1
|
Mansoori B, Kiani S, Mezajin AA, Zandi P, Banaie H, Rostamzadeh D, Cho WC, Duijf PHG, Mansoori B, Baradaran B. MicroRNA-143-5p Suppresses ER-Positive Breast Cancer Development by Targeting Oncogenic HMGA2. Clin Breast Cancer 2023; 23:e480-e490.e3. [PMID: 37596147 DOI: 10.1016/j.clbc.2023.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/22/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND About 70%-80% of breast cancers (BCs) express estrogen receptors (ER-positive). MicroRNAs (miRNAs) are a group of small endogenous noncoding RNAs that play a critical regulatory role in cancer development and progression, including in BC. MiRNA deficiency promotes the development of BCs. MiR-143-5p is one of the most commonly dysregulated miRNAs in BC but its role as a tumor suppressor remains unclear. MATERIALS AND METHODS MiR-143-3p and -5p expression in breast tissue was analyzed using TCGA and StarBase databases. Expression in BC subclasses and survival analyses were conducted. Clinical samples were collected, cell cultures created, and gene expression assays performed following previous studies. Protein expression, luciferase reporter, wound healing, DAPI staining, cell cycle, colony formation, spheroid, CD44 FACS, and proliferation assays were conducted following various protocols. RESULTS Here, we find that both miR-143-3p and miR-143-5p levels are considerably lower in BC tissue compared to normal breast tissue and low miR-143 expression predicts poor prognosis in ER+ BC patients. In-depth analyses identified 3 miR-143-5p binding sites in the 3' untranslated region (UTR) of the DNA binding protein High Mobility Group AT-Hook 2 (HMGA2). Luciferase reporter assays using wild-type and mutant HMGA2 3'UTR sequences and Western blot analyses demonstrated that HMGA2 is a direct and bona fide miR-143-5p target in BC cells. In addition, we show that restoration of miR-143-5p expression suppresses metastasis-related features of ER+ BC cells, including reduced tumor cell migration, increased E-cadherin expression, and decreased vimentin and N-cadherin expression. Furthermore, miR-143-5p reduces cell proliferation, cell cycle entry, and stemness, while promoting apoptosis moderately. Finally, patient sample pathway analyses demonstrated that these mechanisms are also active in BC. CONCLUSIONS Altogether, our findings shed new light on miR-143-5p's anticancer biological functions in BC progression by directly targeting HMGA2. This suggests that restoration of miR-143-5p could be a promising new therapeutic approach for the treatment of ER+ BC.
Collapse
Affiliation(s)
- Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran; Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shiva Kiani
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | - Homadokht Banaie
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Khuzestan, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran; Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Pascal H G Duijf
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane QLD, Australia; Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane QLD, Australia; Centre for Data Science, Queensland University of Technology, Brisbane QLD, Australia; Cancer and Aging Research Program, Queensland University of Technology, Brisbane QLD, Australia; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Behzad Mansoori
- The Wistar Institute, Molecular and Cellular Oncogenesis Program, Philadelphia, PA.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran.
| |
Collapse
|
2
|
Heiat M, Rezaei E, Gharechahi J, Abbasi M, Behroozi J, Abyazi MA, Baradaran B. Knockdown of SIX4 inhibits pancreatic cancer cells via apoptosis induction. Med Oncol 2023; 40:287. [PMID: 37656231 DOI: 10.1007/s12032-023-02163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Sine oculis homeobox 4 (SIX4), a critical transcription factor modulating organ development, potentially participates in tumorigenesis through numerous pathways. Here, we investigated siRNA-mediated knockdown effects of SIX4 on pancreatic cancer cells and underlying molecular mechanisms. The expression of SIX4 in pancreatic cancer and adjacent tissues were investigated in clinical tissue samples and bioinformatically approved by gene expression omnibus (GEO) database. Appropriate siRNA transfected into PANC1 pancreatic cancer cells in order to SIX4 knockdown. The survival, migration, invasion, colony formation, mitochondrial membrane potential, apoptosis, autophagy, and cell cycle in the cancer cells were investigated after knockdown of SIX4. In addition, expression of genes involved in apoptosis and metastasis were assessed in the transfected cancer cells in mRNA and protein levels. High-throughput analysis using GEO database confirmed the overexpression of SIX4 in pancreatic cancer tissues by six independent pancreatic cancer microarrays. Knockdown of SIX4 by specific siRNA significantly decreased survival, colony formation, and mitochondrial membrane potential of the cancer cells. Further assessments demonstrated that knockdown of SIX4 increases the apoptosis and autophagy rates in the cancer cells through modifying the expression of related genes. Moreover, a significant decrease in migration and invasion rates were observed in SIX4 suppressed group. Furthermore, frequency of the cells transfected with SIX4 siRNA increased slightly in G1 and Sub-G1 phases of cell cycle. Our study suggested that siRNA-mediated knockdown of SIX4 increases the pancreatic cancer cells death and reduces the invasion and migration of the cancer cells through different molecular pathways.
Collapse
Affiliation(s)
- Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ehsan Rezaei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Javad Gharechahi
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoumeh Abbasi
- Department of Microbiology, Malekan Branch, Islamic Azad University, Malekan, Iran
| | - Javad Behroozi
- Department of Genetics and Biotechnology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Abyazi
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, 5166614766, Tabriz, Iran.
| |
Collapse
|
3
|
Hindi N, Carrillo-García J, Blanco-Alcaina E, Renshaw M, Luna P, Durán J, Jiménez N, Sancho P, Ramos R, Moura DS, Martín-Broto J. Platinum-Based Regimens Are Active in Advanced Pediatric-Type Rhabdomyosarcoma in Adults and Depending on HMGB1 Expression. Int J Mol Sci 2023; 24:ijms24010856. [PMID: 36614297 PMCID: PMC9821763 DOI: 10.3390/ijms24010856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
Rhabdomyosarcoma (RMS) in adults is a rare and aggressive disease, which lacks standard therapies for relapsed or advanced disease. This retrospective study aimed to describe the activity of BOMP-EPI (bleomycin, vincristine, methotrexate and cisplatin alternating with etoposide, cisplatin and ifosfamide), an alternative platinum-based regimen, in adult patients with relapsed/metastatic RMS. In the study, 10 patients with RMS with a median age at diagnosis of 20.8 years and a female/male distribution of 6/4 received a mean of 2.5 cycles of BOMP-EPI. The best RECIST response was a complete response in 1/10 (10%) patients, a partial response in 5/10 (50%), stable disease in 3/10 (30%) and progression in 1/10 (10%). With a median follow-up in the alive patients from the start of therapy of 30.5 months (15.7-258), all patients progressed with a median progression-free survival of 8.47 months (95% CI 8.1-8.8), and 7/10 patients died with a median overall survival of 24.7 months (95% CI 13.7-35.6). BOMP-EPI was an active chemotherapy regimen in adults with pediatric-type metastatic RMS, with outcomes in terms of survival that seem superior to what was expected for this poor-prognosis population. Low HMGB1 expression level was identified as a predictive factor of better response to this treatment.
Collapse
Affiliation(s)
- Nadia Hindi
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
- Medical Oncology Department, University Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Jaime Carrillo-García
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
| | - Elena Blanco-Alcaina
- Institute of Biomedicine of Seville (IBIS), HUVR-CSIC-University of Seville, 41013 Seville, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Renshaw
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
| | - Pablo Luna
- Medical Oncology Department, University Hospital Son Espases, 07210 Palma, Spain
| | - José Durán
- Medical Oncology Department, University Hospital Son Espases, 07210 Palma, Spain
| | - Natalia Jiménez
- Medical Oncology Department, San Vicente de Paúl Hospital, Heredia 40101, Costa Rica
| | - Pilar Sancho
- Medical Oncology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, 07210 Palma, Spain
| | - David S. Moura
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
| | - Javier Martín-Broto
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
- Medical Oncology Department, University Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-910-908-102 (ext. 52831)
| |
Collapse
|
4
|
The Transcription Factor Otc4A Stimulates the Proliferation, Invasion, and Stemness of Colorectal Cancer Cells by Inhibiting the Regulation of miR-7-5p on TLR4. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7856629. [PMID: 36199554 PMCID: PMC9529417 DOI: 10.1155/2022/7856629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/19/2022]
Abstract
Background To investigate the effects and mechanism of octamer-binding transcription factor 4 (Otc4A) on proliferation, invasion, and stemness of colorectal cancer (CRC) cells. Methods Firstly, normal fetal human cells (FHC, colon epithelial cells) and HT29 cells (CRC cells) were cultured. The expression levels of Otc4A, miR-7-5p, and TLR4 in cells were then detected by qRT-PCR. CCK-8 was adopted to measure cell proliferation rate after Otc4A, miR-7-5p, and TLR4, respectively, were either knocked out or overexpressed in HT29 cells. Later, the cell viability was detected by cell cloning assay; cell invasion by transwell; cell sphere-forming ability by sphere-formation assay; protein expression level of Otc4A, p65, p-p65, and TLR4 by western blot; and the targeting relationships between miR-7-5p and Otc4A as well as miR-7-5p and TLR4 by dual-luciferase reporter assay. Finally, chromatin immunoprecipitation was applied to verify the interaction between Otc4A and miR-7-5p. Results In HT29 cells, Otc4A expression was significantly increased. Additionally, the knockdown of Otc4A prevented HT29 cells from proliferating, migrating, forming spheres, and activating NF–B signaling. Otc4A could negatively regulate miR-7-5p, and miR-7-5p could target TLR4 expression. Besides, a negative correlation was found between Otc4A and miR-7-5p. Finally, the knockdown of miR-7-5p or overexpression of TLR4 could significantly reverse the effect of the knockdown of Otc4A on HT29 cells. Conclusion The transcription factor Otc4A can regulate the level of TLR4 by inhibiting the expression of miR-7-5p and then promote the proliferation and invasion of CRC cell HT29 as well as enhance cell stemness.
Collapse
|
5
|
Wang L, Weng W, Yang S, Yang S, Du R. Circle RNA circ_0007331 promotes colorectal carcinoma by targeting miR-205-5p/high-mobility group A2 axis. Bioengineered 2022; 13:9312-9321. [PMID: 35400282 PMCID: PMC9161998 DOI: 10.1080/21655979.2022.2051857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a common malignancy of the gastrointestinal tract. CircRNAs have been reported to play regulatory roles in many cancers, including CRC. This study focuses on the role of circ_0007331 in CRC. Differentially expressed circRNAs in CRC were screened using the GEO database. RT-qPCR was used to analyze mRNA expression. StarBase and TargetScan were used to predict targeting relationships and then verified by the dual luciferase reporter assay along with the RNA pull-down assay. CCK-8 as well and transwell assays were used to measure cell viability, migration, and invasion. Protein levels were determined using western blotting. circ_0007331 is expressed more frequently in patients with CRC. The inhibition of circ_0007331 expression reduced the viability, colony formation, migration, and invasion of CRC cells. However, inhibition of miR-205-5p or elevation of high-mobility group A2 (HMGA2) can reverse the function of inhibited circ_0007331 in tumor cells. This study demonstrated that the circ_0007331/miR-205-5p/HMGA2 axis promotes CRC development. Thus, circ_0007331 may be a potential biomarker for CRC.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Pathology, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| | - Weiming Weng
- Department of Gastrointestinal Surgery, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| | - Shuhui Yang
- Department of Pathology, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| | - Shasha Yang
- Department of Pathology, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| | - Richang Du
- Department of Pathology, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| |
Collapse
|
6
|
Cui J, Dean D, Hornicek FJ, Yi G, Duan Z. Expression and Clinical Significance of High-Mobility Group AT-hook 2 (HMGA2) in Osteosarcoma. Orthop Surg 2022; 14:955-966. [PMID: 35388973 PMCID: PMC9087380 DOI: 10.1111/os.13167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 09/27/2021] [Accepted: 10/09/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Although high‐mobility group AT‐hook 2 (HMGA2) has been shown to have crucial roles in the pathogenesis and metastasis of various malignancies, its expression and significance in osteosarcoma remain unknown. Here we evaluate the expression, clinical prognostic value, and overall function of HMGA2 in osteosarcoma. Methods Sixty‐nine osteosarcoma patient specimens within a tissue microarray (TMA) were analyzed by immunohistochemistry for HMGA2 expression. Demographics and clinicopathological information including age, gender, tumor location, metastasis, recurrence, chemotherapy response, follow‐up time, and disease status were also collected. After validation of expression, we determined whether there was a correlation between HMGA2 expression and patient clinicopathology. HMGA2 expression was also evaluated in osteosarcoma cell lines and patient tissues by Western blot, we analyzed the expression of HMGA2 in the human osteosarcoma cell lines MG63, 143B, U2OS, Saos‐2, MNNG/HOS, and KHOS. HMGA2‐specific siRNA and clonogenic assays were then used to determine the effect of HMGA2 inhibition on osteosarcoma cell proliferation, growth, and chemosensitivity. Results HMGA2 expression was elevated in the osteosarcoma patient specimens and human osteosarcoma cell lines. HMGA2 was differentially expressed in human osteosarcoma cell lines. Specifically, a relatively high expression of HMGA2 was present in KHOS, MNNG/HOS, 143B and a relatively low expression was in MG63, U2OS as well as Saos‐2. HMGA2 expression is correlated with metastasis and shorter overall survival. High HMGA2 expression is an independent predictor of poor osteosarcoma prognosis. There was no significant correlation between HMGA2 expression and the age, gender, or tumor site of the patient. HMGA2 expression is predominantly within the nucleus. The expression of HMGA2 also directly correlated to neoadjuvant chemoresistance. There was a significant reduction of HMGA2 expression in the siRNA transfection group. After the use of siRNA, the proliferation of osteosarcoma cells is decreased and the chemosensitivity of osteosarcoma cells is significantly increased. Conclusion Our study supports HMGA2 as a potential prognostic biomarker and therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Juncheng Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Dylan Dean
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Guoliang Yi
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
7
|
miR-142-3p simultaneously targets HMGA1, HMGA2, HMGB1, and HMGB3 and inhibits tumorigenic properties and in-vivo metastatic potential of human cervical cancer cells. Life Sci 2021; 291:120268. [PMID: 34973275 DOI: 10.1016/j.lfs.2021.120268] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
AIMS High-mobility group (HMG) proteins are oncogenic in different cancers, including cervical cancer; silencing their individual expression using sh-RNAs, siRNAs, and miRNAs has had anti-tumorigenic effects, but the consequences of their collective downregulation are not known. Since multiple gene targeting is generally very effective in cancer therapy, the present study highlighted the consequences of silencing the expression of HMGA1, A2, B1, and B3 using sh-RNAs or miR-142-3p (that can potentially target HMGA1, A2, B1, and B3) in cervical cancer cell lines. MAIN METHODS 3' UTR luciferase reporter assays were performed to validate HMGA1, A2, B1, and B3 as targets of miR-142-3p in human cervical cancer cells. Annexin V/PI dual staining and flow cytometry analyses were used to detect apoptotic cells. miR-142-3p-mediated regulation of cell death, colony formation, migration, and invasion was investigated in human cervical cancer cells together with in vivo metastasis in zebrafish. KEY FINDINGS Concurrent knockdown of HMGA1, A2, B1, and B3 through their corresponding sh-RNAs inhibited cell viability and colony formation but induced apoptosis, and these effects were relatively reduced upon their individual knockdown. miR-142-3p targeted HMGA1, A2, B1, and B3 by binding to their 3'UTRs and induced apoptosis but inhibited proliferation, migration, and invasion of human cervical cancer cells. In addition, miR-142-3p expression decreased phospho-p65 and EMT-related proteins in cervical cancer cells and their in vivo metastatic potential upon implantation in zebrafish. SIGNIFICANCE These findings suggest that miR-142-3p acts as a tumor-suppressive miRNA by targeting HMGA1, A2, B1, and B3 and may serve as a potential therapeutic agent in human cervical cancer.
Collapse
|
8
|
Mansoori B, Terp MG, Mohammadi A, Pedersen CB, Ditzel HJ, Baradaran B, Gjerstorff MF. HMGA2 Supports Cancer Hallmarks in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:5197. [PMID: 34680349 PMCID: PMC8533747 DOI: 10.3390/cancers13205197] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that exhibits a high proliferation rate and early metastasis leading to a poor prognosis. HMGA2 is a DNA binding transcriptional regulator implicated in tumorigenesis. Here, we demonstrate that the HMGA2 promoter is demethylated in TNBC tumors, leading to increased expression of HMGA2 at both mRNA and protein levels. Importantly, high HMGA2 levels in TNBC tumors are correlated with poor prognosis. To detail the role of HMGA2 in TNBC development and progression, we studied its effect on core cancer phenotypes. Stable knockdown of HMGA2 in TNBC cells revealed that HMGA2 may support cell proliferation, cell migration and invasion. In addition, HMGA2 knockdown decreased cancer stem cell (CSC) features. Importantly, we found that silencing HMGA2 inhibited NF-kB signaling and lead to decreased expression of the downstream molecules IL-6 and IL-8 and reduced STAT3 pathway activation. Our results demonstrate that HMGA2 supports cancer hallmarks in TNBC and may represent a promising target for TNBC treatment.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Golghasht St., Tabriz 51666-14731, Iran; (B.M.); (B.B.)
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
- Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Golghasht St., Tabriz 51656-65811, Iran
| | - Mikkel Green Terp
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
| | - Christina Bøg Pedersen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
| | - Henrik Jørn Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
- Department of Oncology, Odense University Hospital, DK-5000 Odense C, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, DK-5000 Odense C, Denmark
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Golghasht St., Tabriz 51666-14731, Iran; (B.M.); (B.B.)
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 3, DK-5000 Odense C, Denmark; (M.G.T.); (A.M.); (C.B.P.); (H.J.D.)
- Department of Oncology, Odense University Hospital, DK-5000 Odense C, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, DK-5000 Odense C, Denmark
| |
Collapse
|
9
|
Expression profiles of miR-196, miR-132, miR-146a, and miR-134 in human colorectal cancer tissues in accordance with their clinical significance : Comparison regarding KRAS mutation. Wien Klin Wochenschr 2021; 133:1162-1170. [PMID: 34463887 DOI: 10.1007/s00508-021-01933-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/25/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most widespread malignancies in the world. MicroRNA (miRNA) has been identified as an important modulator of the biological processes of the cells. This group of noncoding RNAs also has a pivotal function in the growth and development of human cancers, including CRC. Among these miRNAs, miR-196, miR-132, miR-146a, and miR-134 have fundamental impacts on the regulation of cancers. The current study aimed to investigate the involvement of these miRNAs in CRC patients. METHODS In this study, 50 pairs of tumor and tumor margin samples of CRC patients were investigated to assess the expression levels of miR-196, miR-132, miR-146a, and miR-134 in this cancer. For this purpose, firstly, quantitative real-time PCR (qRT-PCR) was applied. Also, KRAS mutation and clinicopathological characteristics of the CRC patients were analyzed in the study groups. RESULTS The findings demonstrated the overexpression of miR-196 (P-value = 0.0045) and miR-146a (P-value = 0.0033) in tumor tissues compared to controls. Conversely, the expression levels of miR-132 (P-value = 0.00032) and miR-134 (P-value < 0.0001) were downregulated in tumor tissues. Also, miR-146a was the only miRNA with significant expression change in the case of the KRAS gene mutation. Interestingly, the expression ratio of these miRNAs was significantly associated with some of the clinicopathological features of the patients, such as lymph node and distant metastases. CONCLUSION Our data demonstrated that these miRNAs appear to be promising novel biomarkers for early diagnosis of CRC and may pave the way for the future establishment of novel therapeutic options for CRC.
Collapse
|
10
|
Divisato G, Piscitelli S, Elia M, Cascone E, Parisi S. MicroRNAs and Stem-like Properties: The Complex Regulation Underlying Stemness Maintenance and Cancer Development. Biomolecules 2021; 11:biom11081074. [PMID: 34439740 PMCID: PMC8393604 DOI: 10.3390/biom11081074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Embryonic stem cells (ESCs) have the extraordinary properties to indefinitely proliferate and self-renew in culture to produce different cell progeny through differentiation. This latter process recapitulates embryonic development and requires rounds of the epithelial-mesenchymal transition (EMT). EMT is characterized by the loss of the epithelial features and the acquisition of the typical phenotype of the mesenchymal cells. In pathological conditions, EMT can confer stemness or stem-like phenotypes, playing a role in the tumorigenic process. Cancer stem cells (CSCs) represent a subpopulation, found in the tumor tissues, with stem-like properties such as uncontrolled proliferation, self-renewal, and ability to differentiate into different cell types. ESCs and CSCs share numerous features (pluripotency, self-renewal, expression of stemness genes, and acquisition of epithelial-mesenchymal features), and most of them are under the control of microRNAs (miRNAs). These small molecules have relevant roles during both embryogenesis and cancer development. The aim of this review was to recapitulate molecular mechanisms shared by ESCs and CSCs, with a special focus on the recently identified classes of microRNAs (noncanonical miRNAs, mirtrons, isomiRs, and competitive endogenous miRNAs) and their complex functions during embryogenesis and cancer development.
Collapse
|
11
|
Abedi Gaballu F, Cho WCS, Dehghan G, Zarebkohan A, Baradaran B, Mansoori B, Abbaspour-Ravasjani S, Mohammadi A, Sheibani N, Aghanejad A, Ezzati Nazhad Dolatabadi J. Silencing of HMGA2 by siRNA Loaded Methotrexate Functionalized Polyamidoamine Dendrimer for Human Breast Cancer Cell Therapy. Genes (Basel) 2021; 12:genes12071102. [PMID: 34356120 PMCID: PMC8303903 DOI: 10.3390/genes12071102] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
The transcription factor high mobility group protein A2 (HMGA2) plays an important role in the pathogenesis of some cancers including breast cancer. Polyamidoamine dendrimer generation 4 is a kind of highly branched polymeric nanoparticle with surface charge and highest density peripheral groups that allow ligands or therapeutic agents to attach it, thereby facilitating target delivery. Here, methotrexate (MTX)- modified polyamidoamine dendrimer generation 4 (G4) (G4/MTX) was generated to deliver specific small interface RNA (siRNA) for suppressing HMGA2 expression and the consequent effects on folate receptor (FR) expressing human breast cancer cell lines (MCF-7, MDA-MB-231). We observed that HMGA2 siRNA was electrostatically adsorbed on the surface of the G4/MTX nanocarrier for constructing a G4/MTX-siRNA nano-complex which was verified by changing the final particle size and zeta potential. The release of MTX and siRNA from synthesized nanocomplexes was found in a time- and pH-dependent manner. We know that MTX targets FR. Interestingly, G4/MTX-siRNA demonstrates significant cellular internalization and gene silencing efficacy when compared to the control. Besides, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay demonstrated selective cell cytotoxicity depending on the folate receptor expressing in a dose-dependent manner. The gene silencing and protein downregulation of HMGA2 by G4/MTX-siRNA was observed and could significantly induce cell apoptosis in MCF-7 and MDA-MB-231 cancer cells compared to the control group. Based on the findings, we suggest that the newly developed G4/MTX-siRNA nano-complex may be a promising strategy to increase apoptosis induction through HMGA2 suppression as a therapeutic target in human breast cancer.
Collapse
Affiliation(s)
- Fereydoon Abedi Gaballu
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran; (F.A.G.); (B.B.); (B.M.)
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | | | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
- Correspondence: or (G.D.); (J.E.N.D.); Tel.: +98-33392739 (G.D.); +98-41-33367914 (J.E.N.D.); Fax: +98-33356027 (G.D.); +98-41-33367929 (J.E.N.D.)
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran;
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran; (F.A.G.); (B.B.); (B.M.)
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran; (F.A.G.); (B.B.); (B.M.)
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark;
| | | | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark;
| | - Nader Sheibani
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA;
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz 51666-16471, Iran;
| | - Jafar Ezzati Nazhad Dolatabadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran;
- Correspondence: or (G.D.); (J.E.N.D.); Tel.: +98-33392739 (G.D.); +98-41-33367914 (J.E.N.D.); Fax: +98-33356027 (G.D.); +98-41-33367929 (J.E.N.D.)
| |
Collapse
|
12
|
Mansoori B, Najafi S, Mohammadi A, AsadollahSeraj H, Savadi P, Mansoori B, Nazari A, Mokhtarzadeh A, Roshani E, Duijf PH, Cho WCS, Baradaran B. The synergy between miR-486-5p and tamoxifen causes profound cell death of tamoxifen-resistant breast cancer cells. Biomed Pharmacother 2021; 141:111925. [PMID: 34323695 DOI: 10.1016/j.biopha.2021.111925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 11/24/2022] Open
Abstract
Breast cancer (BC) is the most common type of malignancy in women. A subset of breast cancers show resistance to endocrine-based therapies. The estrogen receptor (ER) plays a critical role in developing hormone-dependent BC. Loss of ER contributes to resistance to tamoxifen therapy and may contribute to mortality. Thus, it is crucial to overcome this problem. Here, using luciferase reporter assays, qRT-PCR, and Western blot analyses, we demonstrate that the microRNA miR-486-5p targets HMGA1 mRNA, decreasing its mRNA and protein levels in ER-positive (ER+) BC cells. Consistently, miR-486-5p is significantly downregulated, whereas HMGA1 is considerably upregulated in ER+ BC samples. Remarkably, while both miR-486-5p and tamoxifen individually cause G2/M cell cycle arrest, combination treatment synergistically causes profound cell death, specifically in tamoxifen-resistant ER+ cells but not in tamoxifen-sensitive ER+ cells. Combined treatment with miR-486-5p and tamoxifen also additively reduces cell migration, invasion, colony formation, mammary spheroid formation and a CD24-CD44+ cell population, representing decreased cancer stemness. However, these phenomena are independent of the tamoxifen responsiveness of the ER+ BC cells. Thus, miR-486-5p and tamoxifen exhibit additive and synergistic tumor-suppressive effects, most importantly causing profound cell death specifically in tamoxifen-resistant BC cells. Therefore, our work suggests that combining miR-486-5p replacement therapy with tamoxifen treatment is a promising strategy to treat endocrine therapy-resistant BC.
Collapse
Affiliation(s)
- Behzad Mansoori
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Pouria Savadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Mansoori
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Afsaneh Nazari
- Department of Genetics, Faculty of Basic Sciences, Islamic Azad University, Zanjan Branch, Zanjan, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Roshani
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Pascal Hg Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Centre for Data Science, Queensland University of Technology, Brisbane, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Sang R, Stratton B, Engel A, Deng W. Liposome technologies towards colorectal cancer therapeutics. Acta Biomater 2021; 127:24-40. [PMID: 33812076 DOI: 10.1016/j.actbio.2021.03.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth most common deadly cancer worldwide. After treatment with curative intent recurrence rates vary with staging 0-13% in Stage 1, 11-61% in S2 and 28-73% in Stage 3. The toxicity to healthy tissues from chemotherapy and radiotherapy and drug resistance severely affect the quality of life and cancer specific outcomes of CRC patients. To overcome some of these limitations, many efforts have been made to develop nanomaterial-based drug delivery systems. Among these nanocarriers, liposomes represented one of the most successful candidates in delivering targeted oncological treatment, improving safety profile and therapeutic efficacy of encapsulated drugs. In this review we will discuss liposome design with a particular focus on the targeting feature and triggering functions. We will also summarise the recent advances in liposomal delivery system for CRC treatment in both the preclinical and clinical studies. We will finally provide our perspectives on the liposome technology development for the future clinical translation. STATEMENT OF SIGNIFICANCE: Conventional treatments for colorectal cancer (CRC) severely affect the therapeutic effects for advanced patients. With the development of nanomedicines, liposomal delivery system appears to be one of the most promising nanocarriers for CRC treatment. In last three years several reviews in this area have been published focusing on the preclinical research and drug delivery function, which is a fairly narrow focus in the field of liposome technology for CRC therapy. Our review presented the most recent advances of the liposome technology (both clinical and preclinical applications) for CRC with strong potential for further clinical translation. We believe it will attract lots of attention from various audiences, including researchers, clinicians and the industry.
Collapse
|
14
|
Alswady-Hoff M, Erdem JS, Phuyal S, Knittelfelder O, Sharma A, Fonseca DDM, Skare Ø, Slupphaug G, Zienolddiny S. Long-Term Exposure to Nanosized TiO 2 Triggers Stress Responses and Cell Death Pathways in Pulmonary Epithelial Cells. Int J Mol Sci 2021; 22:ijms22105349. [PMID: 34069552 PMCID: PMC8161419 DOI: 10.3390/ijms22105349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/03/2023] Open
Abstract
There is little in vitro data available on long-term effects of TiO2 exposure. Such data are important for improving the understanding of underlying mechanisms of adverse health effects of TiO2. Here, we exposed pulmonary epithelial cells to two doses (0.96 and 1.92 µg/cm2) of TiO2 for 13 weeks and effects on cell cycle and cell death mechanisms, i.e., apoptosis and autophagy were determined after 4, 8 and 13 weeks of exposure. Changes in telomere length, cellular protein levels and lipid classes were also analyzed at 13 weeks of exposure. We observed that the TiO2 exposure increased the fraction of cells in G1-phase and reduced the fraction of cells in G2-phase, which was accompanied by an increase in the fraction of late apoptotic/necrotic cells. This corresponded with an induced expression of key apoptotic proteins i.e., BAD and BAX, and an accumulation of several lipid classes involved in cellular stress and apoptosis. These findings were further supported by quantitative proteome profiling data showing an increase in proteins involved in cell stress and genomic maintenance pathways following TiO2 exposure. Altogether, we suggest that cell stress response and cell death pathways may be important molecular events in long-term health effects of TiO2.
Collapse
Affiliation(s)
- Mayes Alswady-Hoff
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
| | - Johanna Samulin Erdem
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
| | - Santosh Phuyal
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, NO-0316 Oslo, Norway
| | | | - Animesh Sharma
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (A.S.); (D.d.M.F.); (G.S.)
- Proteomics and Metabolomics Core Facility (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Davi de Miranda Fonseca
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (A.S.); (D.d.M.F.); (G.S.)
- Proteomics and Metabolomics Core Facility (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Øivind Skare
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
| | - Geir Slupphaug
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (A.S.); (D.d.M.F.); (G.S.)
- Proteomics and Metabolomics Core Facility (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Shanbeh Zienolddiny
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
- Correspondence: ; Tel.: +47-23195284
| |
Collapse
|
15
|
Mansoori B, Duijf PHG, Mohammadi A, Safarzadeh E, Ditzel HJ, Gjerstorff MF, Cho WCS, Baradaran B. MiR-142-3p targets HMGA2 and suppresses breast cancer malignancy. Life Sci 2021; 276:119431. [PMID: 33785332 DOI: 10.1016/j.lfs.2021.119431] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022]
Abstract
MicroRNAs (miRNAs) have the ability to regulate gene expression programs in cells. Hence, altered expression of miRNAs significantly contributes to breast cancer development and progression. Here, we demonstrate that the miRNA miR-142-3p directly targets the 3' untranslated region of HMGA2, which encodes an onco-embryonic protein that is overexpressed in most cancers, including breast cancer. Down regulation of miR-142-3p predicting poor patient survival in grade 3 breast cancer (P-value = 0.045). MiR-142-3p downregulates HMGA2 mRNA and protein levels. Higher miR-142-3p and lower HMGA2 expressed are found in breast cancer versus normal breast tissue (P-value<0.05), and their levels inversely correlate in breast cancers (P-value = 1.46 × 10-4). We demonstrate that miR-142-3p induces apoptosis and G2/M cell cycle arrest in breast cancer cells. In addition, it inhibits breast cancer stem cell properties and decreases SOX2, NANOG, ALDH and c-Myc expression. MiR-142-3p also decreases cell proliferation through inhibition of the ERK/AKT/STAT3 signaling pathways. Finally, pathway analyses of patient samples suggest that these mechanisms also acting in the tumors of breast cancer patients. Thus, our work identifies HMGA2 as a direct miR-142-3p target and indicates that miR-142-3p is an important suppressor of breast cancer oncogenesis. This identifies miR-142-3p may candidate as a therapeutic molecule for breast cancer treatment.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Elham Safarzadeh
- Department of Microbiology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
HMGA2 as a Critical Regulator in Cancer Development. Genes (Basel) 2021; 12:genes12020269. [PMID: 33668453 PMCID: PMC7917704 DOI: 10.3390/genes12020269] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human malignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, and therapeutic purposes.
Collapse
|
17
|
Mansoori B, Silvestris N, Mohammadi A, Khaze V, Baghbani E, Mokhtarzadeh A, Shanehbandi D, Derakhshani A, Duijf PHG, Baradaran B. miR-34a and miR-200c Have an Additive Tumor-Suppressive Effect on Breast Cancer Cells and Patient Prognosis. Genes (Basel) 2021; 12:267. [PMID: 33673143 PMCID: PMC7918749 DOI: 10.3390/genes12020267] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common women's malignancy in the world and, for subgroups of patients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR-34a and miR-200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR-34a and miR-200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR-34a and miR-200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness and epithelial-to-mesenchymal transition (EMT). Mechanistically, both miRNA-34a and miR-200c directly target HIF1-α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA-34a and miR-200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co-delivery of miR-34a and miR-200c represents a promising novel therapeutic strategy for breast cancer patients.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000C Odense, Denmark;
| | - Nicola Silvestris
- Medical Oncology Unit-IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, DIMO-University of Bari, 70124 Bari, Italy
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000C Odense, Denmark;
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Pascal H. G. Duijf
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Brisbane, QLD 4102, Australia
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
| |
Collapse
|
18
|
Zhao YC, Jiao Y, Li YQ, Fu Z, Yang ZY, He M. Elevated high mobility group A2 expression in liver cancer predicts poor patient survival. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2021; 112:27-33. [PMID: 31823639 DOI: 10.17235/reed.2019.6365/2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND liver cancer is a malignant tumor with a high morbidity and mortality that endangers human health. High mobility group A2 (HMGA2) is a chromosome associated protein that participates in embryogenesis, tissue development, tumorigenesis and development. OBJECTIVE to explore the relationship between HMGA2 expression and the clinicopathological parameters and survival of liver cancer patients using The Cancer Genome Atlas Liver Hepatocellular Carcinoma (HCC) data. METHODS RNA-sequencing data and the corresponding clinical characteristics of the patients were downloaded from the Atlas database. The Chi-squared test was used to assess the relationship between HMGA2 expression and clinical variables. Cox regression analysis was used to compare survival rates between the high- and low-expressing groups; the p-values and Kaplan-Meier survival curves were compared using the log-rank test. RESULTS RNA-seq data from 373 cases of liver cancer cases were analyzed. HMGA2 was overexpressed in liver cancer and significantly associated with gender (p = 0.0357), T classification (p = 0.0063), clinical classification (p = 0.0026) and overall survival (p = 0.0386). According to the multivariate analysis, HMGA2 could independently predict overall survival in liver cancer. CONCLUSIONS HMGA2 independently predicts poor prognosis in liver cancer and serves as a molecular marker to determine disease prognosis.
Collapse
Affiliation(s)
- Yue-Chen Zhao
- Radiation Oncology, The Second Hospital of Jilin University, China
| | - Yan Jiao
- Department of hepatobiliary and pancreatic surgery, Jilin University
| | - Yan-Qing Li
- Pathophysiology, College of Basic Medical Sciences, Jilin University, China
| | - Zhuo Fu
- Hand and Foot Surgery, The First Hospital of Jilin University
| | - Zhao-Ying Yang
- Breast Surgery, China-Japan Union Hospital of Jilin University, China
| | - Miao He
- Anesthesia, The Second Hospital of Jilin University, China
| |
Collapse
|
19
|
Wang MD, Xing H, Li C, Liang L, Wu H, Xu XF, Sun LY, Wu MC, Shen F, Yang T. A novel role of Krüppel-like factor 8 as an apoptosis repressor in hepatocellular carcinoma. Cancer Cell Int 2020; 20:422. [PMID: 32874135 PMCID: PMC7456055 DOI: 10.1186/s12935-020-01513-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/21/2020] [Indexed: 01/04/2023] Open
Abstract
Background Krüppel-like factor 8 (KLF8), a cancer-promoting factor that regulates critical gene transcription and cellular cancer-related events, has been implicated in tumor development and progression. However, the functional role of KLF8 in the pathogenesis of hepatocellular carcinoma (HCC) remains largely unknown. Methods The gene expression patterns and genome-wide regulatory profiles of HCC cells after KLF8 knockout were analyzed by using RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) of histone H3 lysine 27 acetylation (H3K27ac) combined with bioinformatics analysis. Transcription factor-binding motifs that recognized by KLF8 were evaluated by motif analysis. For the predicted target genes, transcriptional changes were examined by ChIP, and loss of function experiments were conducted by siRNA transfection. Results KLF8 functioned as a transcription repressor in HCC and mainly regulated apoptotic-related genes directly. A total of 1,816 differentially expressed genes after KLF8 knockout were identified and significantly corresponded to global changes in H3K27ac status. Furthermore, two predicted target genes, high-mobility group AT-hook 2 (HMGA2) and matrix metalloproteinase 7 (MMP7), were identified as important participants in KLF8-mediated anti-apoptotic effect in HCC. Knockout of KLF8 enhanced cell apoptosis process and caused increase in the associated H3K27ac, whereas suppression HMGA2 or MMP7 attenuated these biological effects. Conclusions Our work suggests a novel role and mechanism for KLF8 in the regulation of cell apoptosis in HCC and facilitates the discovery of potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Hao Xing
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Lei Liang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Han Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Xin-Fei Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Li-Yang Sun
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China.,Department of Clinical Medicine, Second Military Medical University (Navy Medical University), Shanghai, China
| | - Meng-Chao Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| |
Collapse
|
20
|
Lotfinejad P, Kazemi T, Mokhtarzadeh A, Shanehbandi D, Jadidi Niaragh F, Safaei S, Asadi M, Baradaran B. PD-1/PD-L1 axis importance and tumor microenvironment immune cells. Life Sci 2020; 259:118297. [PMID: 32822718 DOI: 10.1016/j.lfs.2020.118297] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 12/23/2022]
Abstract
Triple-negative breast cancer (TNBC) is heterogeneous cancer with poor prognosis among the other breast tumors. Rapid recurrence and increased progression rate could be reasons for the poor prognosis of this type of breast cancer. Recently, because of the lack of specific targets in multiple cancer treatment, immune checkpoint blockade therapies with targeting PD-1/PD-L1 axis have displayed significant advances and improved survival. Among different types of breast cancers, TNBC is considered more immunogenic with high T-cell and other immune cells infiltration compared to other breast cancer subtypes. This immunogenic characteristic of TNBC is a beneficial marker in the immunotherapy of these tumors. Clinical studies with a focus on immune checkpoint therapy have demonstrated promising results in TNBC treatment. In this review, we summarize clinical trials with the immunotherapy-based treatment of different cancers and also discuss the interaction between infiltrating immune cells and breast tumor microenvironment. In addition, we focus on the signaling pathway that controls PD-L1 expression and continues with CAR T-cell therapy and siRNA as novel strategies and potential tools in targeted therapy.
Collapse
Affiliation(s)
- Parisa Lotfinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Mohammadi A, Mansoori B, Duijf PHG, Safarzadeh E, Tebbi L, Najafi S, Shokouhi B, Sorensen GL, Holmskov U, Baradaran B. Restoration of miR-330 expression suppresses lung cancer cell viability, proliferation, and migration. J Cell Physiol 2020; 236:273-283. [PMID: 32583462 DOI: 10.1002/jcp.29840] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/29/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022]
Abstract
Lung cancer is one of the most common cancers and its incidence is rising around the world. Various studies suggest that miR-330 acts as a tumor-suppressor microRNA (miRNA) in different types of cancers, but precisely how has remained unclear. In this study, we investigate miR-330 expression in lung cancer patient samples, as well as in vitro, by studying how normalization of miR-330 expression affects lung cancer cellular phenotypes such as viability, apoptosis, proliferation, and migration. We establish that low miR-330 expression predicts poor lung cancer prognosis. Stable restoration of reduced miR-330 expression in lung cancer cells reduces cell viability, increases the fraction of apoptotic cells, causes G2/M cell cycle arrest, and inhibits cell migration. These findings are substantiated by increased mRNA and protein expression of markers for apoptosis via the intrinsic pathway, such as caspase 9, and decreased mRNA and protein expression of markers for cell migration, such as vimentin, C-X-C chemokine receptor type 4, and matrix metalloproteinase 9. We showed that reduced miR-330 expression predicts poor lung cancer survival and that stable restoration of miR-330 expression in lung cancer cells has a broad range of tumor-suppressive effects. This indicates that miR-330 is a promising candidate for miRNA replacement therapy for lung cancer patients.
Collapse
Affiliation(s)
- Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Elham Safarzadeh
- Department of Microbiology & Immunology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Leila Tebbi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrooz Shokouhi
- Departmentof Infectious Diseases, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Grith L Sorensen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Esmaeili M, Keshani M, Vakilian M, Esmaeili M, Peymani M, Seyed Forootan F, Chau TL, Göktuna SI, Zaker SR, Nasr Esfahani MH, Ghaedi K. Role of non-coding RNAs as novel biomarkers for detection of colorectal cancer progression through interaction with the cell signaling pathways. Gene 2020; 753:144796. [PMID: 32450203 DOI: 10.1016/j.gene.2020.144796] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/06/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer which affects the colon and the rectum. Approximately one third of annual CRC mortality occurs due to the late detection of this type of cancer. Therefore, there is an urgent need for more powerful diagnostic and prognostic tools for identification and treatment of colorectal tumorigenesis. Non-coding RNAs (ncRNAs) have been implicated in the pathology of CRC and also linked to metastasis, proliferation, differentiation, migration, angiogenesis and apoptosis in numerous cancers. Recently, attention has turned towards ncRNAs as specific targets for diagnosis, prognosis and treatment of various types of cancers, including CRC. In this review, we have tried to outline the roles of ncRNAs, and their involvement in signaling pathways responsible for the progression of CRC.
Collapse
Affiliation(s)
- Mohadeseh Esmaeili
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran.
| | - Maryam Keshani
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran.
| | - Mehrdad Vakilian
- Department of Cell Regeneration and Advanced Therapies, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain; Department of Cellular Biology, Genetics and Physiology, Faculty of Science, University of Malaga (UMA), Málaga, Spain.
| | - Maryam Esmaeili
- Department of Cellular Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Department of Cellular Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Farzad Seyed Forootan
- Department of Cellular Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Legal Medicine Research Centre, Legal Medicine Organization, Tehran, Iran.
| | - Tieu Lan Chau
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey.
| | - Serkan Ismail Göktuna
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey; National Nanotechnology Research Institute (UNAM), Bilkent University, Ankara, Turkey.
| | - Sayed Rasoul Zaker
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Mohammad Hossein Nasr Esfahani
- Department of Cellular Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran; Department of Cellular Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
23
|
Zhu WJ, Yan Y, Zhang JW, Tang YD, Han B. Effect and Mechanism of miR-26a-5p on Proliferation and Apoptosis of Hepatocellular Carcinoma Cells. Cancer Manag Res 2020; 12:3013-3022. [PMID: 32431544 PMCID: PMC7200261 DOI: 10.2147/cmar.s237752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Aim This study aimed to investigate the effect and mechanism of miR-26a-5p on proliferation and apoptosis of hepatocellular carcinoma (HCC) cells. Methods RT-PCR was used to analyze the expression of miR-26a-5p in HCC cells and its targeted gene HMGA2 mRNA determined by biological information prediction. The rate of proliferation, invasion, apoptosis, and expression levels of related proteins of HCC cells overexpressing miR-26a-5p or those after knocking down HMGA2 expression were detected by MTT, invasion and apoptosis rate tests. Moreover, the apoptosis-promoting protein bax was upregulated and the anti-apoptosis-related protein Bcl-2 was downregulated. Results RT-qPCR results showed that the level of miR-26a-5p was downregulated in HCC tissues and cells, and the expression of HMGA2 was upregulated; besides, the expression of miR-26a-5p and HMGA2 was negatively correlated; miR-26a-5p was correlated with tumor diameter, differentiation degree, TNM staging and lymph node metastasis. Cell tests confirmed that miR-26a-5p functioned in tumor suppression, including inhibiting cell proliferation and invasion in two hepatocellular carcinoma cell lines and promoting apoptosis. Bioinformatics prediction and subsequent experiments proved that HMGA2 was the direct target of miR-26a-5p; moreover, after knocking down HMGA2 expression in HCC cells, cell proliferation and invasion ability were significantly inhibited, and apoptosis rate increased significantly. Conclusion miR-26a-5p can inhibit the proliferation and invasion of HCC cells and promote their apoptosis by directly targeting HMGA2. Abnormal decrease of miR-26a-5p and increase of its target HMGA2 are important factors that may participate in the occurrence and development of HCC. miR-26a-5p may be a new potential target for its treatment.
Collapse
Affiliation(s)
- Wen-Jing Zhu
- Abdominal Ultrasonic Department, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ying Yan
- Oncology Department, The First Hospital of Harbin, Harbin, Heilongjiang Province, People's Republic of China
| | - Jiu-Wei Zhang
- Abdominal Ultrasonic Department, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Yan-Dong Tang
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang Province, People's Republic of China
| | - Bo Han
- Oncology Department, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
24
|
Unachukwu U, Chada K, D’Armiento J. High Mobility Group AT-Hook 2 (HMGA2) Oncogenicity in Mesenchymal and Epithelial Neoplasia. Int J Mol Sci 2020; 21:ijms21093151. [PMID: 32365712 PMCID: PMC7246488 DOI: 10.3390/ijms21093151] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022] Open
Abstract
High mobility group AT-hook 2 (HMGA2) has been associated with increased cell proliferation and cell cycle dysregulation, leading to the ontogeny of varied tumor types and their metastatic potentials, a frequently used index of disease prognosis. In this review, we deepen our understanding of HMGA2 pathogenicity by exploring the mechanisms by which HMGA2 misexpression and ectopic expression induces mesenchymal and epithelial tumorigenesis respectively and distinguish the pathogenesis of benign from malignant mesenchymal tumors. Importantly, we highlight the regulatory role of let-7 microRNA family of tumor suppressors in determining HMGA2 misexpression events leading to tumor pathogenesis and focused on possible mechanisms by which HMGA2 could propagate lymphangioleiomyomatosis (LAM), benign mesenchymal tumors of the lungs. Lastly, we discuss potential therapeutic strategies for epithelial and mesenchymal tumorigenesis based on targeting the HMGA2 signaling pathway.
Collapse
Affiliation(s)
- Uchenna Unachukwu
- Department of Anesthesiology, Columbia University Medical Center, 630 West 168th Street, P&S 12-402, New York, NY 10032, USA;
| | - Kiran Chada
- Department of Biochemistry & Molecular Biology; Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854, USA;
| | - Jeanine D’Armiento
- Department of Anesthesiology, Columbia University Medical Center, 630 West 168th Street, P&S 12-402, New York, NY 10032, USA;
- Correspondence: ; Tel.: +212-305-3745
| |
Collapse
|
25
|
miR-4651 inhibits cell proliferation of gingival mesenchymal stem cells by inhibiting HMGA2 under nifedipine treatment. Int J Oral Sci 2020; 12:10. [PMID: 32231210 PMCID: PMC7105500 DOI: 10.1038/s41368-020-0076-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/12/2020] [Accepted: 02/16/2020] [Indexed: 12/11/2022] Open
Abstract
Drug-induced gingival overgrowth (DIGO) is recognized as a side effect of nifedipine (NIF); however, the underlying molecular mechanisms remain unknown. In this study, we found that overexpressed miR-4651 inhibits cell proliferation and induces G0/G1-phase arrest in gingival mesenchymal stem cells (GMSCs) with or without NIF treatment. Furthermore, sequential window acquisition of all theoretical mass spectra (SWATH-MS) analysis, bioinformatics analysis, and dual-luciferase report assay results confirmed that high-mobility group AT-hook 2 (HMGA2) is the downstream target gene of miR-4651. Overexpression of HMGA2 enhanced GMSC proliferation and accelerated the cell cycle with or without NIF treatment. The present study demonstrates that miR-4651 inhibits the proliferation of GMSCs and arrests the cell cycle at the G0/G1 phase by upregulating cyclin D and CDK2 while downregulating cyclin E through inhibition of HMGA2 under NIF stimulation. These findings reveal a novel mechanism regulating DIGO progression and suggest the potential of miR-4651 and HMGA2 as therapeutic targets.
Collapse
|
26
|
Mansoori B, Duijf PHG, Mohammadi A, Najafi S, Roshani E, Shanehbandi D, Hajiasgharzadeh K, Shirjang S, Ditzel HJ, Kazemi T, Mokhtarzadeh A, Gjerstorff MF, Baradaran B. Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness. Expert Opin Ther Targets 2020; 24:255-265. [PMID: 32172636 DOI: 10.1080/14728222.2020.1736559] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 02/18/2020] [Indexed: 01/07/2023]
Abstract
Despite improved therapeutic strategies for early-stage breast cancer, the most common cancer type in women, relapse remains common and the underlying mechanisms for this progression remain poorly understood. To gain more insight, we studied the DNA-binding protein HMGA2 in breast cancer development and stemness. We demonstrated that HMGA2 is overexpressed in breast cancer tissues at the mRNA and protein levels (P value <0.0001). HMGA2 knockdown and overexpression in breast cancer cells revealed that HMGA2 promotes cell proliferation and protects against apoptosis via the intrinsic pathway. HMGA2 knockdown also causes cell cycle arrest in G2/M phase. In addition, we found that HMGA2 increases breast cancer cell migration and invasion (P value <0.001) and promotes the acquisition of cancer stem cell features, both in vitro, in colony formation (P value <0.01) and spheroid assays, and in breast cancer tissues. Overexpression of HMGA2 in breast cancer spurs the acquisition of several hallmarks of cancer, including increased cell proliferation, migration, invasion and stemness, and decreased apoptosis. Thus, targeting HMGA2 could represent an effective strategy to block breast cancer progression.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Roshani
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Institute for Clinical Research, Odense University Hospital, Odense, Denmark
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
An Integrated Bioinformatics Analysis Repurposes an Antihelminthic Drug Niclosamide for Treating HMGA2-Overexpressing Human Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11101482. [PMID: 31581665 PMCID: PMC6826424 DOI: 10.3390/cancers11101482] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/11/2022] Open
Abstract
Aberrant overexpression of high mobility group AT-hook 2 (HMGA2) is frequently found in cancers and HMGA2 has been considered an anticancer therapeutic target. In this study, a pan-cancer genomics survey based on Cancer Cell Line Encyclopedia (CCLE) and The Cancer Genome Atlas (TCGA) data indicated that HMGA2 was mainly overexpressed in gastrointestinal cancers including colorectal cancer. Intriguingly, HMGA2 overexpression had no prognostic impacts on cancer patients’ overall and disease-free survivals. In addition, HMGA2-overexpressing colorectal cancer cell lines did not display higher susceptibility to a previously identified HMGA2 inhibitor (netroposin). By microarray profiling of HMGA2-driven gene signature and subsequent Connectivity Map (CMap) database mining, we identified that S100 calcium-binding protein A4 (S100A4) may be a druggable vulnerability for HMGA2-overexpressing colorectal cancer. A repurposing S100A4 inhibitor, niclosamide, was found to reverse the HMGA2-driven gene signature both in colorectal cancer cell lines and patients’ tissues. In vitro and in vivo experiments validated that HMGA2-overexpressing colorectal cancer cells were more sensitive to niclosamide. However, inhibition of S100A4 by siRNAs and other inhibitors was not sufficient to exert effects like niclosamide. Further RNA sequencing analysis identified that niclosamide inhibited more cell-cycle-related gene expression in HMGA2-overexpressing colorectal cancer cells, which may explain its selective anticancer effect. Together, our study repurposes an anthelminthic drug niclosamide for treating HMGA2-overexpression colorectal cancer.
Collapse
|
28
|
Xu X, Zou H, Luo L, Wang X, Wang G. MicroRNA-9 exerts antitumor effects on hepatocellular carcinoma progression by targeting HMGA2. FEBS Open Bio 2019; 9:1784-1797. [PMID: 31408273 PMCID: PMC6768112 DOI: 10.1002/2211-5463.12716] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/24/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence has demonstrated that the aberrant expression of microRNAs (miRs or miRNAs) may contribute to the initiation and progression of various types of human cancer and may also constitute biomarkers for cancer diagnosis and therapy. However, the specific function of miR‐9 in hepatocellular carcinoma (HCC) remains unclear, and the mechanisms that underlie HCC are incompletely understood. Here, we report that miR‐9 expression was significantly decreased in clinical tumor tissue samples, as well as in a cohort of HCC cell lines. In addition, it was demonstrated that overexpression of miR‐9 suppressed the proliferative and migratory capacity of HCC cells and impaired cell cycle progression. Furthermore, high mobility group AT‐hook 2 (HMGA2) was verified as a downstream target gene of miR‐9 using a luciferase reporter assay. Quantitative RT‐PCR and western blotting implicated HMGA2 in the miR‐9‐mediated reduction of HCC cell growth. In vivo, transfection with miR‐9 mimics down‐regulated the expression of HMGA2, thus leading to a dramatic reduction in tumor growth in a mouse xenograft model. These results suggest that miR‐9 may exert critical antitumor effects on HCC by directly targeting HMGA2, and the miR9/HMGA2 signaling pathway may be of use for the diagnosis and prognosis of patients with HCC.
Collapse
Affiliation(s)
- Xiangang Xu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Haibo Zou
- Department of Hepatobiliary Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Lanyun Luo
- Department of Hepatobiliary Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Xiankui Wang
- Department of Hepatobiliary Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Guan Wang
- Department of Hepatobiliary Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
29
|
Zarredar H, Farajnia S, Ansarin K, Baradaran B, Aria M, Asadi M. Synergistic Effect of Novel EGFR Inhibitor AZD8931 and p38α siRNA in Lung Adenocarcinoma Cancer Cells. Anticancer Agents Med Chem 2019; 19:638-644. [DOI: 10.2174/1871520619666190301125203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/26/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
Background:Lung cancer is the leading cause of cancer-related death with less than 5-year survival rate for both men and women worldwide. EGFR and MAPK signaling pathways have a critical role in proliferation and progression of various cancers, including lung cancer. P38 map kinase plays different role in various tissue hence showing a tissue-dependent behavior. It acts as an oncogene in some tissues while plays as tumor suppressor in some other tissues. The aim of this study was to investigate the combined effect of P38 αspecific siRNA and EGFR inhibitor on apoptosis and proliferation of A549 lung cancer cell line.Objective:This article is dedicated to the synergistic effect of novel EGFR inhibitor AZD8931 and P38 α siRNA in lung adenocarcinoma cancer cells proliferation and apoptosis.Methods and Materials:The A549 lung cancer cells were treated with P38 α- siRNA and EGFR inhibitor alone or in combination. The cytotoxic effects of P38 α- siRNA and EGFR inhibitor were determined using MTT assay. Relative P38 α and EGFR mRNA levels were measured by QRT-PCR. Induction of apoptosis were measured by FACS analysis.Results:The expression of mRNA related to P38 α, EGFR, and Her2 genes was reduced to 23.4%, 52.4%, and 75, respectively, after treatment of their inhibitors. Also, MTT assay showed that the cell viability after treatment with p38 α SiRNA, EGFR inhibitor and their combination was reduced to 51.02%, 48.9%, and 25.11%, respectively. FACS results indicated that p38 α siRNA, EGFR inhibitor and their combination, reduced the population of live cells to 49.5%, 32.2% and 14.3% in comparison to the population of untreated control cells (99.5%).Conclusion:The results of this study indicated that p38 α and EGFR might play an important role in the development and growth of lung cancer and might be a potential therapeutic target for the treatment of lung cancer.
Collapse
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Aria
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Mansoori B, Mohammadi A, Naghizadeh S, Gjerstorff M, Shanehbandi D, Shirjang S, Najafi S, Holmskov U, Khaze V, Duijf PHG, Baradaran B. miR-330 suppresses EMT and induces apoptosis by downregulating HMGA2 in human colorectal cancer. J Cell Physiol 2019; 235:920-931. [PMID: 31241772 DOI: 10.1002/jcp.29007] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/31/2019] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) are important molecular regulatorsof cellular signaling and behavior. They alter gene expression by targeting messenger RNAs, including those encoding transcriptional regulators, such as HMGA2. While HMGA2 is oncogenic in various tumors, miRNAs may be oncogenic or tumor suppressive. Here, we investigate the expression of HMGA2 and the miRNA miR-330 in a patient with colorectal cancer (CRC) samples and their effects on oncogenic cellular phenotypes. We found that HMGA2 expression is increased and miR-330 expression is decreased in CRCs and each predicts poor long-term patient survival. Stably increased miR-330 expression in human colorectal cancer cells (HCT116) and SW480 CRC cell lines downregulate the oncogenic expression of HMGA2, a predicted miR-330 target. Additionally, this promotes apoptosis and decreases cell migration and viability. Consistently, it also decreases protein-level expression of markers for epithelial-to-mesenchymal-transition (Snail-1, E-cadherin, and Vascular endothelial growth factor receptors) and transforming growth factor β signaling (SMAD3), as well as phospho- Protein kinase B (AKT) and phospho-STAT3 levels. We conclude that miR-330 acts as a tumor suppressor miRNA in CRC by suppressing HMGA2 expression and reducing cell survival, proliferation, and migration. Thus, we identify miR-330 as a promising candidate for miRNA replacement therapy for patients with CRC.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sanaz Naghizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Translational Research Institute, University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Mansoori B, Mohammadi A, Asadzadeh Z, Shirjang S, Minouei M, Abedi Gaballu F, Shajari N, Kazemi T, Gjerstorff MF, Duijf PHG, Baradaran B. HMGA2 and Bach-1 cooperate to promote breast cancer cell malignancy. J Cell Physiol 2019; 234:17714-17726. [PMID: 30825204 DOI: 10.1002/jcp.28397] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
During breast cancer progression, tumor cells acquire multiple malignant features. The transcription factors and cell cycle regulators high mobility group A2 (HMGA2) and BTB and CNC homology 1 (Bach-1) are overexpressed in several cancers, but the mechanistic understanding of how HMGA2 and Bach-1 promote cancer development has been limited. We found that HMGA2 and Bach-1 are overexpressed in breast cancer tissues and their expression correlates positively in tumors but not in normal tissues. Individual HMGA2 or Bach-1 knockdown downregulates expression of both proteins, suggesting a mutual stabilizing effect between the two proteins. Importantly, combined HMGA2 and Bach-1 knockdown additively decrease cell proliferation, migration, epithelial-to-mesenchymal transition, and colony formation, while promoting apoptotic cell death via upregulation of caspase-3 and caspase-9. First the first time, we show that HMGA2 and Bach-1 overexpression in tumors correlate positively and that the proteins cooperatively suppress a broad range of malignant cellular properties, such as proliferation, migration, clonogenicity, and evasion of apoptotic cell death. Thus, our observations suggest that combined targeting of HMGA2 and Bach1 may be an effective therapeutic strategy to treat breast cancer.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Minouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Khordadmehr M, Shahbazi R, Sadreddini S, Baradaran B. miR-193: A new weapon against cancer. J Cell Physiol 2019; 234:16861-16872. [PMID: 30779342 DOI: 10.1002/jcp.28368] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
microRNAs (miRNAs) are known as a large group of short noncoding RNAs, which structurally consist of 19-22 nucleotides in length and functionally act as one of the main regulators of gene expression in important biological and physiological contexts like cell growth, apoptosis, proliferation, differentiation, movement (cell motility), and angiogenesis as well as disease formation and progression importantly in cancer cell invasion, migration, and metastasis. Among these notable tiny molecules, many studies recently presented the important role of the miR-193 family comprising miR-193a-3p, miR-193a-5p, miR-193b-3p, and miR-193b-5p in health and disease biological processes by interaction with special targeting and signaling, which mainly contribute as a tumor suppressor. Therefore, in the present paper, we review the functional role of this miRNA family in both health and disease conditions focusing on various tumor developments, diagnoses, prognoses, and treatment.
Collapse
Affiliation(s)
- Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Roya Shahbazi
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Sanam Sadreddini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Mohammadi A, Mansoori B, Savadi P, Khaze V, Minouei M, McMillan NAJ, Hallaj-Nezhadi S, Baradaran B. Targeting of high mobility group A2 by small interfering RNA-loaded nanoliposome-induced apoptosis and migration inhibition in gastrointestinal cancer cells. J Cell Biochem 2018; 120:9203-9212. [PMID: 30507008 DOI: 10.1002/jcb.28196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Considering the complex nature of gastrointestinal cancer, different methods including surgery, radiotherapy, and chemotherapy are considered for the treatment. Novel strategies including silencing of oncogenes using safe delivery systems could be considered as a novel approach in colorectal cancer treatment. The aim of this study was to investigate the silencing effect of high mobility group A2 (HMGA2) small interfering RNA (siRNA)-loaded nanoliposomes on gastrointestinal cancers. METHODS The siRNA-lipoplexes were prepared using dioleoyl trimethylammonium propane (DOTAP)/cholesterol (Chol)/1, 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) through the freeze-drying of a monophase solution method. The size, polydispersity index (PDI), and zeta-potential of nanoliposomes were determined using Zetasizer analyzer. The morphology of the nanoliposomes was determined by transmission electron microscopy (TEM). The agarose gel-retardation assay was carried out to confirm the loading of siRNAs into liposome. The silencing of the HMGA2 in cancer cells was evaluated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The effect of liposomes on cell cytotoxicity was studied by MTT assay. The inhibitory effect of siRNA-loaded liposomes was evaluated by a wound-healing assay. The apoptosis induction was investigated via the annexin V/propidium iodide assay. RESULTS The size, PDI, and zeta-potential of the prepared liposomes were found to be 350 nm, 0.67, and 86.3 mV, respectively. They were spherical in shape and could efficiently associate with siRNA. The results of gene silencing showed that the optimum condition of HMGA2 silencing was 80 pmol HMGA2 and 24 hours after treatment in each cancer cell lines. MTT assays indicated that silencing of HMGA2 in optimal condition could reduce the viability of the cancer cells more than 60% in the three cell lines. The result of the apoptosis assay showed more than 50% of the cell deaths related to the apoptosis in all three cell lines. The gene expression evaluation confirmed that apoptosis was induced via the intrinsic pathway inducing both caspase-3 and -9 expressions. Also, the reduction in Bcl2 expression confirmed the activation apoptosis pathway in the treated cancer cells. The wound-healing assay showed the suppression of cancer cell migration after treatment with the prepared nanoliposomes. CONCLUSION The results of this study showed the HMGA2 siRNA-loaded nanoliposomes could be effective in the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouria Savadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Minouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nigel A J McMillan
- School of Medical Sciences and Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Baradaran B, Shahbazi R, Khordadmehr M. Dysregulation of key microRNAs in pancreatic cancer development. Biomed Pharmacother 2018; 109:1008-1015. [PMID: 30551350 DOI: 10.1016/j.biopha.2018.10.177] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is mentioned as one of the fourth major cause of cancer-related deaths and also is considered as one of the most malignancies worldwide. Sadly, widely metastasis is frequently observed at the time of PC detection and there are, thereby, almost poor prognosis and ineffective treatment in PC patients. microRNAs (miRNAs), a group of short non-coding RNAs, regulate various cellular and developmental mechanisms, such as cell growth, proliferation, apoptosis, differentiation and angiogenesis. Also, they have essential roles even on the progression of different human and animal diseases. In recent years, extensive studies confirmed the important role of miRNAs in various steps of PC developments, including; tumor initiation, invasion and metastasis, which can use valuably for cancer detection, prognosis and therapy. Therefore, the present study reviewed the new recent investigations in miRNAs involvement in the biology of PC associated with their clinical implications.
Collapse
Affiliation(s)
- Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Shahbazi
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran
| | - Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| |
Collapse
|
35
|
Hajiasgharzadeh K, Somi MH, Shanehbandi D, Mokhtarzadeh A, Baradaran B. Small interfering RNA-mediated gene suppression as a therapeutic intervention in hepatocellular carcinoma. J Cell Physiol 2018; 234:3263-3276. [PMID: 30362510 DOI: 10.1002/jcp.27015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/25/2018] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the lethal and difficult-to-cure cancers worldwide. Owing to the late diagnosis and drug resistance of malignant hepatocytes, treatment of this cancer by conventional chemotherapy agents is challenging, and researchers are seeking new alternative treatment options to overcome therapy resistance in this neoplasm. RNA interference (RNAi) is a potent and specific approach in targeting gene expression and has emerged as a novel therapeutic tool for many diseases, including cancers. Small interfering RNA (siRNA) is a type of RNAi that is produced intracellularly from exogenous synthetic oligonucleotides and can selectively knock down target gene expression in a sequence-specific manner. Various factors play roles in the initiation and progression of HCC and provide multiple candidate targets for siRNA intervention. In addition, due to the liver's unique architecture and availability of some hepatic siRNA delivery methods, this organ has received much more attention as a target tissue for such oligonucleotide action. Recent advances in designing nanoparticle systems for the in vivo delivery of siRNAs have markedly enhanced the potency of siRNA-mediated gene silencing under clinical development for HCC therapy. The utility of siRNAs as anti-HCC agents is the subject of the current review. siRNA-based gene therapies could be one of the main feasible approaches for HCC therapy in the future.
Collapse
Affiliation(s)
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Balachandran A, Zambre A, Kainth JS, Nagarajha Selvan LD, Parameswaran S, Afrasiabi Z, Krishnakumar S, Kannan R, Upendran A. Targeting HMGA protein inhibits retinoblastoma cell proliferation. RSC Adv 2018; 8:31510-31514. [PMID: 35548247 PMCID: PMC9085636 DOI: 10.1039/c8ra06026f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 08/26/2018] [Indexed: 01/03/2023] Open
Abstract
We describe a novel synthetic strategy for conjugating HMGA2 siRNA and the HMGA aptamer to the nucleolin aptamer and nucleolin antibody, respectively. Our studies demonstrate that these conjugates inhibit cell proliferation in retinoblastoma cells. A novel approach to target HMGA proteins in retinoblastoma using HMGA2 siRNA–nucleolin aptamer and HMGA aptamer–nucleolin antibody conjugates was developed.![]()
Collapse
Affiliation(s)
- Akilandeswari Balachandran
- Department of Nanobiotechnology, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Chennai India
| | - Ajit Zambre
- Department of Radiology, University of Missouri Columbia MO USA
| | - Jagjot Singh Kainth
- Department of Radiology, University of Missouri Columbia MO USA .,Department of Life Sciences, Lincoln University Jefferson City MO USA
| | - Lakshmi Dhevi Nagarajha Selvan
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Chennai India
| | - Sowmya Parameswaran
- Radheshyam Kanoi Stem Cell Laboratory, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Chennai India
| | - Zahra Afrasiabi
- Department of Life Sciences, Lincoln University Jefferson City MO USA
| | - Subramanian Krishnakumar
- Department of Nanobiotechnology, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Chennai India .,L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Chennai India.,Radheshyam Kanoi Stem Cell Laboratory, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Chennai India
| | - Raghuraman Kannan
- Department of Radiology, University of Missouri Columbia MO USA .,Department of Biological Engineering, University of Missouri Columbia MO USA
| | - Anandhi Upendran
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia MO USA.,Institute of Clinical and Translational Sciences (MU-iCATS), School of Medicine, University of Missouri Columbia MO USA
| |
Collapse
|
37
|
Xie J, Ubango J, Ban Y, Chakravarti D, Kim JJ, Wei JJ. Comparative analysis of AKT and the related biomarkers in uterine leiomyomas with MED12, HMGA2, and FH mutations. Genes Chromosomes Cancer 2018; 57:485-494. [PMID: 29790226 DOI: 10.1002/gcc.22643] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 11/07/2022] Open
Abstract
Uterine leiomyomas (ULM) are histologically and molecularly heterogeneous and clinically they grow at vastly different rates. Several driver gene mutations have been identified in ULM, including MED12 mutations, HMGA2 overexpression, and biallelic FH inactivation. ULM with different driver mutant genes may use different molecular pathways, but currently no clear correlation between gene mutations and growth related pathways has been established. To better define this relationship, we collected ULM with MED12 (n = 25), HMGA2 (n = 15), and FH (n = 27) mutations and examined the sex steroid hormone, cell cycle, and AKT pathway genes by immunohistochemistry. While ER and PR were highly expressed in all types of ULM, FH ULM showed lower ER expression and higher PR expression. HMGA2 tumors had significantly higher levels of AKT signaling and mitogenic activity than other ULM types. HMGA2 activated AKT signaling through upregulation of IGF2BP2. Silencing HMGA2 in ULM cells resulted in downregulation of AKT and upregulation of p16 and p21, which eventually led to cell senescence. HMGA2 overexpression in ULM is not only related to tumor development but also plays a role in controlling cellular proliferation through the AKT pathway.
Collapse
Affiliation(s)
- Jia Xie
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Julianne Ubango
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Yanli Ban
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - J Julie Kim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jian-Jun Wei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
38
|
Zhao H, Zhao H, Xia X, Liu X. MicroRNA-599 targets high-mobility group AT-hook 2 to inhibit cell proliferation and invasion in clear cell renal carcinoma. Mol Med Rep 2018; 17:7451-7459. [PMID: 29568870 DOI: 10.3892/mmr.2018.8755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/06/2018] [Indexed: 11/05/2022] Open
Abstract
Dysregulation of microRNAs (miRNAs) is associated with the occurrence and development of clear cell renal cell carcinoma (ccRCC) through their participation in a number of critical biological processes. Therefore, an in‑depth investigation into miRNAs and their biological roles within ccRCC may provide useful insights and lead to the identification of novel therapeutic methods for patients with ccRCC. miRNA‑599 (miR‑599) serves critical roles in different types of human cancer. However, the expression pattern, biological function and molecular mechanism of miR‑599 in ccRCC remain unknown. The present study aimed to detect the expression level of miR‑599 in ccRCC, examine its effect on ccRCC progression and further explore the possible underlying mechanisms. It was observed that miR‑599 was significantly underexpressed in ccRCC tissues and cell lines compared with the control. Functional assays revealed that restored expression of miR‑599 restricted the proliferation and invasion of ccRCC cells. Bioinformatics analysis, luciferase reporter assay, reverse transcription‑quantitative polymerase chain reaction and western blot analysis demonstrated that high‑mobility group AT‑hook 2 (HMGA2) was a direct target of miR‑599 in ccRCC. HMGA2 knockdown simulated the suppressive effects caused by miR‑599 overexpression in ccRCC. Recovered HMGA2 expression partially rescued the miR‑599‑mediated inhibition of ccRCC proliferation and invasion. These results suggest that miR‑599 may serve tumour suppressive roles in ccRCC by directly targeting HMGA2, indicating that miR‑599 may have potential as a treatment for patients with ccRCC.
Collapse
Affiliation(s)
- Hailing Zhao
- Department of Pediatrics, Yidu Central Hospital of Weifang, Weifang, Shandong 262550, P.R. China
| | - Huizhen Zhao
- Department of Pediatrics, Yidu Central Hospital of Weifang, Weifang, Shandong 262550, P.R. China
| | - Xiaolin Xia
- Department of Pediatrics, Yidu Central Hospital of Weifang, Weifang, Shandong 262550, P.R. China
| | - Xiujuan Liu
- Department of Nephrology, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
39
|
Zarredar H, Ansarin K, Baradaran B, Ahdi Khosroshahi S, Farajnia S. Potential Molecular Targets in the Treatment of Lung Cancer Using siRNA Technology. Cancer Invest 2018; 36:37-58. [DOI: 10.1080/07357907.2017.1416393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
An analysis of suppressing migratory effect on human urinary bladder cancer cell line by silencing of snail-1. Biomed Pharmacother 2017; 96:545-550. [DOI: 10.1016/j.biopha.2017.10.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 11/24/2022] Open
|