonging to a specific group (pancreatic cancer/colorectal cancer/control) and average protein levels. There was no effect of sex or genotype on the occurrence of elevated levels of TNF-α and TGF-β protein control, despite their variability in particular types of cancer.
Collapse
|
16
|
Nakazawa N, Yokobori T, Kaira K, Turtoi A, Baatar S, Gombodorj N, Handa T, Tsukagoshi M, Ubukata Y, Kimura A, Kogure N, Ogata K, Maeno T, Sohda M, Yajima T, Shimizu K, Mogi A, Kuwano H, Saeki H, Shirabe K. High Stromal TGFBI in Lung Cancer and Intratumoral CD8-Positive T Cells were Associated with Poor Prognosis and Therapeutic Resistance to Immune Checkpoint Inhibitors. Ann Surg Oncol 2019; 27:933-942. [PMID: 31571056 DOI: 10.1245/s10434-019-07878-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND We investigated whether the expression of transforming growth factor-beta-induced protein (TGFBI) and intratumoral immune cells including CD8- and Forkhead box protein P3 (Foxp3)-positive T cells in clinical lung cancer patients could predict the therapeutic response to nivolumab. METHODS Thirty-three patients who were treated with nivolumab were enrolled in this study. Immunohistochemical analyses of TGFBI, PD-L1, CD8, Foxp3, and vimentin expression were conducted. Serum concentrations of TGFBI and transforming growth factor-beta1 (TGF-β1) were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS Cancer TGFBI was not associated with prognosis and therapeutic response to nivolumab, but cancer stromal TGFBI and intratumoral CD8-positive T cells were associated with them. Therefore, we evaluated cancer stromal TGFBI and intratumoral CD8-positive T cells. The high-TGFBI-expression group had poorer clinical responses than did the low-TGFBI-expression group (p < 0.0001). The number of times nivolumab was administered in the high-CD8-expression group was significantly higher than that in the low-CD8-expression group (p = 0.0046). The high-CD8-expression group had better clinical responses than did the low-CD8-expression group (p = 0.0013). Interestingly, all patients in the high-TGFBI/low-CD8-expression group had progressive disease (PD). In contrast, all patients in the low-TGFBI/high-CD8-expression group had PR + SD (partial response + stable disease) by the Response Evaluation Criteria in Solid Tumors (RECIST 1.1). CONCLUSIONS The dual evaluation of stromal TGFBI and intratumoral CD8-positive T cells could be a useful predictive marker for nivolumab.
Collapse
Affiliation(s)
- Nobuhiro Nakazawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan.
| | - Takehiko Yokobori
- Department of Innovative Cancer Immunotherapy, Graduate School of Medicine, Gunma University, Maebashi, Japan.
| | - Kyoichi Kaira
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama University Hospital, Saitama, Japan.
| | - Andrei Turtoi
- Tumor Microenvironment Laboratory, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier, France
| | - Seded Baatar
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Navchaa Gombodorj
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Tadashi Handa
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Mariko Tsukagoshi
- Department of Innovative Cancer Immunotherapy, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Yasunari Ubukata
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Akiharu Kimura
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Norimichi Kogure
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Kyoichi Ogata
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Toshitaka Maeno
- Department of Respiratory Medicine, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Toshiki Yajima
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Kimihiro Shimizu
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Akira Mogi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hiroyuki Kuwano
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hiroshi Saeki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
17
|
Zou J, Huang R, Li H, Wang B, Chen Y, Chen S, Ou K, Wang X. Secreted TGF-beta-induced protein promotes aggressive progression in bladder cancer cells. Cancer Manag Res 2019; 11:6995-7006. [PMID: 31440088 PMCID: PMC6664251 DOI: 10.2147/cmar.s208984] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 07/05/2019] [Indexed: 01/08/2023] Open
Abstract
Background: Transforming growth factor-beta-induced (TGFBI) is an exocrine protein, which has been found to be able to promote the development of nasopharyngeal carcinoma, glioma, pancreatic cancer, and other tumors. However, there is currently no report concerning the relationship between TGFBI and invasive progression of bladder cancer (BCa). Methods: IHC staining, qRT-PCR and Western blot were used to analyze TGFBI and EMT markers levels. In vivo tumorigenesis was performed by xenograft tumor model. Results: In this study, we found that both mRNA and protein levels of TGFBI were significantly up-regulated in muscle invasive bladder cancer (MIBC) tissues compared with non-muscle-invasive bladder cancer (NMIBC) tissues. The high expression level of TGFBI was positively correlated with high histological grade and advanced clinical stage, and BCa patients with high TGFBI levels exhibited poor prognoses. We further confirmed that high expression level of TGFBI can promote proliferation, invasive progression, and epithelial-to-mesenchymal transition (EMT) of BCa cells in vitro, as well as promote tumor growth and EMT in vivo, while silencing of TGFBI inhibited these malignant phenotypes. TGFBI was involved in the up-regulation of EMT by inducing the expression level of Slug, Vimentin, Snail, MMP2, and MMP9 genes, while it down-regulated the expression level of E-cadherin. Moreover, Western blot analysis was carried out to demonstrate that BCa cell lines stably transfected with expression of TGFBI, a secreted protein. Furthermore, conditional medium containing TGFBI protein also resulted in enhanced EMT and malignant phenotype of BCa cells. Conclusion: Our results indicate that high expression level of TGFBI promotes EMT, proliferation, and invasive progression of BCa cells, and TGFBI is a potential therapeutic target and prognostic marker for BCa. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://www.youtube.com/watch?v=GkmU8GAfOv0
Collapse
Affiliation(s)
- Jun Zou
- Department of Emergency Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ruiyan Huang
- Department of Ultrasonography and Electrocardiograms, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Huajun Li
- Department of Emergency Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Bin Wang
- Department of Urology, Affiliated Cancer Hospital & Institue of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yanfei Chen
- Department of Urology, Affiliated Cancer Hospital & Institue of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shuwei Chen
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Kaifu Ou
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xutao Wang
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
18
|
Goehrig D, Nigri J, Samain R, Wu Z, Cappello P, Gabiane G, Zhang X, Zhao Y, Kim IS, Chanal M, Curto R, Hervieu V, de La Fouchardière C, Novelli F, Milani P, Tomasini R, Bousquet C, Bertolino P, Hennino A. Stromal protein βig-h3 reprogrammes tumour microenvironment in pancreatic cancer. Gut 2019; 68:693-707. [PMID: 30415234 PMCID: PMC6580775 DOI: 10.1136/gutjnl-2018-317570] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Pancreatic cancer is associated with an abundant stromal reaction leading to immune escape and tumour growth. This massive stroma drives the immune escape in the tumour. We aimed to study the impact of βig-h3 stromal protein in the modulation of the antitumoural immune response in pancreatic cancer. DESIGN We performed studies with p48-Cre;KrasG12D, pdx1-Cre;KrasG12D;Ink4a/Arffl/fl, pdx1-Cre;KrasG12D; p53R172H mice and tumour tissues from patients with pancreatic ductal adenocarcinoma (PDA). Some transgenic mice were given injections of anti-βig-h3, anti-CD8, anti-PD1 depleting antibodies. Tumour growth as well as modifications in the activation of local immune cells were analysed by flow cytometry, immunohistochemistry and immunofluorescence. Tissue stiffness was measured by atomic force microscopy. RESULTS We identified βig-h3 stromal-derived protein as a key actor of the immune paracrine interaction mechanism that drives pancreatic cancer. We found that βig-h3 is highly produced by cancer-associated fibroblasts in the stroma of human and mouse. This protein acts directly on tumour-specific CD8+ T cells and F4/80 macrophages. Depleting βig-h3 in vivo reduced tumour growth by enhancing the number of activated CD8+ T cell within the tumour and subsequent apoptotic tumour cells. Furthermore, we found that targeting βig-h3 in established lesions released the tissue tension and functionally reprogrammed F4/80 macrophages in the tumour microenvironment. CONCLUSIONS Our data indicate that targeting stromal extracellular matrix protein βig-h3 improves the antitumoural response and consequently reduces tumour weight. Our findings present βig-h3 as a novel immunological target in pancreatic cancer.
Collapse
Affiliation(s)
- Delphine Goehrig
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | | | | | - Zhichong Wu
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Gaëlle Gabiane
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | - Xinyi Zhang
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | - Yajie Zhao
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | - In-San Kim
- KU-KIST School, Korea University, Seongbuk-gu, Korea
| | - Marie Chanal
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | - Roberta Curto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | | | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Pascale Milani
- Ecole Normale Supérieure de Lyon, Lyon, France
- Biomeca, Lyon
| | | | | | - Philippe Bertolino
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| | - Ana Hennino
- Cancer Research Center of Lyon, UMR INSERM 1052, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Centre Léon Bérard, Lyon, France
| |
Collapse
|
19
|
Seok Y, Lee WK, Park JY, Kim DS. TGFBI Promoter Methylation is Associated with Poor Prognosis in Lung Adenocarcinoma Patients. Mol Cells 2019; 42:161-165. [PMID: 30726660 PMCID: PMC6399005 DOI: 10.14348/molcells.2018.0322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/01/2018] [Accepted: 12/10/2018] [Indexed: 11/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide and has high rates of metastasis. Transforming growth factor beta-inducible protein (TGFBI) is an extracellular matrix component involved in tumour growth and metastasis. However, the exact role of TGFBI in NSCLC remains controversial. Gene silencing via DNA methylation of the promoter region is common in lung tumorigenesis and could thus be used for the development of molecular biomarkers. We analysed the methylation status of the TGFBI promoter in 138 NSCLC specimens via methylation-specific PCR and evaluated the correlation between TGFBI methylation and patient survival. TGFBI promoter methylation was detected in 25 (18.1%) of the tumours and was demonstrated to be associated with gene silencing. We observed no statistical correlation between TGFBI methylation and clinicopathological characteristics. Univariate and multivariate analyses showed that TGFBI methylation is significantly associated with poor survival outcomes in adenocarcinoma cases (adjusted hazard ratio = 2.88, 95% confidence interval = 1.19-6.99, P = 0.019), but not in squamous cell cases. Our findings suggest that methylation in the TGFBI promoter may be associated with pathogenesis of NSCLC and can be used as a predictive marker for lung adenocarcinoma prognosis. Further large-scale studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yangki Seok
- Department of Thoracic Surgery, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| | - Won Kee Lee
- Department of Preventive Medicine, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| | - Dong Sun Kim
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu 702-422,
Korea
| |
Collapse
|
20
|
Zhang H, Dong S, Feng J. Epigenetic profiling and mRNA expression reveal candidate genes as biomarkers for colorectal cancer. J Cell Biochem 2019; 120:10767-10776. [PMID: 30672027 DOI: 10.1002/jcb.28368] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/29/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Hui Zhang
- Department of Medical Oncology The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing Jiangsu People's Republic of China
| | - Shuchen Dong
- Department of Medical Oncology The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing Jiangsu People's Republic of China
| | - Jifeng Feng
- Department of Medical Oncology The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing Jiangsu People's Republic of China
| |
Collapse
|
21
|
Chen Y, Xu H, Shi Q, Gu M, Wan X, Chen Q, Wang Z. Hypoxia-inducible factor 1α (HIF-1α) mediates the epithelial-mesenchymal transition in benign prostatic hyperplasia. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:295-304. [PMID: 31933745 PMCID: PMC6944022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 09/27/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) based cancer cell invasion and metastasis has been thoroughly studied in prostate cancer. It was well known that EMT markers which have been found in benign prostatic hyperplasia (BPH) tissues, but system descriptions have not been described. METHODS First, in order to construct the epithelial cells to mesenchymal cell transformation model, BPH-1 cells were cultured with supernatant of prostate matrix normal prostate stromal WPMY-1 cells, after obtaining the culture medium through a filter. After that, we observed the morphology of cells cultured for a period of time by microscopy, detected cell invasion ability by transwell assay, detected cell proliferation ability by MTT, and detected EMT marker expression by western. Finally, we treated the cells with anti-HIF-1α drugs to study their effects on EMT, and then tested several related proteins simultaneously. RESULTS The results showed that the morphology of BPH-1 cells gradually changed to fusiform after cultured with WSCM. At the same time, E-cadherin and cytokeratin levels were significantly lower than those in normal medium. Simultaneous detection of vimentin (SMA) and Snail was positive compared to normal cultured cells. At the same time, the cells were cultured with WSCM and the invasive ability was up-regulated. After treatment with anti-HIF-1α drug, E-cadherin and CK5/8 protein expression was up-regulated, but vimentin, α-SMA, and Snail expression was down-regulated, and in addition, p-Smad3 protein expression was also down-regulated after anti-HIF-1α drug was added. CONCLUSION The above results indicated that WSCM-1 stromal cell supernatant WSCM can induced BPH-1 cell interstitialization, and at the same time, by inducing EMT, secreting HIF-1α activates Smad3 signaling. Our study shows that inhibition of HIF-1α expression provides a new reference for clinical treatment of BPH.
Collapse
|
22
|
Choi D, Montermini L, Kim DK, Meehan B, Roth FP, Rak J. The Impact of Oncogenic EGFRvIII on the Proteome of Extracellular Vesicles Released from Glioblastoma Cells. Mol Cell Proteomics 2018; 17:1948-1964. [PMID: 30006486 DOI: 10.1074/mcp.ra118.000644] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/16/2018] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and heterogeneous form of primary brain tumors, driven by a complex repertoire of oncogenic alterations, including the constitutively active epidermal growth factor receptor (EGFRvIII). EGFRvIII impacts both cell-intrinsic and non-cell autonomous aspects of GBM progression, including cell invasion, angiogenesis and modulation of the tumor microenvironment. This is, at least in part, attributable to the release and intercellular trafficking of extracellular vesicles (EVs), heterogeneous membrane structures containing multiple bioactive macromolecules. Here we analyzed the impact of EGFRvIII on the profile of glioma EVs using isogenic tumor cell lines, in which this oncogene exhibits a strong transforming activity. We observed that EGFRvIII expression alters the expression of EV-regulating genes (vesiculome) and EV properties, including their protein composition. Using mass spectrometry, quantitative proteomic analysis and Gene Ontology terms filters, we observed that EVs released by EGFRvIII-transformed cells were enriched for extracellular exosome and focal adhesion related proteins. Among them, we validated the association of pro-invasive proteins (CD44, BSG, CD151) with EVs of EGFRvIII expressing glioma cells, and downregulation of exosomal markers (CD81 and CD82) relative to EVs of EGFRvIII-negative cells. Nano-flow cytometry revealed that the EV output from individual glioma cell lines was highly heterogeneous, such that only a fraction of vesicles contained specific proteins (including EGFRvIII). Notably, cells expressing EGFRvIII released EVs double positive for CD44/BSG, and these proteins also colocalized in cellular filopodia. We also detected the expression of homophilic adhesion molecules and increased homologous EV uptake by EGFRvIII-positive glioma cells. These results suggest that oncogenic EGFRvIII reprograms the proteome and uptake of GBM-related EVs, a notion with considerable implications for their biological activity and properties relevant for the development of EV-based cancer biomarkers.
Collapse
Affiliation(s)
- Dongsic Choi
- From the ‡Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Laura Montermini
- From the ‡Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Dae-Kyum Kim
- §Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto, Toronto, Ontario, M5S 3E1, Canada.,¶Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Brian Meehan
- From the ‡Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Frederick P Roth
- §Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto, Toronto, Ontario, M5S 3E1, Canada.,¶Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada.,‖Canadian Institute for Advanced Research, Toronto, Ontario, M5G 1M1, Canada
| | - Janusz Rak
- From the ‡Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada;
| |
Collapse
|
23
|
DNMT1 mediated promoter methylation of GNAO1 in hepatoma carcinoma cells. Gene 2018; 665:67-73. [DOI: 10.1016/j.gene.2018.04.080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/20/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
|
24
|
Lee YJ, Lee SB, Beak SK, Han YD, Cho MS, Hur H, Lee KY, Kim NK, Min BS. Temporal changes in immune cell composition and cytokines in response to chemoradiation in rectal cancer. Sci Rep 2018; 8:7565. [PMID: 29765096 PMCID: PMC5953940 DOI: 10.1038/s41598-018-25970-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
We measured systemic changes in the immune response in 92 patients receiving preoperative chemoradiation therapy (CRT) and subsequent surgery for rectal cancer. The peripheral blood was sampled five times from the onset of CRT until surgery. Lymphocytes decreased continuously during CRT but increased after CRT. The increased lymphocyte population was predominantly CD8+ T lymphocytes, which accounted for a significantly larger proportion in patients without residual lymph node metastasis than in those with residual lymph node metastasis. Neutrophils and monocytes decreased during the initial two weeks of CRT but were maintained or increased afterwards. Neutrophil and monocyte counts were significantly lower in patients with a pCR (pathologic complete response) than in those without a pCR two weeks after CRT began but not at the initiation of CRT. All cytokines showed dramatic changes one month after the termination of CRT. Cytokines related to the antitumour immune response increased, and those related to tumour progression decreased. The predictive value of cytokines was not clear. In short, we observed that immune components in peripheral blood are affected by CRT and show dynamic changes over time. We identified biomarker candidates to predict the pathologic response in the future.
Collapse
Affiliation(s)
- Yong Joon Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sat Byol Lee
- Open NBI Convergence Technology Laboratory, Avison Biomedical Research Centre, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suk Kyung Beak
- Open NBI Convergence Technology Laboratory, Avison Biomedical Research Centre, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Dae Han
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Soo Cho
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyuk Hur
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kang Young Lee
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nam Kyu Kim
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Soh Min
- Open NBI Convergence Technology Laboratory, Avison Biomedical Research Centre, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Peng J, Chen W, Chen J, Yuan Y, Zhang J, He Y. Overexpression of chloride channel-3 predicts unfavorable prognosis and promotes cellular invasion in gastric cancer. Cancer Manag Res 2018; 10:1163-1175. [PMID: 29795988 PMCID: PMC5958948 DOI: 10.2147/cmar.s159790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Chloride channel-3 (CLC-3) has been reported to promote the proliferation and invasion in various tumors, yet little is known about its role in gastric cancer. In the present study, we investigated the clinical significance of CLC-3 and its biological role in gastric cancer. METHODS Bioinformatic analysis, immunohistochemical staining, quantitative real-time polymerase chain reaction and Western blot assay were used to assess the expression of CLC-3 and its clinical significance in gastric cancer. The biological role of CLC-3 and its underlying mechanism were detected through in vitro experiments. RESULTS CLC-3 was highly expressed in gastric cancer tissues and cell lines, and high levels of CLC-3 were significantly associated with adverse clinicopathological parameters and shorter overall survival time in patients with gastric cancer. Functional studies revealed that silencing of CLC-3 decreased, while overexpression promoted, the proliferation, migration and invasion of gastric cancer cells in vitro. Mechanistic studies suggested that canonical TGF-β/Smad signaling pathway is involved in CLC-3-induced gastric cancer cells proliferation, migration and invasion. CONCLUSION These findings indicate the vital role of CLC-3 in gastric cancer progression and its potential role of a therapeutic target for treatment.
Collapse
Affiliation(s)
- Jianjun Peng
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianhui Chen
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yujie Yuan
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jian Zhang
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yulong He
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Verrecchia F, Rédini F. Transforming Growth Factor-β Signaling Plays a Pivotal Role in the Interplay Between Osteosarcoma Cells and Their Microenvironment. Front Oncol 2018; 8:133. [PMID: 29761075 PMCID: PMC5937053 DOI: 10.3389/fonc.2018.00133] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
Osteosarcomas are the most frequent form of primary bone tumors and mainly affect children, adolescents, and young adults. Despite encouraging progress in therapeutic management, including the advent of multidrug chemotherapy, the survival rates have remained unchanged for more than four decades: 75% at 5 years for localized disease, but two groups of patients are still at high risk: metastatic at diagnosis (overall survival around 40% at 5 years) and/or poor responders to chemotherapy (20% at 5 years). Because these tumors are classified as “complex genomic,” it is extremely difficult to determine the signaling pathways that might be targeted by specific therapies. A hypothesis has thus emerged, stating that the particular microenvironment of these tumors may interfere with the tumor cells that promote chemoresistance and the dissemination of metastases. The stroma is composed of a large number of cell types (immune cells, endothelial cells, mesenchymal stromal cells, etc.) which secrete growth factors, such as transforming growth factor-β (TGF-β), which favors the development of primary tumors and dissemination of metastases by constituting a permissive niche at primary and distant sites. Rather than targeting the tumor cells themselves, which are very heterogeneous in osteosarcoma, the hypothesis is instead to target the key actors secreted in the microenvironment, such as TGF-βs, which play a part in tumor progression. In the last decade, numerous studies have shown that overexpression of TGF-β is a hallmark of many cancers, including primary bone tumors. In this context, TGF-β signaling has emerged as a crucial factor in the cross talk between tumor cells and stroma cells in poor-prognosis cancers. Secretion of TGF-β by tumor cells or stroma cells can effectively act in a paracrine manner to regulate the phenotype and functions of the microenvironment to stimulate protumorigenic microenvironmental changes. TGF-β can thus exert its protumorigenic function in primary bone tumors by promoting angiogenesis, bone remodeling and cell migration, and by inhibiting immunosurveillance. This review focuses on the involvement of TGF-β signaling in primary bone tumor development, and the related therapeutic options that may be possible for these tumors.
Collapse
Affiliation(s)
- Franck Verrecchia
- UMR1238 INSERM, Université de Nantes, PHY-OS, "Bone Sarcomas and Remodeling of Calcified Tissues", Medical School, Nantes, France
| | - Françoise Rédini
- UMR1238 INSERM, Université de Nantes, PHY-OS, "Bone Sarcomas and Remodeling of Calcified Tissues", Medical School, Nantes, France
| |
Collapse
|
27
|
Zheng J, Mei Y, Xiang P, Zhai G, Zhao N, Xu C, Liu M, Pan Z, Tang K, Jia D. DNA methylation affects metastasis of renal cancer and is associated with TGF-β/RUNX3 inhibition. Cancer Cell Int 2018; 18:56. [PMID: 29651226 PMCID: PMC5894227 DOI: 10.1186/s12935-018-0554-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 04/05/2018] [Indexed: 01/10/2023] Open
Abstract
Background Renal cell carcinoma accounts for 2–3% of all cancers and metastasis increased the malignancy of renal cancer. However, the role of methylation in metastasis of renal cancer is poorly understood. Methods We performed targeted gene array to compare the differential expressions of methylation regulated genes in metastatic and primary renal cancer tissues. Quantitative methylation specific PCR was performed to examine the CpG methylation levels of Runt related transcription factor 3 (RUNX3) and transforming growth factor (TGF)-β. Western blot was performed to detect the expression of target genes. Murine xenograft renal cancer model was established to assay gene expression, methylation level, tumor growth and animal survival in vivo. Results RUNX3 and TGF-β levels were decreased in metastatic renal cancer tissues as a result of their CpG methylation. Metastatic xenograft model displayed decreased expression levels of RUNX3 and TGF-β and higher CpG methylation levels, bigger tumor size and shorter survival time, all which were restored by treatment with a methylation inhibitor. Conclusions Hypermethylation in CpG islands promotes metastasis of renal cancer and is associated with TGF-β and RUNX3 inhibition. Electronic supplementary material The online version of this article (10.1186/s12935-018-0554-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianbo Zheng
- 1Department of Urology, QiLu Hospital of Shandong University, 107 Wenhua Western Road, Jinan, 250012 Shandong Province China.,2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Yanhui Mei
- 3Department of Urology, Binzhou Medical University Hospital, No 2 Huanghe Road, Binzhou, 256603 Shandong Province China
| | - Ping Xiang
- 4Department of Urology, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, 230001 Anhui Province China
| | - Guangsheng Zhai
- 5Department of Radiotherapy, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Ning Zhao
- 2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Chuanbing Xu
- 2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Min Liu
- 2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Zhengsheng Pan
- 2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Kai Tang
- 2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| | - Dongsheng Jia
- 2Department of Urology, Central Hospital of Zibo, No. 54 Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong Province China
| |
Collapse
|
28
|
Fujii R, Jochems C, Tritsch SR, Wong HC, Schlom J, Hodge JW. An IL-15 superagonist/IL-15Rα fusion complex protects and rescues NK cell-cytotoxic function from TGF-β1-mediated immunosuppression. Cancer Immunol Immunother 2018; 67:675-689. [PMID: 29392336 DOI: 10.1007/s00262-018-2121-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells are innate cytotoxic lymphocytes that play a fundamental role in the immunosurveillance of cancers. NK cells of cancer patients exhibit impaired function mediated by immunosuppressive factors released from the tumor microenvironment (TME), such as transforming growth factor (TGF)-β1. An interleukin (IL)-15 superagonist/IL-15 receptor α fusion complex (IL-15SA/IL-15RA; ALT-803) activates the IL-15 receptor on CD8 T cells and NK cells, and has shown significant anti-tumor activity in several in vivo studies. This in vitro study investigated the efficacy of IL-15SA/IL-15RA on TGF-β1-induced suppression of NK cell-cytotoxic function. IL-15SA/IL-15RA inhibited TGF-β1 from decreasing NK cell lysis of four of four tumor cell lines (H460, LNCap, MCF7, MDA-MB-231). IL-15SA/IL-15RA rescued healthy donor and cancer patient NK cell-cytotoxicity, which had previously been suppressed by culture with TGF-β1. TGF-β1 downregulated expression of NK cell-activating markers and cytotoxic granules, such as CD226, NKG2D, NKp30, granzyme B, and perforin. Smad2/3 signaling was responsible for this TGF-β1-induced downregulation of NK cell-activating markers and cytotoxic granules. IL-15SA/IL-15RA blocked Smad2/3-induced transcription, resulting in the rescue of NK cell-cytotoxic function from TGF-β1-induced suppression. These findings suggest that in addition to increasing NK cell function via promoting the IL-15 signaling pathway, IL-15SA/IL-15RA can function as an inhibitor of TGF-β1 signaling, providing a potential remedy for NK cell dysfunction in the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Rika Fujii
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B13, Bethesda, MD, 20892, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B13, Bethesda, MD, 20892, USA
| | - Sarah R Tritsch
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B13, Bethesda, MD, 20892, USA
| | - Hing C Wong
- Altor BioScience Corporation, 2810 North Commerce Parkway, Miramar, FL, 33025, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B13, Bethesda, MD, 20892, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B13, Bethesda, MD, 20892, USA.
| |
Collapse
|
29
|
Szaryńska M, Olejniczak A, Kobiela J, Spychalski P, Kmieć Z. Therapeutic strategies against cancer stem cells in human colorectal cancer. Oncol Lett 2017; 14:7653-7668. [PMID: 29250169 PMCID: PMC5727596 DOI: 10.3892/ol.2017.7261] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third most frequent malignancy and represents the fourth most common cause of cancer-associated mortalities in the world. Despite many advances in the treatment of CRC, the 5-year survival rate of patients with CRC remains unsatisfactory due to tumor recurrence and metastases. Recently, cancer stem cells (CSCs), have been suggested to be responsible for the initiation and relapse of the disease, and have been identified in CRC. Due to their basic biological features, which include self-renewal and pluripotency, CSCs may be novel therapeutic targets for CRC and other cancer types. Conventional therapeutics only act on proliferating and mature cancer cells, while quiescent CSCs survive and often become resistant to chemotherapy. In this review, markers of CRC-CSCs are evaluated and the recently introduced experimental therapies that specifically target these cells by inducing CSC proliferation, differentiation and sensitization to apoptotic signals via molecules including Dickkopf-1, bone morphogenetic protein 4, Kindlin-1, tankyrases, and p21-activated kinase 1, are discussed. In addition, novel strategies aimed at inhibiting some crucial processes engaged in cancer progression regulated by the Wnt, transforming growth factor β and Notch signaling pathways (pyrvinium pamoate, silibinin, PRI-724, P17, and P144 peptides) are also evaluated. Although the metabolic alterations in cancer were first described decades ago, it is only recently that the concept of targeting key regulatory molecules of cell metabolism, such as sirtuin 1 (miR-34a) and AMPK (metformin), has emerged. In conclusion, the discovery of CSCs has resulted in the definition of novel therapeutic targets and the development of novel experimental therapies for CRC. However, further investigations are required in order to apply these novel drugs in human CRC.
Collapse
Affiliation(s)
- Magdalena Szaryńska
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| | - Agata Olejniczak
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| | - Jarosław Kobiela
- Department of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Piotr Spychalski
- Department of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| |
Collapse
|
30
|
Feng Z, Ma J, Hua X. Epigenetic regulation by the menin pathway. Endocr Relat Cancer 2017; 24:T147-T159. [PMID: 28811300 PMCID: PMC5612327 DOI: 10.1530/erc-17-0298] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
There is a trend of increasing prevalence of neuroendocrine tumors (NETs), and the inherited multiple endocrine neoplasia type 1 (MEN1) syndrome serves as a genetic model to investigate how NETs develop and the underlying mechanisms. Menin, encoded by the MEN1 gene, at least partly acts as a scaffold protein by interacting with multiple partners to regulate cellular homeostasis of various endocrine organs. Menin has multiple functions including regulation of several important signaling pathways by controlling gene transcription. Here, we focus on reviewing the recent progress in elucidating the key biochemical role of menin in epigenetic regulation of gene transcription and cell signaling, as well as posttranslational regulation of menin itself. In particular, we will review the progress in studying structural and functional interactions of menin with various histone modifiers and transcription factors such as MLL, PRMT5, SUV39H1 and other transcription factors including c-Myb and JunD. Moreover, the role of menin in regulating cell signaling pathways such as TGF-beta, Wnt and Hedgehog, as well as miRNA biogenesis and processing will be described. Further, the regulation of the MEN1 gene transcription, posttranslational modifications and stability of menin protein will be reviewed. These various modes of regulation by menin as well as regulation of menin by various biological factors broaden the view regarding how menin controls various biological processes in neuroendocrine organ homeostasis.
Collapse
Affiliation(s)
- Zijie Feng
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Ma
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- State Key Laboratory of Veterinary BiotechnologyHarbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xianxin Hua
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
31
|
Chen L, Jin M, Li C, Shang Y, Zhang Q. The tissue distribution and significance of B7-H4 in laryngeal carcinoma. Oncotarget 2017; 8:92227-92239. [PMID: 29190910 PMCID: PMC5696176 DOI: 10.18632/oncotarget.21152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/26/2017] [Indexed: 12/22/2022] Open
Abstract
The costimulatory signals CD28 and B7 have been shown to control tumor invasion and metastasis by regulating T cell activation, whereas the distribution characteristics of B7-associated proteins in laryngeal carcinoma (LC) tissue are still unclear. Here, the expression of members of the B7 superfamily, including B7-H1 (PD-L1), B7-DC (PD-L2) and B7-H4, in fifty-two LC samples was determined by immunohistochemistry, and the relationship between B7-H4 and epithelial-mesenchymal transition (EMT)-associated markers was further assessed by immunofluorescence double staining. Furthermore, the human LC cell lines, Hep-2 and TU212 cells, were further transfected to overexpress B7-H4, and cell invasion and metastasis were analyzed. The results showed that B7-H1, B7-DC and B7-H4 were expressed in the tumor cells, and their expression was restricted to the cell membrane and the cytoplasm. The positive rates of these molecules in the tumor tissues were 57.7% (30/52), 32.7% (17/52) and 34.6% (18/52), respectively. Interestingly, double immunofluorescence staining showed that B7-H4 is coexpression with EMT-related markers, including p-Smad2/3, Snail and Vimentin, in carcinoma cells. Moreover, overexpression of B7-H4 in Hep-2 cells promotes the expression of pSmad2/3 and Snail by activating AKT-STAT3 signaling. Transwell and wound-healing assays demonstrated that B7-H4 enhanced both Hep-2 and TU212 cell invasion and metastasis. Our results suggest that B7-H4 transmits feedback signaling to tumor cells and promotes invasion and metastasis by promoting EMT progression. Therefore, blocking B7-H4 signaling might be a novel treatment strategy for LC.
Collapse
Affiliation(s)
- Lili Chen
- Medical College, Dalian University, Dalian, People's Republic of China.,Department of Clinical Laboratory, Laiwu City People Hospital, Laiwu, People's Republic of China
| | - Meihua Jin
- Medical College, Dalian University, Dalian, People's Republic of China
| | - Chunshi Li
- Medical College, Dalian University, Dalian, People's Republic of China.,School of Pharmacy, Yanbian University, Yanji, People's Republic of China
| | - Yongjun Shang
- Medical College, Dalian University, Dalian, People's Republic of China.,Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng, People's Republic of China
| | - Qinggao Zhang
- Medical College, Dalian University, Dalian, People's Republic of China
| |
Collapse
|
32
|
Immune Components in Human Milk Are Associated with Early Infant Immunological Health Outcomes: A Prospective Three-Country Analysis. Nutrients 2017; 9:nu9060532. [PMID: 28538696 PMCID: PMC5490511 DOI: 10.3390/nu9060532] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 01/22/2023] Open
Abstract
The role of breastfeeding in improving allergy outcomes in early childhood is still unclear. Evidence suggests that immune mediators in human milk (HM) play a critical role in infant immune maturation as well as protection against atopy/allergy development. We investigated relationships between levels of immune mediators in colostrum and mature milk and infant outcomes in the first year of life. In a large prospective study of 398 pregnant/lactating women in the United Kingdom, Russia and Italy, colostrum and mature human milk (HM) samples were analysed for immune active molecules. Statistical analyses used models adjusting for the site of collection, colostrum collection time, parity and maternal atopic status. Preliminary univariate analysis showed detectable interleukin (IL) 2 and IL13 in HM to be associated with less eczema. This finding was further confirmed in multivariate analysis, with detectable HM IL13 showing protective effect OR 0.18 (95% CI 0.04-0.92). In contrast, a higher risk of eczema was associated with higher HM concentrations of transforming growth factor β (TGFβ) 2 OR 1.04 (95% CI 1.01-1.06) per ng/mL. Parental-reported food allergy was reported less often when IL13 was detectable in colostrum OR 0.10 (95% CI 0.01-0.83). HM hepatocyte growth factor (HGF) was protective for common cold incidence at 12 months OR 0.19 (95% CI 0.04-0.92) per ng/mL. Data from this study suggests that differences in the individual immune composition of HM may have an influence on early life infant health outcomes. Increased TGFβ2 levels in HM are associated with a higher incidence of reported eczema, with detectable IL13 in colostrum showing protective effects for food allergy and sensitization. HGF shows some protective effect on common cold incidence at one year of age. Future studies should be focused on maternal genotype, human milk microbiome and diet influence on human milk immune composition and both short- and long-term health outcomes in the infant.
Collapse
|