1
|
Obaidur Rab S, Altalbawy FMA, Chandra M, Ariffin IA, Kaur P, Rathore G, Rizaev J, Aloraibi F, Najeeb MA, Abdulhussain MA, Zwamel AH. Targeting the lung tumor microenvironment by phytochemicals and their nanoformulations. Pathol Res Pract 2024; 264:155679. [PMID: 39500198 DOI: 10.1016/j.prp.2024.155679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024]
Abstract
Lung malignancies are among the most prevalent and foremost causes of tumor-related deaths. Despite significant advancements in the understanding and management of lung cancer, resistance to traditional treatments remains a significant challenge. Understanding and targeting tumor microenvironment (TME) have attracted interest in the recent decade for eliminating various solid tumors. The lung TME has a crucial position in tumor expansion and therapy failure, driving it an engaging target for novel medicinal interventions. Plant-derived products offer a promising avenue for targeting TME due to their diverse chemical structures and biological activities. However, their clinical use is hindered by insufficient bioavailability and also possible systemic toxicity. The use of nanoparticles as delivery vehicles for natural products can overcome these challenges and enhance their therapeutic efficacy. This review article explores the potential of plant-derived products as medicinal agents for targeting lung TME. We provide an outline of the present knowledge of lung TME and explain the mechanisms by which plant-derived products can modulate key components of this microenvironment. The promising impacts and properties of nanoparticles for the delivery of these derivatives into lung tumors will also be discussed. We also review the preclinical and clinical findings for supporting the usefulness of these agents in targeting lung TME. Additionally, we highlight the challenges and forthcoming trends in the development of plant-derived products as targeted therapies for lung cancer, with a particular focus on combination therapies.
Collapse
Affiliation(s)
- Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Muktesh Chandra
- Department of Bioinformatics, Marwadi University Research Center, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat 360003, India
| | - I A Ariffin
- Management and Science University, Shah Alam, Selangor, Malaysia
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Gulshan Rathore
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Jasur Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Farah Aloraibi
- Department of Density, Al-Manara College for Medical Sciences, Maysan, Iraq
| | - Maryam Ali Najeeb
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Valdivia-Silva J, Chinney-Herrera A. Chemokine receptors and their ligands in breast cancer: The key roles in progression and metastasis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:124-161. [PMID: 39260935 DOI: 10.1016/bs.ircmb.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Chemokines and their receptors are a family of chemotactic cytokines with important functions in the immune response in both health and disease. Their known physiological roles such as the regulation of leukocyte trafficking and the development of immune organs generated great interest when it was found that they were also related to the control of early and late inflammatory stages in the tumor microenvironment. In fact, in breast cancer, an imbalance in the synthesis of chemokines and/or in the expression of their receptors was attributed to be involved in the regulation of disease progression, including invasion and metastasis. Research in this area is progressing rapidly and the development of new agents based on chemokine and chemokine receptor antagonists are emerging as attractive alternative strategies. This chapter provides a snapshot of the different functions reported for chemokines and their receptors with respect to the potential to regulate breast cancer progression.
Collapse
Affiliation(s)
- Julio Valdivia-Silva
- Centro de Investigación en Bioingenieria (BIO), Universidad de Ingenieria y Tecnologia-UTEC, Barranco, Lima, Peru.
| | - Alberto Chinney-Herrera
- Facultad de Medicina, Universidad Nacional Autonoma de Mexico-UNAM, Ciudad Universitaria, Coyoacan, Ciudad de Mexico, Mexico
| |
Collapse
|
3
|
Lujan DA, Ochoa JL, Beswick EJ, Howard TA, Hathaway HJ, Perrone-Bizzozero NI, Hartley RS. Cold-Inducible RNA Binding Protein Impedes Breast Tumor Growth in the PyMT Murine Model for Breast Cancer. Biomedicines 2024; 12:340. [PMID: 38397942 PMCID: PMC10886683 DOI: 10.3390/biomedicines12020340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
RNA binding proteins (RBPs) post-transcriptionally regulate gene expression by associating with regulatory sequences in the untranslated regions of mRNAs. Cold-inducible RBP (CIRP) is a stress-induced RBP that was recently shown to modulate inflammation in response to cellular stress, where it increases or decreases pro-tumorigenic (proinflammatory) cytokines in different contexts. CIRP expression is altered in several cancers, including breast cancer, but the effects of CIRP on inflammation in breast cancer is not known. Here, we investigate if CIRP alters growth and the inflammatory profile of breast tumors. Transgenic mice overexpressing CIRP in the mammary epithelium were crossed with the PyMT mouse model of breast cancer, and the effects on both early and late tumorigenesis and inflammation were assessed. The effects of CIRP knockdown were also assessed in Py2T cell grafts. Overexpression of CIRP led to decreased tumorigenesis in the PyMT mouse model. Conversely, the knockdown of CIRP in Py2T cell grafts led to increased tumor growth. Luminex cytokine assays assessed the effects on the inflammatory environment. CIRP/PyMT mammary glands/mammary tumors and serum had decreased cytokines that promote inflammation, angiogenesis, and metastasis compared to PyMT mammary glands and serum, documenting a shift towards an environment less supportive of tumorigenesis. CIRP overexpression also decreased CD4+ helper T cells and increased CD8+ cytotoxic T cells in mammary tumors. Overall, these data support a role for CIRP as a potent antitumor molecule that suppresses both local and systemic pro-tumorigenic inflammation.
Collapse
Affiliation(s)
- Daniel A. Lujan
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Joey L. Ochoa
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Ellen J. Beswick
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA;
| | - Tamara A. Howard
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Helen J. Hathaway
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
| | - Rebecca S. Hartley
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| |
Collapse
|
4
|
Zubair M, Khalil S, Rasul I, Nadeem H, Noor F, Ahmad S, Alrumaihi F, Allemailem KS, Almatroudi A, Alshehri FF, Alshehri ZS. Integrated molecular modeling and dynamics approaches revealed potential natural inhibitors of NF-κB transcription factor as breast cancer therapeutics. J Biomol Struct Dyn 2023; 41:14715-14729. [PMID: 37301608 DOI: 10.1080/07391102.2023.2214209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/08/2023] [Indexed: 06/12/2023]
Abstract
Breast cancer is a silent killer malady among women and a serious economic burden in health care management. A case of breast cancer is diagnosed among women every 19 s, and every 74 s, a woman dies of breast cancer somewhere in the world. Despite the pop-up of progressive research, advanced treatment approaches, and preventive measures, breast cancer remains amplifying ailment. The nuclear factor kappa B (NF-κB) is a key transcription factor that links inflammation with cancer and is demonstrated as being involved in the tumorigenesis of breast cancer. The NF-κB transcription factor family in mammals consists of five proteins; c-Rel, RelA(p65), RelB, NF-κB1(p50), and NF-κB2(p52). The antitumor effect of NF-κB has also been explored in breast cancer, however, the actual treatment for breast cancer is yet to be discovered. This study is attributed to the identification of novel drug targets against breast cancer by targeting c-Rel, RelA(p65), RelB, NF-κB1(p50), and NF-κB2(p52) proteins. To identify the putative active compounds, a structure-based 3D pharmacophore model to the protein active site cavity was generated followed by virtual screening, molecular docking, and molecular dynamics (MD) simulation. Initially, a library of 45000 compounds were docked against the target protein and five compounds namely Z56811101, Z653426226, Z1097341967, Z92743432, and Z464101066 were selected for further analysis. The relative binding affinity of Z56811101, Z653426226, Z1097341967, Z92743432, and Z464101066 with NF-κB1 (p50), NF-κB2 (p52), RelA (p65), RelB, and c-Rel proteins were -6.8, -8, -7.0, -6.9, and -7.2 kcal/mol, respectively which remained stable throughout the simulations of 200 ns. Furthermore, all of these compounds depict maximum drug-like properties. Therefore, the proposed compounds can be a potential candidate for patients with breast cancer, but, experimental validation is needed to ensure their safety.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Sidra Khalil
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Ijaz Rasul
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Habibullah Nadeem
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Fatima Noor
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Faez Falah Alshehri
- College of Applied Medical Sciences, Shaqra University, Aldawadmi, Saudi Arabia
| | - Zafer Saad Alshehri
- College of Applied Medical Sciences, Shaqra University, Aldawadmi, Saudi Arabia
| |
Collapse
|
5
|
The Combination of Immune Checkpoint Blockade with Tumor Vessel Normalization as a Promising Therapeutic Strategy for Breast Cancer: An Overview of Preclinical and Clinical Studies. Int J Mol Sci 2023; 24:ijms24043226. [PMID: 36834641 PMCID: PMC9964596 DOI: 10.3390/ijms24043226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have a modest clinical activity when administered as monotherapy against breast cancer (BC), the most common malignancy in women. Novel combinatorial strategies are currently being investigated to overcome resistance to ICIs and promote antitumor immune responses in a greater proportion of BC patients. Recent studies have shown that the BC abnormal vasculature is associated with immune suppression in patients, and hampers both drug delivery and immune effector cell trafficking to tumor nests. Thus, strategies directed at normalizing (i.e., at remodeling and stabilizing) the immature, abnormal tumor vessels are receiving much attention. In particular, the combination of ICIs with tumor vessel normalizing agents is thought to hold great promise for the treatment of BC patients. Indeed, a compelling body of evidence indicates that the addition of low doses of antiangiogenic drugs to ICIs substantially improves antitumor immunity. In this review, we outline the impact that the reciprocal interactions occurring between tumor angiogenesis and immune cells have on the immune evasion and clinical progression of BC. In addition, we overview preclinical and clinical studies that are presently evaluating the therapeutic effectiveness of combining ICIs with antiangiogenic drugs in BC patients.
Collapse
|
6
|
Wei X, Yang M. Cell- and subcellular organelle-targeting nanoparticle-mediated breast cancer therapy. Front Pharmacol 2023; 14:1180794. [PMID: 37089933 PMCID: PMC10117787 DOI: 10.3389/fphar.2023.1180794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Breast cancer (BC) is the most prevalent malignant tumor, surpassing lung cancer as the most frequent malignancy in women. Drug resistance, metastasis, and immune escape are the major factors affecting patient survival and represent a huge challenge in BC treatment in clinic. The cell- and subcellular organelle-targeting nanoparticles-mediated targeted BC therapy may be an effective modality for immune evasion, metastasis, and drug resistance. Nanocarriers, efficiently delivering small molecules and macromolecules, are used to target subcellular apparatuses with excellent targeting, controlled delivery, and fewer side effects. This study summarizes and critically analyzes the latest organic nanoparticle-mediated subcellular targeted therapeutic based on chemotherapy, gene therapy, immunotherapy, and combination therapy in detail, and discusses the challenges and opportunities of nanoparticle therapy.
Collapse
Affiliation(s)
- Xue Wei
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Ming Yang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Ming Yang,
| |
Collapse
|
7
|
Sweeney C, Lazennec G, Vogel CFA. Environmental exposure and the role of AhR in the tumor microenvironment of breast cancer. Front Pharmacol 2022; 13:1095289. [PMID: 36588678 PMCID: PMC9797527 DOI: 10.3389/fphar.2022.1095289] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Activation of the aryl hydrocarbon receptor (AhR) through environmental exposure to chemicals including polycyclic aromatic hydrocarbons (PAHs) and polychlorinated dibenzo-p-dioxins (PCDDs) can lead to severe adverse health effects and increase the risk of breast cancer. This review considers several mechanisms which link the tumor promoting effects of environmental pollutants with the AhR signaling pathway, contributing to the development and progression of breast cancer. We explore AhR's function in shaping the tumor microenvironment, modifying immune tolerance, and regulating cancer stemness, driving breast cancer chemoresistance and metastasis. The complexity of AhR, with evidence for both oncogenic and tumor suppressor roles is discussed. We propose that AhR functions as a "molecular bridge", linking disproportionate toxin exposure and policies which underlie environmental injustice with tumor cell behaviors which drive poor patient outcomes.
Collapse
Affiliation(s)
- Colleen Sweeney
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Gwendal Lazennec
- Centre National de la Recherche Scientifique, SYS2DIAG-ALCEN, Cap Delta, Montpellier, France
| | - Christoph F. A. Vogel
- Center for Health and the Environment, University of California Davis, Davis, CA, United States
- Department of Environmental Toxicology, University of California Davis, Davis, CA, United States
| |
Collapse
|
8
|
Villarreal-García V, Estupiñan-Jiménez JR, Vivas-Mejía PE, Gonzalez-Villasana V, Vázquez-Guillén JM, Reséndez-Pérez D. A vicious circle in breast cancer: The interplay between inflammation, reactive oxygen species, and microRNAs. Front Oncol 2022; 12:980694. [PMID: 36226048 PMCID: PMC9548555 DOI: 10.3389/fonc.2022.980694] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women worldwide. This highly heterogeneous disease is molecularly stratified into luminal A, luminal B, HER2, triple-negative/basal-like, and normal-like subtypes. An important aspect in BC progression is the activation of inflammatory processes. The activation of CD8+/Th1, NK, and M1 tumor associated macrophages (TAMs), leads to tumor destruction. In contrast, an anti-inflammatory response mediated by CD4+/Th2 and M2 TAMs will favor tumor progression. Inflammation also stimulates the production of inflammatory mediators like reactive oxygen species (ROS). In chronic inflammation, ROS activates oxidative stress and endothelial dysfunction. In cancer, ROS plays a dual role with anti-tumorigenic and pro-tumorigenic effects in cell signaling pathways that control proliferation, survival, apoptosis, and inflammation. MicroRNAs (miRNAs), which are known to be involved in BC progression and inflammation, can be regulated by ROS. At the same time, miRNAs regulate the expression of genes modulating oxidative stress. In this review, we will discuss the interplay between inflammation, ROS, and miRNAs as anticancer and tumor promoter molecules in BC. A clear understanding of the role of miRNAs in the regulation of ROS production and inflammation, may lead to new opportunities for therapy in BC.
Collapse
Affiliation(s)
- Valeria Villarreal-García
- Departmento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - José Roberto Estupiñan-Jiménez
- Departmento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Pablo E. Vivas-Mejía
- Department of Biochemestry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
- Comprehensive Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Vianey Gonzalez-Villasana
- Departmento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - José Manuel Vázquez-Guillén
- Departamento de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Diana Reséndez-Pérez
- Departmento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
- Departamento de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| |
Collapse
|
9
|
Hu Y, Ding J, Wu C, Gao H, Ge M, Shao Q, Liu Y, Ye Q. Differential Expression and Prognostic Correlation of Immune Related Factors Between Right and Left Side Colorectal Cancer. Front Oncol 2022; 12:845765. [PMID: 35936748 PMCID: PMC9353740 DOI: 10.3389/fonc.2022.845765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Growing evidence suggests that colorectal cancer (CRC) should be considered a heterogeneous disease. The right side (RCC) and left side (LCC) colorectal cancer have different clinical characteristics and immune landscapes. The aim of this study was to analyze differential expression and prognostic correlation of immune-related factors between RCC and LCC. Methods The gene expression profile and clinical characteristics of CRC patients were retrieved from The Cancer Genome Atlas data portal (n=525). Using a deconvolution algorithm, immune cell infiltration in RCC and LCC based on the RNA-seq data was analyzed. Differentially expressed genes (DEGs) were obtained by performing differential gene expression analysis. Immune-related DEGs were derived by the intersection with immune-related factors downloaded from the IMMPORT database. To further validate the findings, we applied immunohistochemical (IHC) staining of a CRC tissue microarray (TMA). The distribution of immune cells in RCC and LCC and changes in the expression of immune molecules on their membranes were verified. The expression levels of circulating cytokines were measured by flow cytometry to detect the cytokines secreted by immune cells in RCC and LCC. Furthermore, to reveal the prognostic value of differential immune factors on RCC and LCC patients, survival analysis based on mRNA levels using TCGA cohort and survival analysis using protein levels was performed using our CRC patients. Results The infiltration of immune cells differed between RCC and LCC, the infiltration degree of macrophages M0 was significantly higher in LCC, while the infiltration degree of differentiated macrophages M1 and M2, CD4+ T and CD8+ T cells was significantly higher in RCC. The expression of related molecules by immune cells also differed between RCC and LCC. The expression of 7 genes in RCC was higher than that in LCC, which were CCR5, CD209, CD8A, HCK, HLA-DPB1, HLA-DQA1, HLA-DRA, respectively. Meanwhile, the expression of 2 genes in LCC was higher than in RCC, which were IL-34 and PROCR. Patients with RCC having high expression of HLA-DQA1 mRNA or proteins had better survival and LCC patients with high expression of IL 34 mRNA or protein had better survival. Conclusions In this study, we comprehensively compared differences in immune cells and regulating factors between left and right colorectal cancer. Different expression patterns and their effects on survival were identified. The analysis of immune-related factors may provide a theoretical basis for precise immunotherapy of RCC and LCC.
Collapse
Affiliation(s)
- Yue Hu
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jie Ding
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chengjiang Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Gao
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Meiling Ge
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qixiang Shao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yanhong Liu
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Yanhong Liu, ; Qing Ye,
| | - Qing Ye
- Department of Pathology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Yanhong Liu, ; Qing Ye,
| |
Collapse
|
10
|
Mohammed AK. Comparison of TNF-α and IL-19 concentrations at different stages of breast cancer. J Med Life 2022; 15:845-849. [PMID: 35928364 PMCID: PMC9321485 DOI: 10.25122/jml-2021-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/14/2022] [Indexed: 11/17/2022] Open
Abstract
This study investigated the alteration of tumour necrosis factor (TNF-α) and interleukin-19 (IL-19) at different clinical disease stages, lymph node metastasis, and ductal carcinoma in women with breast cancer. Serum samples were collected from 90 individuals with an age range of 25-61 years. These individuals were divided into a control group (healthy people), consisting of 31 individuals, and a breast cancer patients (BCP) group, consisting of 59 individuals. The pathological data (tumour stage, lymph node metastasis, and ductal carcinoma) was obtained from the medical record of patients and confirmed by experienced histopathology. Enzyme-linked immunosorbent assay (ELISAs) technology was used to measure the serum concentrations of IL-19 and TNF-α. The results showed significant differences (P≤0.002) in the mean of BMI, interleukin-19, and TNF-α in BCP compared to controls, while the age factor did not play an important role. The stages of breast cancer caused clear differences in the levels of TNF-α and IL-19. According to the findings, BCPs had a greater level of TNF-α in lymph node metastases than healthy persons. The concentration of serum IL-19 in BCP with lymph node metastasis was significantly different in non-lymph node metastasis patients and healthy people. Additionally, BCP with ductal carcinoma showed significant differences in the mean levels of IL-19 and TNF-α in comparison with healthy people.
Collapse
Affiliation(s)
- Amera Kamal Mohammed
- Clinical Laboratory Sciences Department, College of Pharmacy, University of Kirkuk, Kirkuk, Iraq,Corresponding Author: Amera Kamal Mohammed, Clinical Laboratory Sciences Department, College of Pharmacy, University of Kirkuk, Kirkuk, Iraq. E-mail:
| |
Collapse
|
11
|
Shi J, Shen X, Kang Q, Yang X, Denzinger M, Kornmann M, Traub B. Loss of Interleukin-13-Receptor-Alpha-1 Induces Apoptosis and Promotes EMT in Pancreatic Cancer. Int J Mol Sci 2022; 23:3659. [PMID: 35409019 PMCID: PMC8998778 DOI: 10.3390/ijms23073659] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022] Open
Abstract
In search of new therapies for pancreatic cancer, cytokine pathways have attracted increasing interest in recent years. Cytokines play a vital role in the crosstalk between tumour cells and the tumour microenvironment. The related inflammatory cytokines IL-4 and IL-13 can regularly be detected at increased levels in the microenvironment of pancreatic cancer. They share a receptor heterodimer consisting of IL-4Rα and IL-13Rα1. While IL-4Rα induces a more oncogenic phenotype, the role of IL-13Rα1 was yet to be determined. ShRNA-based knockdown of IL-13Rα1 was performed in Capan-1 and MIA PaCa-2. We assessed cell growth and migratory capacities under the influence of IL-13Rα1. Pathway alterations were detected by immunoblot analysis. We now have demonstrated that the loss of IL-13Rα1 induces apoptosis in pancreatic cancer cells. This was associated with an epithelial-to-mesenchymal transition. Loss of IL-13Rα1 also abolished the effects of exogenous IL-4 and IL-13 stimulation. Interestingly, in wild type cells, cytokine stimulation caused a similar increase in migratory capacities as after IL-13Rα1 knockdown. Overall, our results indicate the vital role of IL-13Rα1 in the progression of pancreatic cancer. The differential expression of IL-4Rα and IL-13Rα1 has to be taken into account when considering a cytokine-targeted therapy in pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Benno Traub
- Department of General and Visceral Surgery, Ulm Univsersity Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (J.S.); (X.S.); (Q.K.); (X.Y.); (M.D.); (M.K.)
| |
Collapse
|
12
|
Mohammad Mirzaei N, Changizi N, Asadpoure A, Su S, Sofia D, Tatarova Z, Zervantonakis IK, Chang YH, Shahriyari L. Investigating key cell types and molecules dynamics in PyMT mice model of breast cancer through a mathematical model. PLoS Comput Biol 2022; 18:e1009953. [PMID: 35294447 PMCID: PMC8959189 DOI: 10.1371/journal.pcbi.1009953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/28/2022] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
The most common kind of cancer among women is breast cancer. Understanding the tumor microenvironment and the interactions between individual cells and cytokines assists us in arriving at more effective treatments. Here, we develop a data-driven mathematical model to investigate the dynamics of key cell types and cytokines involved in breast cancer development. We use time-course gene expression profiles of a mouse model to estimate the relative abundance of cells and cytokines. We then employ a least-squares optimization method to evaluate the model's parameters based on the mice data. The resulting dynamics of the cells and cytokines obtained from the optimal set of parameters exhibit a decent agreement between the data and predictions. We perform a sensitivity analysis to identify the crucial parameters of the model and then perform a local bifurcation on them. The results reveal a strong connection between adipocytes, IL6, and the cancer population, suggesting them as potential targets for therapies.
Collapse
Affiliation(s)
- Navid Mohammad Mirzaei
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Navid Changizi
- Department of Civil and Environmental Engineering, University of Massachusetts, Dartmouth, Massachusetts, United States of America
| | - Alireza Asadpoure
- Department of Civil and Environmental Engineering, University of Massachusetts, Dartmouth, Massachusetts, United States of America
| | - Sumeyye Su
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Dilruba Sofia
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Zuzana Tatarova
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon, United States of America
| | - Ioannis K. Zervantonakis
- Department of Bioengineering, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Young Hwan Chang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon, United States of America
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| |
Collapse
|
13
|
Reduction on Proinflammatory Cytokines after Application of Transcutaneous Electrical Nerve Stimulation (TENS) in Patients with a Breast Cancer: A Nonrandomized, Open, and Single-Arm Study Protocol with Paired Analysis. Mediators Inflamm 2022; 2022:1350813. [PMID: 35241969 PMCID: PMC8886802 DOI: 10.1155/2022/1350813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 12/15/2021] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
Background. Transcutaneous electrical nerve stimulation (TENS) has been used as analgesic therapy in many diseases. It is already known that studies that have observed the relationship between pain and cytokines have found that patients who report less severe pain have less production of proinflammatory cytokines. However, one another accepted mechanism is that decreasing proinflammatory cytokines results in decreased pain intensity. Analyzing the literature, the authors describe that, in addition to the analgesic effect, TENS has shown systemic effects, and clinically, the reduction of proinflammatory cytokines could be a protective factor against inflammation. To test the inflammatory effect of TENS, we researched the literature for clinical conditions that suggest that proinflammatory cytokines are one of the main mediators of the disease process. Chronic inflammation is one of the risk factors mentioned for the development of a new cancer; at the same time, it is indicated as an indicator of the worst prognosis. Studies also suggest that the worst prognosis of breast cancer, one of the types with the highest incidence in the world, may be related to increased inflammatory activity. Considering that inflammation is increased in breast cancer and that TENS can reduce proinflammatory cytokines even without blocking the pain pathway, our hypothesis is that the anti-inflammatory effect of TENS can bring benefits to these patients. The aim of this study will be to evaluate the effect of TENS on blood reduction of proinflammatory cytokines in breast cancer patients. Methods. This study will evaluate at least 59 patients, over 18 years of age, diagnosed with breast cancer, but who have not yet started any treatment. All patients will be submitted to TENS intervention (Ibramed, Model Neurodyn III, parameters: VIF—turn on, frequency—2-247 Hz, pulse size—50-500 μs, and intensity (mA)—maximum tolerated by the patient), and the data will be analyzed in the pre- and postintervention of each patient. The application has a total duration of 30 minutes, and 8 ml of blood will be collected before and after the intervention. Proinflammatory (IL-1, IL-2, IL-6, IL-7, and TNF-α) and anti-inflammatory (IL-4, IL-10, IL-13, and FTCβ) cytokines will be analyzed. As a primary endpoint, we will analyze the reduction in blood concentration of proinflammatory cytokines, and as secondary endpoints, we will analyze the size of the effect according to each type of proinflammatory cytokine, describe the effect size of the reduction according to the breast cancer immunohistochemistry, and analyze the effect of TENS on anti-inflammatory cytokines. This study is approved by the Research Ethics Committee (Centro Universitário FMABC, Brazil) and registered in the Brazilian Clinical Trials (Search text: RBR-10jbwh47).
Collapse
|
14
|
He J, Zhong Y, Sun Y, Xie C, Yu T. Construction of an immune-related prognostic model by exploring the tumor microenvironment of clear cell renal cell carcinoma. Anal Biochem 2022; 643:114567. [PMID: 35122734 DOI: 10.1016/j.ab.2022.114567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE In this study, bioinformatics methods were performed to screen the candidate prognosis-related genes of clear cell renal cell carcinoma (ccRCC) by analyzing the tumor microenvironment (TME). METHODS Gene expression and clinical data of ccRCC patients were accessed from TCGA, and R package ESTIMATE was applied to calculate immune, stromal, and ESTIMATE scores of the patients. Survival analysis was conducted per median of these three scores. Based on the scoring results, differentially expressed genes (DEGs) were screened. Regression algorithms were utilized to screen prognostic genes and establish a risk model. Finally, pathway activity differences were analyzed through GSEA. RESULTS Patients with the unfavorable prognosis had high immune scores. 619 DEGs (499 up-regulated and 120 down-regulated) were screened based on the differences in gene expression of the patients with high and low immune scores. These genes mainly participated in immune-related signaling pathways. A prognostic risk model for ccRCC patients was constructed and 7 immune-related signature genes (RORB, TNFSF14, UCN2, USP2, TOX3, KLRC2, SLAMF9) were obtained through regression analysis. The constructed prognostic risk model could be used for determining prognoses of patients with ccRCC. CONCLUSION We unraveled the association between TME and prognosis of ccRCC patients and established a prognostic risk model based on the differentially expressed genes. These results contributed to understanding of TME that affected patients' prognosis and progression of ccRCC and conduced to finding potential biomarkers of ccRCC.
Collapse
Affiliation(s)
- Jia He
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Yun Zhong
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Yanli Sun
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Chao Xie
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Tianqiang Yu
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China.
| |
Collapse
|
15
|
Ye Y, Xu C, Chen F, Liu Q, Cheng N. Targeting Innate Immunity in Breast Cancer Therapy: A Narrative Review. Front Immunol 2021; 12:771201. [PMID: 34899721 PMCID: PMC8656691 DOI: 10.3389/fimmu.2021.771201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023] Open
Abstract
Although breast cancer has been previously considered "cold" tumors, numerous studies are currently conducted to explore the great potentials of immunotherapies in improving breast cancer patient outcomes. In addition to the focus on stimulating adaptive immunity for antitumor responses, growing evidence showed the importance of triggering host innate immunity to eradicate established tumors and/or control tumor metastasis of breast cancer. In this review, we first briefly introduce the breast tumor immune microenvironment. We also discuss innate immune targets and pathways and mechanisms of their synergy with the adaptive antitumor response and other treatment strategies. Lastly, we review clinical trials targeting innate immune pathways for breast cancer therapies.
Collapse
Affiliation(s)
- Yanqi Ye
- Zenomics. Inc. Magnify at California NanoSystems Institute, Los Angeles, CA, United States
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, QLD, Australia
| | - Fengqian Chen
- School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Qi Liu
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ning Cheng
- Department of Otolaryngology - Head and Neck Surgery, University of California at San Francisco, San Francisco, CA, United States
| |
Collapse
|
16
|
Malara FA, Matos DC, Ribeiro LCA, Falcoski TOR, Andrade TJAS, Santos VNC, Lima NM, Carlos IZ. Medicinal Plants from Brazilian Cerrado Biome: Potential sources of new anti-inflammatory compounds and antitumor agents on Ehrlich carcinoma. AN ACAD BRAS CIENC 2021; 93:e20191101. [PMID: 34730736 DOI: 10.1590/0001-3765202120191101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 05/26/2020] [Indexed: 11/21/2022] Open
Abstract
This work describes a pharmacological screening of Brazilian medicinal plants through their anti-inflammatory and cytotoxicity activities. Cytotoxicity activity of Mouriri elliptica and Alchornea glandulosa as well as the drugs celecoxib and doxorubicin were evaluated in cultures of peritoneal macrophages. The immune system influence of these samples was analyzed by determining production/inhibition of NO, production of tumor necrosis factor-α and production of interleukin-10. Regarding the production/inhibition of NO, there was NO production by M. elliptica and NO inhibition when the cells were exposed to A. glandulosa; Macrophages generally produce more NO, plus TNF-α and less IL-10, when associated to the tumor phenomenon, characterizing the inflammation involved in cancer. A. glandulosa showed anti-inflammatory effect, inhibited NO production and it was associated with low TNF-α production, although not as low as the macrophages associated with celecoxib and doxorubicin. These cytokines were not different in animals with tumor. Celecoxib confirms its anti-inflammatory action by markedly inhibiting NO and TNF-α, but also inhibiting IL-10 which is an anti-inflammatory cytokine. Doxorubicin inhibited NO in a higher percentage in the group of animals with tumor, although the literature reports that this drug stimulates the production of NO and this collaborates with its cytotoxic effect.
Collapse
Affiliation(s)
- Fábio A Malara
- Universidade Estadual Paulista "Júlio de Mesquita Filho"/UNESP, Faculdade de Ciências Farmacêuticas, Rod. Araraquara-Jaú Km 1, Machados, 14800-901 Araraquara, SP, Brazil
| | - Djamile C Matos
- Universidade Estadual Paulista "Júlio de Mesquita Filho"/UNESP, Faculdade de Ciências Farmacêuticas, Rod. Araraquara-Jaú Km 1, Machados, 14800-901 Araraquara, SP, Brazil
| | - Lívia C A Ribeiro
- Universidade Estadual Paulista "Júlio de Mesquita Filho"/UNESP, Faculdade de Ciências Farmacêuticas, Rod. Araraquara-Jaú Km 1, Machados, 14800-901 Araraquara, SP, Brazil
| | - Thais O R Falcoski
- Universidade Estadual Paulista "Júlio de Mesquita Filho"/UNESP, Faculdade de Ciências Farmacêuticas, Rod. Araraquara-Jaú Km 1, Machados, 14800-901 Araraquara, SP, Brazil
| | - Teresinha J A S Andrade
- Universidade Estadual Paulista "Júlio de Mesquita Filho"/UNESP, Faculdade de Ciências Farmacêuticas, Rod. Araraquara-Jaú Km 1, Machados, 14800-901 Araraquara, SP, Brazil
| | - Vanessa N C Santos
- Universidade Federal do Amazonas/UFAM, Instituto de Ciências Biológicas/ICB, Av. General Rodrigo Octavio Jordão Ramos, 1200, Coroado I, 69067-005 Manaus, AM, Brazil
| | - Nerilson M Lima
- Universidade Federal de Juiz de Fora/UFJF, Instituto de Ciências Exatas/ICE, Rua José Lourenço Kelmer, s/n, São Pedro, 36036-900 Juiz de Fora, MG, Brazil
| | - Iracilda Z Carlos
- Universidade Estadual Paulista "Júlio de Mesquita Filho"/UNESP, Faculdade de Ciências Farmacêuticas, Rod. Araraquara-Jaú Km 1, Machados, 14800-901 Araraquara, SP, Brazil
| |
Collapse
|
17
|
Tobin AJ, Noel NP, Christian SL, Brown RJ. Lipoprotein lipase hydrolysis products induce pro-inflammatory cytokine expression in triple-negative breast cancer cells. BMC Res Notes 2021; 14:315. [PMID: 34404457 PMCID: PMC8369739 DOI: 10.1186/s13104-021-05728-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
Objectives Breast cancer cell growth and proliferation requires lipids for energy production, cell membrane synthesis, or as signaling molecules. Lipids can be delivered to cells by lipoprotein lipase (LPL), an extracellular lipase that hydrolyzes triacylglycerols and phospholipids from lipoproteins, that is expressed by adipose tissue and some breast cancer cell lines. Studies have shown that lipoprotein hydrolysis products induce pro-inflammatory cytokine secretion by endothelial cells. Thus, our objective was to determine if hydrolysis products generated by LPL from total lipoproteins can also promote pro-inflammatory cytokine secretion from breast cancer cells. Results Using cytokine arrays, we found that MDA-MB-231 cells increased secretion of seven cytokines in response to treatment with lipoprotein hydrolysis products. In contrast, MCF-7 cells showed decreased secretion of two cytokines. Expanding the analysis to additional cell lines by ELISA, we found increased secretion of TNF-α and IL-6 by MDA-MB-468 cells, and increased secretion of IL-4 by MDA-MB-468 and SKBR3 cells. The changes to cytokine secretion profiles of the breast cancer cell types examined, including the non-cancerous MCF-10a breast cells, were independent of increased cell metabolic activity. These results provide information on how lipoprotein hydrolysis products within the tumor microenvironment might affect breast cancer cell viability and progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-021-05728-z.
Collapse
Affiliation(s)
- Alexandria J Tobin
- Department of Biochemistry, Memorial University of Newfoundland, 232 Elizabeth Ave., St. John's, NL, A1B 3X9, Canada
| | - Nicholas P Noel
- Department of Biochemistry, Memorial University of Newfoundland, 232 Elizabeth Ave., St. John's, NL, A1B 3X9, Canada
| | - Sherri L Christian
- Department of Biochemistry, Memorial University of Newfoundland, 232 Elizabeth Ave., St. John's, NL, A1B 3X9, Canada.
| | - Robert J Brown
- Department of Biochemistry, Memorial University of Newfoundland, 232 Elizabeth Ave., St. John's, NL, A1B 3X9, Canada.
| |
Collapse
|
18
|
Sarangthem V, Yi A, Kim Y, Rehemtulla A, Lee BH, Jeon YH, Singh TD, Park RW. Therapeutic Effect of IL-4 Receptor-Targeting Pro-Apoptotic Peptide (AP1-ELP-KLAK) in Glioblastoma Tumor Model. Int J Nanomedicine 2021; 16:5039-5052. [PMID: 34335025 PMCID: PMC8318221 DOI: 10.2147/ijn.s316388] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/28/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Thermal-responsive self-assembled elastin-like polypeptide (ELP)-based nanoparticles are an emerging platform for controlled delivery of therapeutic peptides, proteins and small molecular drugs. The antitumor effect of bioengineered chimeric polypeptide AP1-ELP-KLAK containing an interleukin-4 receptor (IL-4R) targeting peptide and pro-apoptotic peptide (KLAKLAK) was evaluated in glioblastoma (GBM) in vitro and in vivo. METHODS AND RESULTS Herein, the therapeutic effect of AP1-ELP-KLAK was tested in advanced, and less curable glioblastoma cells with higher expression of IL-4R. Glioblastoma cell lines stably expressing different reporter systems i.e., caspase-3 sensor (surrogate marker for cellular apoptosis) or effluc/enhanced firefly luciferase (cellular viability) were established to measure cell death non-invasively. Bioluminescence imaging (BLI) of D54/effluc and U97MG/effluc treated with AP1-ELP-KLAK exhibited higher cell death up to 2~3-fold than the control. Treatment with AP1-ELP-KLAK resulted in time-dependent increase of caspase-3 sensor BLI activity in D54/C cells and D54/C tumor-bearing mice. Intravenous injection of AP1-ELP-KLAK dramatically reduced tumor growth by inducing cellular apoptosis in D54/effluc tumor-bearing mice. Further, the immuno-histological examination of the excised tumor tissue confirmed the presence of apoptotic cells as well as caspase-3 activation. CONCLUSION Collectively, AP1-ELP-KLAK effectively induced cellular apoptosis of glioblastoma cells and non-invasive imaging provides a window for real-time monitoring of anti-tumor effect with the provision of improving therapeutic efficacy in a glioblastoma mice model.
Collapse
Affiliation(s)
- Vijaya Sarangthem
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Aena Yi
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Yunjae Kim
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Young Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Republic of Korea
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| |
Collapse
|
19
|
Leonel ECR, Ruiz TFR, Bedolo CM, Campos SGP, Taboga SR. Inflammatory repercussions in female steroid responsive glands after perinatal exposure to bisphenol A and 17-β estradiol. Cell Biol Int 2021; 45:2264-2274. [PMID: 34288236 DOI: 10.1002/cbin.11665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/18/2022]
Abstract
The mammary gland (MG) and female prostate are plastic reproductive organs which are highly responsive to hormones. Thus, endocrine disruptors, such as bisphenol A (BPA) and exogenous estrogens, negatively affect glandular homeostasis. In addition to previously described alterations, changes in inflammatory markers expression also trigger the development of a microenvironment that contributes to tumor progression. The current work aimed to evaluate the inflammatory responses of the MG and prostate gland to BPA (50 µg/kg) and 17-β estradiol (35 µg/kg) exposure during the perinatal window of susceptibility. The results showed that at 6 months of age there was an increase in the number of phospho-STAT3 (P-STAT3) positive cells in the female prostate from animals perinatally exposed to 50 µg/kg BPA daily. In addition, the number of macrophages increased in these animals in comparison with nonexposed animals, as shown by the F4/80 marker. Despite an increase in the incidence of lobuloalveolar and intraductal hyperplasia, the MG did not show any difference in the expression of the four inflammatory markers evaluated: tumor necrosis factor-α, COX-2, P-STAT3, and F4/80. Analysis of both glands from the same animal led to the conclusion that exposure to endocrine disruptors during the perinatal window of susceptibility leads to different inflammatory responses in different reproductive organs. As the prostate is more susceptible to these inflammatory mechanisms, it is reasonable to affirm that possible neoplastic alterations in this organ are related to changes in the inflammatory pattern of the stroma, a characteristic that is not evident in the MG.
Collapse
Affiliation(s)
- Ellen Cristina Rivas Leonel
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil.,Department of Histology, Embriology, and Cell Biology, Institute of Biological Sciences (ICB III), Federal University of Goiás (UFG), Goiânia, Goiás, Brazil
| | - Thalles Fernando Rocha Ruiz
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Carolina Marques Bedolo
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Silvana Gisele Pegorin Campos
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
20
|
Yuan SJ, Wang C, Xu HZ, Liu Y, Zheng MY, Li K, Sun SK, Komatsu N, Zhao L, Chen X. Conjugation with nanodiamonds via hydrazone bond fundamentally alters intracellular distribution and activity of doxorubicin. Int J Pharm 2021; 606:120872. [PMID: 34246743 DOI: 10.1016/j.ijpharm.2021.120872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 12/18/2022]
Abstract
Doxorubicin (DOX) has been widely incorporated in various delivery forms for tareted treatment of malignant tumors such as triple-negative breast cancer (TNBC), with numerous studies reporting higher therapeutic efficacy and lower toxicity at the same time. However, little attention has been paid to whether DOX in a delivery form acts with the same actions and processes as in free form at the cellular level. This question was investigated in the present study wherein DOX conjugated with polyglycerol-coated nanodiamonds through the pH-sensitive hydrazone bond (Nano-DOX) was compared with DOX in free form on the 4T1 mouse TNBC model. We first found Nano-DOX to have a distinct intracellular distribution profile from DOX. Internalized Nano-DOX mainly stayed in the lysosomes slowly releasing DOX into the cytoplasm and then the nucleus whereas DOX displayed both nuclear and lysosomal distribution after cell uptake. Next, Nano-DOX was shown to induce endoplasmic reticulum (ER) stress without substantial DNA damage while DOX caused massive DNA damage as well as ER stress. Consequently, Nano-DOX only caused minimal activation of pro-inflammatory signaling mediated by MAPK/ERK, NF-κB and STAT3 as seen in response to DOX-inflicted DNA damage. Consistently, DOX-induced activities of ABC transporters, CXCL-1, GM-CSF and IL-6, which are tumor protective events downstream to the pro-inflammatory signaling, were also minimal in Nano-DOX-treated cancer cells. These findings are compelling proof that a chemotherapy in nano form can have distinct intracellular pharmacokinetics from its free from, which can result in altered cellular effects of the drug. Implications of these findings are discussed with an emphasis on nano-drug design, tumor pharmacology and chemoresistance.
Collapse
Affiliation(s)
- Shen-Jun Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No. 185, Wuhan 430072, China; Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Chao Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No. 185, Wuhan 430072, China
| | - Hua-Zhen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No. 185, Wuhan 430072, China
| | - Yan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No. 185, Wuhan 430072, China
| | - Mei-Yan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No. 185, Wuhan 430072, China
| | - Ke Li
- Demonstration Center for Experimental Basic Medicine Education, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Shi-Kuan Sun
- School of Material Science and Energy Engineering, Foshan University, Foshan, Guangdong 528000, China
| | - Naoki Komatsu
- Graduate School of Human and Environmental Studies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Li Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No. 185, Wuhan 430072, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430072, China.
| |
Collapse
|
21
|
Wang X, Priya Veeraraghavan V, Krishna Mohan S, Lv F. Anticancer and immunomodulatory effect of rhaponticin on Benzo(a)Pyrene-induced lung carcinogenesis and induction of apoptosis in A549 cells. Saudi J Biol Sci 2021; 28:4522-4531. [PMID: 34354438 PMCID: PMC8324936 DOI: 10.1016/j.sjbs.2021.04.052] [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: 03/29/2021] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 01/14/2023] Open
Abstract
In worldwide, one of the most important cancer-related death is lung cancer. Also has the highest mortality rate between various cancer types. The count of lung cancer occurrence is increasing with an increased frequency by smoking. Proficient chemoprevention approaches are needed to prevent the occurrence of lung cancer. Therefore, the aim of this exploration is to determine the therapeutic impact on the immune modulatory effect of rhaponticin on lung tumorigenesis in vivo and in vitro cytotoxicity effect in A549 cells of human lung cancer. Lung cancer tumorigenesis in mice was challenged with benzo(a)pyrene (BaP) with 50 mg/kg bodyweight (b.wt) as oral administration for 6 weeks (two times/week). Rhaponticin were given orally 30 mg/kg b.wt (two times/week) in BaP induced mice from 12 weeks to 18 weeks. After treatment completes, the body weight was measured and then blood, lung tissue was collected for various parameters detection. The results evidenced that BaP induced mice decreased the bodyweight, increased lung weight, increased tumor markers (AHH, CEA and LDH), and increased the proinflammatory cytokines. The enzyme catalase, superoxide dismutase activity was decreased and increased lipid peroxidation in immune comprising cells compared with the control cells. Moreover, rhaponticin treatment improves in chemical assays and also the histopathological alteration of lung tissues. The present findings provide evidence about the therapeutic potentials of rhaponticin against BaP triggered lung tumorigenesis.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Surapaneni Krishna Mohan
- Department of Biochemistry, Department of Clinical Skills & Simulation and Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, India
| | - Feng Lv
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
- Corresponding author.
| |
Collapse
|
22
|
Zhang C, Cui X, Feng L, Han Z, Peng D, Fu W, Xing Y. The deficiency of FKBP-5 inhibited hepatocellular progression by increasing the infiltration of distinct immune cells and inhibiting obesity-associated gut microbial metabolite. J Gastrointest Oncol 2021; 12:711-721. [PMID: 34012660 DOI: 10.21037/jgo-21-71] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Gut microbiota has a number of essential roles in nutrition metabolism and immune homeostasis, and is closely related to hepatocellular progression. In recent years, studies have also shown that FK506 binding protein 5 (FKBP-5) plays a crucial role in immune regulation. However, it is not yet clear whether FKBP-5 promotes the development of hepatocellular carcinoma (HCC) by affecting immune function and gut microbiota. Methods FKBP-5 expression was verified by immunochemistry and western blot and reverse transcription polymerase chain reaction (RT-qPCR) assays. After treatment in WT and FKBP-5-/- mice, the histological characteristic of mice liver tissue was assessed by H&E staining, and hepatic leukocytes and hepatic NKT cells were identified by flow cytometer. Meanwhile, primary bile acids (BAs), secondary BAs, serum total cholesterol, and the weight of abdomen adipose tissues were examined, and the gut microbiota was evaluated by 16S ribosomal ribonucleic acid (rRNA) sequencing. Results We discovered that FKBP-5 was highly expressed in HCC tissues. Meanwhile, FKBP-5 deletion inhibited tumor progression by increasing CD8+ T, CD4+ T, NKT and CD4+NKT cells in mice after diethylnitrosamine (DEN) injection. Besides, we proved that FKBP-5 deletion generated rapid and significant reductions in the intestinal BAs, the weight of abdomen adipose tissues and the serum total cholesterol. FKBP-5 deletion also led to a change in the composition of gut microbiota, suggesting that BAs are the main dietary factor regulating gut microbiota, which could be affected by FKBP-5 deletion. Further, we uncovered that anti-CD4 and anti-CD8 treatments facilitated hepatocellular progression by modulating gut microbiota composition in FKBP-5-/- mice. Conclusions Therefore, we demonstrated that FKBP-5 deletion inhibited hepatocellular progression by modulating immune response and gut microbiome-mediated BAs metabolism.
Collapse
Affiliation(s)
- Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Cui
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Lian Feng
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhiyi Han
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Deti Peng
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Wenjun Fu
- South China Research Center for Acupuncture and Moxibustion, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yufeng Xing
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
23
|
Hausmann LD, de Almeida BS, de Souza IR, Drehmer MN, Fernandes BL, Wilkens RS, Vieira DSC, Lofgren SE, Lindenau JDR, de Toledo E Silva G, Muniz YCN. Association of TNFRSF1A and IFNLR1 Gene Polymorphisms with the Risk of Developing Breast Cancer and Clinical Pathologic Features. Biochem Genet 2021; 59:1233-1246. [PMID: 33751344 DOI: 10.1007/s10528-021-10060-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 03/10/2021] [Indexed: 12/27/2022]
Abstract
Several genes have been associated with breast cancer (BC) susceptibility. The tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A), and interferon lambda receptor 1 (IFNLR1) genes encode receptors that mediate the action of inflammatory cytokines. Previous studies have demonstrated the association of the variants rs1800693 (TNFRSF1A) and rs4649203 (IFNLR1) with some inflammatory diseases. The present study aimed to verify a possible association of these variants with BC, its clinical pathologic features, as well as epidemiological data in a Brazilian population. A total of 243 patients and 294 individuals without history of BC were genotyped for these polymorphisms through TaqMan® SNP genotyping assays by qPCR. For the TNFRSF1A gene, no significant results were found. For IFNLR1, the AA genotype (p = 0.008) and the A allele (p = 0.02) were significantly associated with a lower risk of developing BC. When analyzing the age, it was observed that each increase of one year contributes to the development of BC (p < 0.001). Also, the smoking habit (p < 0.001) and body mass index (p = 0.018) increase the risk of disease development. Analyzing progesterone receptor factor an association was found with the AA genotype of the IFNLR1 (p = 0.02). The findings suggest that polymorphism in the immune-related IFNLR1 gene contribute to BC susceptibility in a Brazilian population. These findings can contribute to the further understanding of the role this gene and pathways in BC development.
Collapse
Affiliation(s)
- Leili Daiane Hausmann
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil.
| | - Bibiana Sgorla de Almeida
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| | - Ilíada Rainha de Souza
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| | - Manuela Nunes Drehmer
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| | - Braulio Leal Fernandes
- Polydoro Ernani de São, Thiago University Hospital (HU/UFSC), Florianópolis, 88036-800, Brazil
| | - Renato Salerno Wilkens
- Polydoro Ernani de São, Thiago University Hospital (HU/UFSC), Florianópolis, 88036-800, Brazil
| | | | - Sara Emelie Lofgren
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| | - Juliana Dal-Ri Lindenau
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| | - Guilherme de Toledo E Silva
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| | - Yara Costa Netto Muniz
- Department of Cell Biology, Embryology and Genetics (BEG), School of Biological Sciences (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, 88040-900, Brazil
| |
Collapse
|
24
|
Shi J, Song X, Traub B, Luxenhofer M, Kornmann M. Involvement of IL-4, IL-13 and Their Receptors in Pancreatic Cancer. Int J Mol Sci 2021; 22:ijms22062998. [PMID: 33804263 PMCID: PMC8000536 DOI: 10.3390/ijms22062998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 01/05/2023] Open
Abstract
Interleukin (IL)-4 and IL-13 are known as pleiotropic Th2 cytokines with a wide range of biological properties and functions especially in immune responses. In addition, increasing activities have also been determined in oncogenesis and tumor progression of several malignancies. It is now generally accepted that IL-4 and IL-13 can exert effects on epithelial tumor cells through corresponding receptors. Type II IL-4 receptor (IL-4Rα/IL-13Rα1), predominantly expressed in non-hematopoietic cells, is identified to be the main target for both IL-4 and IL-13 in tumors. Moreover, IL-13 can also signal by binding to the IL-13Rα2 receptor. Structural similarity due to the use of the same receptor complex generated in response to IL-4/IL-13 results in overlapping but also distinct signaling pathways and functions. The aim of this review was to summarize knowledge about IL-4 and IL-13 and their receptors in pancreatic cancer in order understand the implication of IL-4 and IL-13 and their receptors for pancreatic tumorigenesis and progression and for developing possible new diagnostic and therapeutic targets.
Collapse
|
25
|
Sigdel I, Gupta N, Faizee F, Khare VM, Tiwari AK, Tang Y. Biomimetic Microfluidic Platforms for the Assessment of Breast Cancer Metastasis. Front Bioeng Biotechnol 2021; 9:633671. [PMID: 33777909 PMCID: PMC7992012 DOI: 10.3389/fbioe.2021.633671] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 12/27/2022] Open
Abstract
Of around half a million women dying of breast cancer each year, more than 90% die due to metastasis. Models necessary to understand the metastatic process, particularly breast cancer cell extravasation and colonization, are currently limited and urgently needed to develop therapeutic interventions necessary to prevent breast cancer metastasis. Microfluidic approaches aim to reconstitute functional units of organs that cannot be modeled easily in traditional cell culture or animal studies by reproducing vascular networks and parenchyma on a chip in a three-dimensional, physiologically relevant in vitro system. In recent years, microfluidics models utilizing innovative biomaterials and micro-engineering technologies have shown great potential in our effort of mechanistic understanding of the breast cancer metastasis cascade by providing 3D constructs that can mimic in vivo cellular microenvironment and the ability to visualize and monitor cellular interactions in real-time. In this review, we will provide readers with a detailed discussion on the application of the most up-to-date, state-of-the-art microfluidics-based breast cancer models, with a special focus on their application in the engineering approaches to recapitulate the metastasis process, including invasion, intravasation, extravasation, breast cancer metastasis organotropism, and metastasis niche formation.
Collapse
Affiliation(s)
- Indira Sigdel
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Niraj Gupta
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Fairuz Faizee
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Vishwa M Khare
- Eurofins Lancaster Laboratories, Philadelphia, PA, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yuan Tang
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
26
|
Subia B, Dahiya UR, Mishra S, Ayache J, Casquillas GV, Caballero D, Reis RL, Kundu SC. Breast tumor-on-chip models: From disease modeling to personalized drug screening. J Control Release 2021; 331:103-120. [PMID: 33417986 PMCID: PMC8172385 DOI: 10.1016/j.jconrel.2020.12.057] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023]
Abstract
Breast cancer is one of the leading causes of mortality worldwide being the most common cancer among women. Despite the significant progress obtained during the past years in the understanding of breast cancer pathophysiology, women continue to die from it. Novel tools and technologies are needed to develop better diagnostic and therapeutic approaches, and to better understand the molecular and cellular players involved in the progression of this disease. Typical methods employed by the pharmaceutical industry and laboratories to investigate breast cancer etiology and evaluate the efficiency of new therapeutic compounds are still based on traditional tissue culture flasks and animal models, which have certain limitations. Recently, tumor-on-chip technology emerged as a new generation of in vitro disease model to investigate the physiopathology of tumors and predict the efficiency of drugs in a native-like microenvironment. These microfluidic systems reproduce the functional units and composition of human organs and tissues, and importantly, the rheological properties of the native scenario, enabling precise control over fluid flow or local gradients. Herein, we review the most recent works related to breast tumor-on-chip for disease modeling and drug screening applications. Finally, we critically discuss the future applications of this emerging technology in breast cancer therapeutics and drug development.
Collapse
Affiliation(s)
- Bano Subia
- Elvesys Microfluidics Innovation Centre, Paris 75011, France..
| | | | - Sarita Mishra
- CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India..
| | - Jessica Ayache
- Elvesys Microfluidics Innovation Centre, Paris 75011, France..
| | | | - David Caballero
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| | - Subhas C Kundu
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| |
Collapse
|
27
|
Liao WC, Yen HR, Chen CH, Chu YH, Song YC, Tseng TJ, Liu CH. CHPF promotes malignancy of breast cancer cells by modifying syndecan-4 and the tumor microenvironment. Am J Cancer Res 2021; 11:812-826. [PMID: 33791155 PMCID: PMC7994168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths in women worldwide. Several studies have indicated that abnormal chondroitin sulfate (CS) chains accumulate in breast cancer tissues; however, the functions and dysregulation of CS synthases are largely unknown. Here, we demonstrate that chondroitin polymerising factor (CHPF) is frequently upregulated in breast cancer tissues and that its high expression is positively associated with tumor metastasis, high stages, and short survival time. CHPF modulates CS formation in breast cancer cells. Additionally, we found that CHPF promotes tumor growth and metastasis accompanied by an increase in G-CSF levels and the number of myeloid-derived suppressor cells in tumor tissue. We revealed that tumor cell-derived G-CSF is co-localised with CS on the cell surface. Interestingly, our study is the first to identify that syndecan-4 (SDC4) is modified by CHPF and that it is involved in CHPF-mediated phenotypes. Moreover, breast cancer patients with high expression of both SDC4 and CHPF had worse overall survival compared to other subsets, which implied the synergistic effects of these two genes. In summary, our results indicated that the upregulation of CHPF in breast cancer contributes to the malignant behaviour of cancer cells, thereby providing novel insights on the significance of CHPF-modified SDC4 in breast cancer pathogenesis.
Collapse
Affiliation(s)
- Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| | - Hung-Rong Yen
- School of Chinese Medicine, China Medical UniversityTaichung, Taiwan
- Department of Chinese Medicine, China Medical University HospitalTaichung, Taiwan
- Chinese Medicine Research Center, China Medical UniversityTaichung, Taiwan
| | - Chia-Hua Chen
- Molecular Medicine Research Center, Chang Gung UniversityTaiwan
| | - Yin-Hung Chu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
| | - Ying-Chyi Song
- School of Chinese Medicine, China Medical UniversityTaichung, Taiwan
- Chinese Medicine Research Center, China Medical UniversityTaichung, Taiwan
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical UniversityTaichung, Taiwan
| | - To-Jung Tseng
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| | - Chiung-Hui Liu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| |
Collapse
|
28
|
Yin L, Chen L, Qi Z, Li J, Wang X, Ma K, Liu X. Gene expression-based immune infiltration analyses of liver cancer and their associations with survival outcomes. Cancer Genet 2021; 254-255:75-81. [PMID: 33647815 DOI: 10.1016/j.cancergen.2021.02.001] [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: 10/28/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 12/09/2022]
Abstract
INTRODUCTION Liver cancer is one of the most lethal malignancies, presenting inferior survival outcomes once diagnosed. Current therapeutic approaches mainly target the tumor cells or vasculature, but rarely take the immune factors into consideration. METHODS In our study, the compositions of tumor-infiltrating immune cells (TIICs) in liver cancer and paracancer samples were analyzed based on the gene expression profiles by CIBERSORT. After calculating the proportions of 22 TIICs subtypes in 51 paired cancer and paracancer samples, we found their proportions varied between intragroup and intergroup. Compared with the paracancer tissues, the relative proportions of macrophages M0 and resting mast cells in liver cancer samples were significantly elevated, while that of M2 macrophages were reduced. RESULTS Univariate Cox regression analysis with the 22 TIICs subtypes as continuous variables showed increased B cells memory and resting NK cells were significantly associated with poor survival outcome. Besides, hierarchical clustering analysis based on the proportions of 22 TIICs subtypes identified 3 clusters, which exhibited distinct prognosis. Among them, cluster 1 had superior survival outcomes, while cluster 3 had inferior survival outcomes. CONCLUSIONS Collectively, our research suggested certain TIICs subpopulations proportions, as well as cluster patterns were associated with the prognosis of liver cancer, which provided potential therapeutic targets for liver cancer.
Collapse
Affiliation(s)
- Liang Yin
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| | - Lei Chen
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| | - Zilong Qi
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| | - Jinmin Li
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| | - Xinning Wang
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| | - Kun Ma
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| | - Xiangyang Liu
- Department of Pediatric Surgery, Cangzhou Central Hospital, Xinhua Road, Yunhe District, Cangzhou, Heibei 061000, China
| |
Collapse
|
29
|
Lu G, Qiu Y, Su X. Targeting CXCL12-CXCR4 Signaling Enhances Immune Checkpoint Blockade Therapy Against Triple Negative Breast Cancer. Eur J Pharm Sci 2021; 157:105606. [PMID: 33131745 DOI: 10.1016/j.ejps.2020.105606] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/21/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Insufficient T cell infiltration in triple-negative breast cancer (TNBC) has limited its response rate to immune checkpoint blockade (ICB) therapies and motivated the development of immunostimulatory approaches to enhance the ICB therapy. CXCR4 is a chemokine receptor highly upregulated both on cell surface and cytoplasm in tumor tissues. Activating CXCR4 has been associated with increased immunosuppression in the tumor microenvironment. Here, we developed a CXCR4-targeted liposomal formulation (Liposomal-AMD3100) to enhance therapeutic efficacy of AMD3100, a CXCR4 antagonist. Particularly, AMD3100 is not only encapsulated into the liposome but coated on the surface of the formulation to serve as a targeting moiety and a dual blocker capable of inhibiting CXCR4 activation extracellularly and intracellularly. The Liposomal-AMD3100 remodeled both immune and stromal microenvironment more efficiently compared with free AMD3100, indicating better pharmacodynamic profile of AMD3100 achieved by liposomal formulation. The combination of anti-PD-L1 with Liposomal-AMD3100 formulation exhibited an increased antitumor effect and prolonged survival time compared with monotherapies in a murine TNBC model (4T1). This work proves that immune activation via liposomal delivery of CXCR4 inhibitors has a great potential to expand ICB therapies to originally ICB-insensitive cancer types.
Collapse
Affiliation(s)
- Guowen Lu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China.
| | - Yier Qiu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| | - Xiaobao Su
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| |
Collapse
|
30
|
Luo Z, Chen X, Zhang Y, Huang Z, Zhao H, Zhao J, Li Z, Zhou J, Liu J, Cai J, Bi X. Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy. Front Cell Dev Biol 2021; 8:577125. [PMID: 33585439 PMCID: PMC7876250 DOI: 10.3389/fcell.2020.577125] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/30/2020] [Indexed: 12/30/2022] Open
Abstract
Background: Post-surgical recurrence of the metastatic colorectal cancer (mCRC) remains a challenge, even with adjuvant therapy. Moreover, patients show variable outcomes. Here, we set to identify gene models based on the perspectives of intrinsic cell activities and extrinsic immune microenvironment to predict the recurrence of mCRC and guide the adjuvant therapy. Methods: An RNA-based gene expression analysis of CRC samples (total = 998, including mCRCs = 344, non-mCRCs = 654) was performed. A metastasis-evaluation model (MEM) for mCRCs was developed using the Cox survival model based on the prognostic differentially expressed genes between mCRCs and non-mCRCs. This model separated the mCRC samples into high- and low-recurrence risk clusters that were tested using machine learning to predict recurrence. Further, an immune prognostic model (IPM) was built using the COX survival model with the prognostic differentially expressed immune-related genes between the two MEM risk clusters. The ability of MEM and IPM to predict prognosis was analyzed and validated. Moreover, the IPM was utilized to evaluate its relationship with the immune microenvironment and response to immuno-/chemotherapy. Finally, the dysregulation cause of IPM three genes was analyzed in bioinformatics. Results: A high post-operative recurrence risk was observed owing to the downregulation of the immune response, which was influenced by MEM genes (BAMBI, F13A1, LCN2) and their related IPM genes (SLIT2, CDKN2A, CLU). The MEM and IPM were developed and validated through mCRC samples to differentiate between low- and high-recurrence risk in a real-world cohort. The functional enrichment analysis suggested pathways related to immune response and immune system diseases as the major functional pathways related to the IPM genes. The IPM high-risk group (IPM-high) showed higher fractions of regulatory T cells (Tregs) and smaller fractions of resting memory CD4+ T cells than the IPM-low group. Moreover, the stroma and immune cells in the IPM-high samples were scant. Further, the IPM-high group showed downregulation of MHC class II molecules. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and GDSC analysis suggested the IPM-low as a promising responder to anti-CTLA-4 therapy and the common FDA-targeted drugs, while the IPM-high was non-responsive to these treatments. However, treatment using anti-CDKN2A agents, along with the activation of major histocompatibility complex (MHC) class-II response might sensitize this refractory mCRC subgroup. The dysfunction of MEIS1 might be the reason for the dysregulation of IPM genes. Conclusions: The IPM could identify subgroups of mCRC with a distinct risk of recurrence and stratify the patients sensitive to immuno-/chemotherapy. Further, for the first time, our study highlights the importance of MHC class-II molecules in the treatment of mCRCs using immunotherapy.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yefan Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyu Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianguo Zhou
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianmei Liu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Lopes C, Pereira C, Medeiros R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers (Basel) 2021; 13:E203. [PMID: 33429909 PMCID: PMC7828050 DOI: 10.3390/cancers13020203] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
The role of the amino acid transporters ASCT2 and LAT1 in cancer has been explored throughout the years. In this review, we report their impact on the hallmarks of cancer, as well as their clinical significance. Overall, both proteins have been associated with cell death resistance through dysregulation of caspases and sustainment of proliferative signaling through mTOR activation. Furthermore, ASCT2 appears to play an important role in cellular energetics regulation, whereas LAT1 expression is associated with angiogenesis and invasion and metastasis activation. The molecular impact of these proteins on the hallmarks of cancer translates into various clinical applications and both transporters have been identified as prognostic factors in many types of cancer. Concerning their role as therapeutic targets, efforts have been undertaken to synthesize competitive or irreversible ASCT2 and LAT1 inhibitors. However, JHP203, a selective inhibitor of the latter, is, to the best of our knowledge, the only compound included in a Phase 1 clinical trial. In conclusion, considering the usefulness of ASCT2 and LAT1 in a variety of cancer-related pathways and cancer therapy/diagnosis, the development and testing of novel inhibitors for these transporters that could be evaluated in clinical trials represents a promising approach to cancer prognosis improvement.
Collapse
Affiliation(s)
- Catarina Lopes
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
| | - Carina Pereira
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
- CINTESIS—Center for Health Technology and Services Research, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
- Research Department of the Portuguese League Against Cancer—North (LPCC-NRNorte), Estrada da Circunvalação, 4200-177 Porto, Portugal
| |
Collapse
|
32
|
Abdellatif AAH, Rasheed Z, Alhowail AH, Alqasoumi A, Alsharidah M, Khan RA, Aljohani ASM, Aldubayan MA, Faisal W. Silver Citrate Nanoparticles Inhibit PMA-Induced TNFα Expression via Deactivation of NF-κB Activity in Human Cancer Cell-Lines, MCF-7. Int J Nanomedicine 2020; 15:8479-8493. [PMID: 33154638 PMCID: PMC7608585 DOI: 10.2147/ijn.s274098] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background The nuclear factor kappa-B (NF-κB) is a major transcription factor responsible for the production of numerous inflammatory mediators, including the tumor necrosis factor (TNFα), which has a lethal association with cancer’s onset. The silver nanoparticles (AgNPs) are widely used in cancer treatment and several other biomedical applications. Objective The study aimed to determine the effects of silver citrate nanoparticles (AgNPs-CIT) on NF-κB activation together with TNFα mRNA/protein expressions in the phorbol myristate acetate (PMA)-stimulated MCF-7 human breast cancer cell-lines. Methods The AgNPs-CIT were synthesized by the reduction method, and the prepared AgNPs-CIT were characterized for their shape, absorption in UV-VIS electromagnetic radiations, size distribution, ζ-potential, and antioxidant activity. The MCF-7 cell-lines were pretreated with AgNPs-CIT and stimulated with PMA. The TNFα mRNA expressions were determined by real-time PCR, whereas the protein production was determined by the ELISA. The NF-κB activity was distinctly observed by highly-specific DNA-based ELISA, and by NF-κB-specific inhibitor, Bay 11–7082. Results The prepared AgNPs-CIT were spherical and have an absorption wavelength range of 381–452 nm wherein the particles size ranged between 19.2±0.1 to 220.77±0.12 nm with the charge range −9.99±0.8 to −34.63±0.1 mV. The prepared AgNPs-CIT showed comparative antioxidant activity at >40% inhibitions level of the DPPH radicals. The AgNPs-CIT were found to be non-toxic to MCF-7 cell-lines and inhibited PMA-induced activation of the NF-κBp65, and also the mRNA/protein expression of TNFα. Conclusion This is the first report that showed AgNPs-CIT inhibited TNFα expression via deactivation of the NF-κB signaling event in stimulated breast cancer cells. The results have important implications for the development of novel therapeutic strategies for the prevention/treatment of cancers and/or inflammatory disorders.
Collapse
Affiliation(s)
- Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Zafar Rasheed
- Department of Medical Biochemistry, College of Medicine, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Ahmad H Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Abdulmajeed Alqasoumi
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Mansour Alsharidah
- Department of Physiology, College of Medicine, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Riaz A Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Maha A Aldubayan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Waleed Faisal
- School of Pharmacy, University College Cork, Cork, Ireland.,Faculty of Pharmacy, Minya University, Minya, Egypt
| |
Collapse
|
33
|
Dahmani Z, Addou-Klouche L, Gizard F, Dahou S, Messaoud A, Chahinez Djebri N, Benaissti MI, Mostefaoui M, Terbeche H, Nouari W, Miliani M, Lefranc G, Fernandez A, Lamb NJ, Aribi M. Metformin partially reverses the inhibitory effect of co-culture with ER-/PR-/HER2+ breast cancer cells on biomarkers of monocyte antitumor activity. PLoS One 2020; 15:e0240982. [PMID: 33108409 PMCID: PMC7591052 DOI: 10.1371/journal.pone.0240982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immune activities of monocytes (MOs) can be altered within the microenvironment of solid malignancies, including breast cancer. Metformin (1,1-dimethylbiguanide hydrochloride, MET), has been shown to decrease tumor cell proliferation, but its effects have yet to be explored with respect to MOs (monocytes) activity during their crosstalk with breast cancer cells. Here, we investigated the effects of MET on overall phenotypic functional activities, including cellular immunometabolism and protective redox signaling based-biomarkers, intracellular free calcium ions (ifCa2+), phagocytosis and co-operative cytokines (IFN-γ and IL-10) of autologous MOs before and during their interplay with primary ER-/PR-/HER2+ breast cancer cells. METHODS Human primary breast cancer cells were either cultured alone or co-cultured with autologous MOs before treatment with MET. RESULTS MET downregulated breast cancer cell proliferation and phagocytosis, while having no significant effect on the ratio of phosphorylated Akt (p-Akt) to total Akt. Additionally, we observed that, in the absence of MET treatment, the levels of lactate dehydrogenase (LDH)-based cytotoxicity, catalase, ifCa2+, IL-10 and arginase activity were significantly reduced in co-cultures compared to levels in MOs cultured alone whereas levels of inducible nitric oxide synthase (iNOS) activity were significantly increased. In contrast, MET treatment reduced the effects measured in co-culture on the levels of LDH-based cytotoxicity, arginase activity, catalase, ifCa2+, and IFN-γ. MET also induced upregulation of both iNOS and arginase in MO cells, although the increase did not reach significant difference for iNOS activity. Moreover, MET induced a robust increase of superoxide dismutase (SOD) activity in MOs, but not in MOs co-cultured with breast cancer cells. Furthermore, MET markedly upregulated the levels of IFN-γ production and downregulated those of IL-10 in isolated MOs, while inducing a slight opposing up-regulation of IL-10 production in co-cultures. CONCLUSIONS Our results show that the biomarkers of phenotypic functional activities of MOs are modified after co-culturing with primary human breast cancer cells. Treatment of co-cultures with MET resulted in increased release of antitumor cytokine IFN-γ and ifCa2+, and increased cell necrosis during breast cancer cells-MOs crosstalk.
Collapse
Affiliation(s)
- Zoheir Dahmani
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Lynda Addou-Klouche
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Florence Gizard
- Cell Biology Unit, IGH CNRS, Université de Montpellier, (UMR 9002), Montpellier, France
| | - Sara Dahou
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Aida Messaoud
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Nihel Chahinez Djebri
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Mahmoud Idris Benaissti
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Meriem Mostefaoui
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Hadjer Terbeche
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Marwa Miliani
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Gérard Lefranc
- IGH, UMR 9002 CNRS-Université de Montpellier, Montpellier, France
| | - Anne Fernandez
- Cell Biology Unit, IGH CNRS, Université de Montpellier, (UMR 9002), Montpellier, France
| | - Ned J. Lamb
- Cell Biology Unit, IGH CNRS, Université de Montpellier, (UMR 9002), Montpellier, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
34
|
Target the human Alanine/Serine/Cysteine Transporter 2(ASCT2): Achievement and Future for Novel Cancer Therapy. Pharmacol Res 2020; 158:104844. [DOI: 10.1016/j.phrs.2020.104844] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
|
35
|
Pupae protein extracts exert anticancer effects by downregulating the expression of IL-6, IL-1β and TNF-α through biomolecular changes in human breast cancer cells. Biomed Pharmacother 2020; 128:110278. [PMID: 32480223 DOI: 10.1016/j.biopha.2020.110278] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 12/14/2022] Open
Abstract
The Pupae of Bombyx mori and Samia ricini are a source of high-quality proteins and essential nutrient elements for human. Recent studies revealed that protein extracted from pupae possessed therapeutic benefits for the treatment of many diseases. However, the anticancer activity of protein extracts from the pupae of B. mori and S. ricini has been rarely reported. Our objective was to study the effect of protein extracts from the pupae of B. mori and S. ricini on cytotoxicity and expression of pro-inflammatory cytokines; IL-6, IL-1β and TNF-α, in breast cancer cells (MCF-7). Additionally, anticancer action of protein extracted from the pupae was further investigated through biomolecular changes in MCF-7 cells using Fourier transform infrared (FTIR) spectroscopy. Pupae protein extracts of B. mori exhibited cytotoxic effects with an IC50 value of 15.23 + 0.4 μg/mL with higher selectivity than doxorubicin on MCF-7 cells. Fourier transform infrared (FTIR) spectroscopy revealed that lipid contents in MCF-7 cells treated with pupae protein extracts of B. mori were higher than untreated cells. Treatment with protein extracts from pupae of B. mori or S. ricini caused significantly reduced protein and nucleic acid contents of MCF-7 cells. The expression of IL-6, IL-1β and TNF-α in MCF-7 treated cells was investigated using RT-qPCR and ELISA. Our results revealed that protein extracts from the pupae of B. mori or S. ricini significantly decreased IL-6, IL-1β and TNF-α in MCF-7 cells both at mRNA and protein levels. Expression of IL-6 and IL-1β in MCF-7 treated cells, especially IL-6, was strongly reduced compared to untreated cells, while TNF-α expression was slightly decreased. These findings suggest that pupae protein extracted from B. mori or S. ricini may play a role in breast cancer through a down-regulatory action on the expression of IL-6, IL-1β and TNF-α, and may also exert anticancer effects by causing biochemical changes of lipids, proteins and nucleic acids. These findings indicate that pupae protein extracted from B. mori or S. ricini may provide a potential novel therapeutic target for breast cancer.
Collapse
|
36
|
Yang H, Jin W, Liu H, Gan D, Cui C, Han C, Wang Z. Immune-Related Prognostic Model in Colon Cancer: A Gene Expression-Based Study. Front Genet 2020; 11:401. [PMID: 32457797 PMCID: PMC7227137 DOI: 10.3389/fgene.2020.00401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/30/2020] [Indexed: 01/17/2023] Open
Abstract
Mounting evidence supports that the malignant phenotypes of cancers are defined not only by the intrinsic activity of tumor cells but also by immune cells that are recruited and activated in tumor-related microenvironment. Here, we developed a diagnostic and prognostic model for colon cancer, based on expression profiles of immune-related genes and immune cell component. As a result, we found that B cell infiltration ratio, CD4+ T cells, as well as immune-related genes of TRIB3, CHGA, CASP7, LGALS4, LEP, NOX4, IL17A, and HSPD1 may be highly relevant with clinical outcome of colon cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Changpeng Han
- Department of Coloproctology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenyi Wang
- Department of Coloproctology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
37
|
Duan M, Hu F, Li D, Wu S, Peng N. Silencing KPNA2 inhibits IL-6-induced breast cancer exacerbation by blocking NF-κB signaling and c-Myc nuclear translocation in vitro. Life Sci 2020; 253:117736. [PMID: 32360571 DOI: 10.1016/j.lfs.2020.117736] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 01/16/2023]
Abstract
AIMS Recently, studies indicated that inflammation could exacerbate the development of BC. Karyopherin α-2 (KPNA2) is a molecule which modulates nucleocytoplasmic transport and is involved in malignant cellular behavior and carcinogenesis. Our study aims to elucidate the role of KPNA2 in BC pathogenesis and explore the mechanism of KPNA2 in regulating inflammation-induced BC exacerbations. MAIN METHODS We measured the expression of KPNA2 in BC cells. Through loss-of-function experiments, the functional role of KPNA2 in MCF-7 and MDA-MB-468 cells was evaluated. SK-BR-3 cells were treated with IL-6 as an inflammatory in vitro model of BC. ELISA determination exhibited the contents of cytokines. RANKL and leptomycin B treatments activated NF-κB signaling and inhibited the nuclear translocation of c-Myc, respectively. KEY FINDINGS The results showed that KPNA2 was significantly up-regulated in BC and silencing KPNA2 inhibited the proliferation, migration and invasion of BC cells, while the cycle arrest was induced, via blocking NF-κB signaling and c-Myc nuclear translocation. IL-6 stimulated the secretions of IL-8 and IL-17 in BC cells, and elevated KPNA2 expression. However, KPNA2 knockdown suppressed the inflammatory responses and malignant progression of BC induced by IL-6. SIGNIFICANCE In conclusion, our study illustrated that KPNA2 regulated BC development, as well as IL-6-induced inflammation and exacerbation, via NF-κB signaling and c-Myc nuclear translocation. This may provide a novel target for BC therapy.
Collapse
Affiliation(s)
- Mingyue Duan
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, China; Shaanxi Institute of Pediatric Diseases, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an 710003, China
| | - Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, China
| | - Dan Li
- Department of Immunology and Rheumatology, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an 710003, China
| | - Shouzhen Wu
- Department of Clinical Laboratory, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an 710003, China
| | - Niancai Peng
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, China; The Biomedical-information Engineering Laboratory of State Ministry of Education Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
38
|
Urbano AC, Nascimento C, Soares M, Correia J, Ferreira F. Clinical Relevance of the serum CTLA-4 in Cats with Mammary Carcinoma. Sci Rep 2020; 10:3822. [PMID: 32123292 PMCID: PMC7052166 DOI: 10.1038/s41598-020-60860-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/22/2020] [Indexed: 12/21/2022] Open
Abstract
Cytotoxic T lymphocyte associated antigen 4 (CTLA-4) serves an important role in breast cancer progression, which has led to the development of novel immunotherapies aimed at blocking tumor immune evasion. Although feline mammary carcinoma is increasingly recognized as a valuable cancer model, no studies on CTLA-4 function had been conducted in this species. The serum CTLA-4, TNF-α and IL-6 levels of 57 female cats with mammary carcinoma were determined by ELISA, and immunohistochemistry was performed to evaluate CTLA-4 and FoxP3 expression in tumor cells and interstitial lymphocytes. The results obtained show that serum CTLA-4 levels are increased in cats with mammary carcinoma (P = 0.022), showing an association with a number of clinicopathological features: smaller tumor size, P < 0.001; absence of tumor necrosis, P < 0.001; non-basal status, P < 0.02 and HER-2-positive status. Additionally, a strong positive correlation was found between serum CTLA-4 levels and serum TNF-α (R = 0.88, P < 0.001) and IL-6 levels (R = 0.72, P < 0.001). Concerning the CTLA-4 and FoxP3 expression, although detected in both interstitial lymphocytes and tumor cells, a positive association was found only between interstitial CTLA-4 and FoxP3 expressions (R = 0.387, P = 0.01), which is negatively associated with the serum CTLA-4 levels (P = 0.03). These findings provide a preliminary step in the characterization of immune profiles in feline mammary carcinoma, uncovering a molecular rationale for targeted therapy with CTLA-4 pathway inhibitors. Finally, by strengthening the hypothesis of an immunomodulatory role for this regulator, we further validate the utility of spontaneous feline mammary carcinoma as a model for human breast cancer.
Collapse
Affiliation(s)
- Ana Catarina Urbano
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, Lisboa, 1300-477, Portugal
| | - Catarina Nascimento
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, Lisboa, 1300-477, Portugal
| | - Maria Soares
- Research Center for Biosciences and Health Technologies (CBiOS), Faculdade de Medicina Veterinária, Universidade Lusófona de Humanidades e Tecnologias (ULHT), Lisboa, 1749-024, Portugal
| | - Jorge Correia
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, Lisboa, 1300-477, Portugal
| | - Fernando Ferreira
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, Lisboa, 1300-477, Portugal.
| |
Collapse
|
39
|
Zhou J, Lei J, Wang J, Lian CL, Hua L, He ZY, Wu SG. Bioinformatics-Based Discovery of CKLF-Like MARVEL Transmembrane Member 5 as a Novel Biomarker for Breast Cancer. Front Cell Dev Biol 2020; 7:361. [PMID: 31998718 PMCID: PMC6962189 DOI: 10.3389/fcell.2019.00361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022] Open
Abstract
Chemokine-like factor (CKLF)-like MARVEL transmembrane members (CMTMs) represent a novel protein family linking the chemokine and transmembrane-4 superfamily families, which potentially play several roles in diverse physiological and pathological processes. The detailed functions and underlying molecular mechanisms of CMTMs remain elusive in breast cancer. Herein, we performed a comprehensive bioinformatic analysis to investigate the prognostic effect, potential functions, and biomolecular regulatory network of CMTMs in breast cancer. The mRNA expression level of CMTM5, in particular, was significantly downregulated in breast cancer; moreover, high mRNA expression level of CMTM5 was significantly associated with better relapse-free survival. DNA promoter hypermethylation of CMTM5 was negatively correlated with its mRNA expression levels. Furthermore, CMTM5 strongly associated with pathway in MARVEL domains, chemotaxis, cytokines, transmembrane structures, and integral component of membrane. For example, genes related to MARVEL domains, transmembrane structures, and chemokines were significantly enriched. Our findings indicate that CMTM5 can be used as a prognostic biomarker and potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Jian Lei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Jun Wang
- Department of Radiation Oncology, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Chen-Lu Lian
- Department of Radiation Oncology, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Li Hua
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Zhen-Yu He
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - San-Gang Wu
- Department of Radiation Oncology, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| |
Collapse
|
40
|
Cruceriu D, Baldasici O, Balacescu O, Berindan-Neagoe I. The dual role of tumor necrosis factor-alpha (TNF-α) in breast cancer: molecular insights and therapeutic approaches. Cell Oncol (Dordr) 2020; 43:1-18. [PMID: 31900901 DOI: 10.1007/s13402-019-00489-1] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer is the most prevalent cancer among women worldwide and the fifth cause of death among all cancer patients. Breast cancer development is driven by genetic and epigenetic alterations, with the tumor microenvironment (TME) playing an essential role in disease progression and evolution through mechanisms like inflammation promotion. TNF-α is one of the essential pro-inflammatory cytokines found in the TME of breast cancer patients, being secreted both by stromal cells, mainly by tumor-associated macrophages, and by the cancer cells themselves. In this review, we explore the biological and clinical impact of TNF-α in all stages of breast cancer development. First of all, we explore the correlation between TNF-α expression levels at the tumor site or in plasma/serum of breast cancer patients and their respective clinical status and outcome. Secondly, we emphasize the role of TNF-α signaling in both estrogen-positive and -negative breast cancer cells. Thirdly, we underline TNF-α involvement in epithelial-to-mesenchymal transition (EMT) and metastasis of breast cancer cells, and we point out the contribution of TNF-α to the development of acquired drug resistance. CONCLUSIONS Collectively, these data reveal a pro-tumorigenic role of TNF-α during breast cancer progression and metastasis. We systemize the knowledge regarding TNF-α-related therapies in breast cancer, and we explain how TNF-α may act as both a target and a drug in different breast cancer therapeutic approaches. By corroborating the known molecular effects of TNF-α signaling in breast cancer cells with the results from several preclinical and clinical trials, including TNF-α-related clinical observations, we conclude that the potential of TNF-α in breast cancer therapy promises to be of great interest.
Collapse
Affiliation(s)
- Daniel Cruceriu
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania.,Department of Molecular Biology and Biotechnology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Oana Baldasici
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Ovidiu Balacescu
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania. .,11th Department of Medical Oncology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 34-36 Republicii Street, 400015, Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania. .,Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania. .,MedFuture Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
| |
Collapse
|
41
|
Murugaiah V, Agostinis C, Varghese PM, Belmonte B, Vieni S, Alaql FA, Alrokayan SH, Khan HA, Kaur A, Roberts T, Madan T, Bulla R, Kishore U. Hyaluronic Acid Present in the Tumor Microenvironment Can Negate the Pro-apototic Effect of a Recombinant Fragment of Human Surfactant Protein D on Breast Cancer Cells. Front Immunol 2020; 11:1171. [PMID: 32733438 PMCID: PMC7360846 DOI: 10.3389/fimmu.2020.01171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/12/2020] [Indexed: 02/05/2023] Open
Abstract
Human surfactant protein D (SP-D) belongs to the family of collectins that is composed of a characteristic amino-terminal collagenous region and a carboxy-terminal C-type lectin domain. Being present at the mucosal surfaces, SP-D acts as a potent innate immune molecule and offers protection against non-self and altered self, such as pathogens, allergens, and tumor. Here, we examined the effect of a recombinant fragment of human SP-D (rfhSP-D) on a range of breast cancer lines. Breast cancer has four molecular subtypes characterized by varied expressions of estrogen (ER), progesterone (PR), and epidermal growth factor (EGF) receptors (HER2). The cell viability of HER2-overexpressing (SKBR3) and triple-positive (BT474) breast cancer cell lines [but not of a triple-negative cell line (BT20)] was reduced following rfhSP-D treatment at 24 h. Upregulation of p21/p27 cell cycle inhibitors and p53 phosphorylation (Ser15) in rfhSP-D-treated BT474 and SKBR3 cell lines signified G2/M cell cycle arrest. Cleaved caspases 9 and 3 were detected in rfhSP-D-treated BT474 and SKBR3 cells, suggesting an involvement of the intrinsic apoptosis pathway. However, rfhSP-D-induced apoptosis was nullified in the presence of hyaluronic acid (HA) whose increased level in breast tumor microenvironment is associated with malignant tumor progression and invasion. rfhSP-D bound to solid-phase HA and promoted tumor cell proliferation. rfhSP-D-treated SKBR3 cells in the presence of HA showed decreased transcriptional levels of p53 when compared to cells treated with rfhSP-D only. Thus, HA appears to negate the anti-tumorigenic properties of rfhSP-D against HER2-overexpressing and triple-positive breast cancer cells.
Collapse
Affiliation(s)
- Valarmathy Murugaiah
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Chiara Agostinis
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Praveen M. Varghese
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Beatrice Belmonte
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Salvatore Vieni
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Fanan A. Alaql
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Salman H. Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Haseeb A. Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anuvinder Kaur
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Terry Roberts
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, ICMR—National Institute for Research in Reproductive Health, Mumbai, India
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
- *Correspondence: Roberta Bulla
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- Uday Kishore ;
| |
Collapse
|
42
|
Markkanen E. Know Thy Model: Charting Molecular Homology in Stromal Reprogramming Between Canine and Human Mammary Tumors. Front Cell Dev Biol 2019; 7:348. [PMID: 31921858 PMCID: PMC6927989 DOI: 10.3389/fcell.2019.00348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/03/2019] [Indexed: 12/24/2022] Open
Abstract
Spontaneous canine simple mammary tumors (CMTs) are often viewed as models of human breast cancer. Cancer-associated stroma (CAS) is central for initiation and progression of human cancer, and is likely to play a key role in canine tumors as well. Until recently, however, canine CAS in general, and in CMT in particular, lacked detailed characterization and it remained unclear how canine and human CAS compare. This void in knowledge regarding canine CAS and the resulting lack of unbiased cross-species analysis of molecular homologies and differences undermined the validity of the canine model for human disease. To assess stromal reprogramming in canine breast tumors, we have recently established a protocol to specifically isolate and analyze CAS and matched normal stroma from archival, formalin-fixed paraffin embedded (FFPE) clinical tumor samples using laser-capture microdissection followed by next-generation RNA-sequencing. Using this approach, we have analyzed stromal reprogramming in both malignant canine mammary carcinomas (mCAs) as well as benign canine mammary adenomas in a series of studies. Our results demonstrate strong stromal reprogramming in CMTs and identify high-grade molecular homology between human and canine CAS. Here, I aim to give a short background on the value of comparative oncology in general, and spontaneous CMT in particular. This will be followed by a concise review of the current knowledge of stromal reprogramming in both malignant canine mCA and benign adenoma. Finally, I will conclude with insights on highly conserved aspects of stromal reprogramming between CMT and human breast cancer that accentuate the relevance of CAS in CMT as a model for the human disease.
Collapse
Affiliation(s)
- Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Yin S, Wang N, Riabov V, Mossel DM, Larionova I, Schledzewski K, Trofimova O, Sevastyanova T, Zajakina A, Schmuttermaier C, Gratchev A, Flatley A, Kremmer E, Zavyalova M, Cherdyntseva N, Simon-Keller K, Marx A, Klüter H, Goerdt S, Kzhyshkowska J. SI-CLP inhibits the growth of mouse mammary adenocarcinoma by preventing recruitment of tumor-associated macrophages. Int J Cancer 2019; 146:1396-1408. [PMID: 31525266 DOI: 10.1002/ijc.32685] [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: 09/13/2018] [Revised: 06/30/2019] [Accepted: 08/07/2019] [Indexed: 02/05/2023]
Abstract
Chitinase-like proteins (CLP) are chitin-binding proteins that lack chitin hydrolyzing activity, but possess cytokine-like and growth factor-like properties, and play crucial role in intercellular crosstalk. Both human and mice express two members of CLP family: YKL-40 and stabilin-1 interacting chitinase-like protein (SI-CLP). Despite numerous reports indicating the role of YKL-40 in the support of angiogenesis, tumor cell proliferation, invasion and metastasis, the role of its structurally related protein SI-CLP in cancer was not reported. Using gain-of-function approach, we demonstrate in the current study that the expression of recombinant SI-CLP in mouse TS/A mammary adenocarcinoma cells results in significant and persistent inhibition of in vivo tumor growth. Using quantitative immunohistochemistry, we show that on the cellular level this phenomenon is associated with reduced infiltration of tumor-associated macrophages (TAMs), CD4+ and FoxP3+ cells in SI-CLP expressing tumors. Gene expression analysis in TAM isolated from SI-CLP-expressing and control tumors demonstrated that SI-CLP does not affect macrophage phenotype. However, SI-CLP significantly inhibited migration of murine bone-marrow derived macrophages and human primary monocytes toward monocyte-recruiting chemokine CCL2 produced in the tumor microenvironment (TME). Mechanistically, SI-CLP did not affect CCL2/CCR2 interaction, but suppressed cytoskeletal rearrangements in response to CCL2. Altogether, our data indicate that SI-CLP functions as a tumor growth inhibitor in mouse breast cancer by altering cellular composition of TME and blocking cytokine-induced TAM recruitment. Taking into consideration weak to absent expression of SI-CLP in human breast cancer, it can be considered as a therapeutic protein to block TAM-mediated support of breast tumor growth.
Collapse
Affiliation(s)
- Shuiping Yin
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Nan Wang
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science andTechnology, Wuhan, China
| | - Vladimir Riabov
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| | - Dieuwertje M Mossel
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia.,Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Kai Schledzewski
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Olga Trofimova
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Tatyana Sevastyanova
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Anna Zajakina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Christina Schmuttermaier
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Alexei Gratchev
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Laboratory for Tumor Stromal Cell Biology, Institute of Carcinogenesis, NN Blokhin Cancer Research Center, Russian Academy of Sciences, Moscow, Russia
| | - Andrew Flatley
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Marina Zavyalova
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Nadezhda Cherdyntseva
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia.,Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Katja Simon-Keller
- Institute of Pathology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Alexander Marx
- Institute of Pathology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Harald Klüter
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Sergij Goerdt
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Julia Kzhyshkowska
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| |
Collapse
|
44
|
Zileuton, a 5-Lipoxygenase Inhibitor, Exerts Anti-Angiogenic Effect by Inducing Apoptosis of HUVEC via BK Channel Activation. Cells 2019; 8:cells8101182. [PMID: 31575085 PMCID: PMC6829222 DOI: 10.3390/cells8101182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023] Open
Abstract
The arachidonic acid metabolism through 5-lipoxygenase (5-LO) pathways is involved in modulating both tumorigenesis and angiogenesis. Although anti-carcinogenic activities of certain 5-LO inhibitors have been reported, the role of zileuton, a well known 5-LO inhibitor, on the endothelial cell proliferation and angiogenesis has not been fully elucidated. Here, we report that zileuton has an anti-angiogenic effect, and the underlying mechanisms involved activation of the large-conductance Ca2+-activated K+ (BK) channel. Our results show that zileuton significantly prevented vascular endothelial growth factor (VEGF)-induced proliferation of human umbilical vein endothelial cells (HUVECs) in vitro, as well as in vivo. However, such anti-angiogenic effect of zileuton was abolished by iberiotoxin (IBTX), a BK channel blocker, suggesting zileuton-induced activation of BK channel was critical for the observed anti-angiogenic effect of zileuton. Furthermore, the anti-angiogenic effect of zileuton was, at least, due to the activation of pro-apoptotic signaling cascades which was also abolished by IBTX. Additionally, zileuton suppressed the expression of VCAM-1, ICAM-1, ETS related gene (Erg) and the production of nitric oxide (NO). Taken together, our results show that zileuton prevents angiogenesis by activating the BK channel dependent-apoptotic pathway, thus highlighting its therapeutic capacity in angiogenesis-related diseases, such as cancer.
Collapse
|
45
|
Walker WH, Borniger JC. Molecular Mechanisms of Cancer-Induced Sleep Disruption. Int J Mol Sci 2019; 20:E2780. [PMID: 31174326 PMCID: PMC6600154 DOI: 10.3390/ijms20112780] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Sleep is essential for health. Indeed, poor sleep is consistently linked to the development of systemic disease, including depression, metabolic syndrome, and cognitive impairments. Further evidence has accumulated suggesting the role of sleep in cancer initiation and progression (primarily breast cancer). Indeed, patients with cancer and cancer survivors frequently experience poor sleep, manifesting as insomnia, circadian misalignment, hypersomnia, somnolence syndrome, hot flushes, and nightmares. These problems are associated with a reduction in the patients' quality of life and increased mortality. Due to the heterogeneity among cancers, treatment regimens, patient populations and lifestyle factors, the etiology of cancer-induced sleep disruption is largely unknown. Here, we discuss recent advances in understanding the pathways linking cancer and the brain and how this leads to altered sleep patterns. We describe a conceptual framework where tumors disrupt normal homeostatic processes, resulting in aberrant changes in physiology and behavior that are detrimental to health. Finally, we discuss how this knowledge can be leveraged to develop novel therapeutic approaches for cancer-associated sleep disruption, with special emphasis on host-tumor interactions.
Collapse
Affiliation(s)
- William H Walker
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA.
| | - Jeremy C Borniger
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
46
|
Wound fluids collected postoperatively from patients with breast cancer induce epithelial to mesenchymal transition but intraoperative radiotherapy impairs this effect by activating the radiation-induced bystander effect. Sci Rep 2019; 9:7891. [PMID: 31133667 PMCID: PMC6536501 DOI: 10.1038/s41598-019-44412-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
Wound fluids (WF) are believed to play a role in the local recurrences by inducing an inflammatory process in scar tissue area. Given that most local relapse in primary breast cancer patients occur within the scar tissue area, researchers have investigated whether localized radiotherapy, such as intraoperative radiotherapy (IORT), could be more effective than postoperative RT in inhibiting local tumor recurrence. The epithelial-mesenchymal transition (EMT) program plays a critical role in promoting metastasis in epithelium-derived carcinoma. Given this background the main aim of the present study was to determine the mechanisms by which IORT decreases the tumorigenic potential of WF. We assumed that postoperative fluids from patients would activate the radiation-induced bystander effect (RIBE) in treated cells, thus altering the tumor microenvironment. To confirm this hypothesis, WF collected from patients after breast conserving surgery (BCS) alone, after BCS followed by IORT treatment or WF from BCS patients together with RIBE medium were incubated with MCF7 and MDA-MB-468 cells. Changes in the CSC phenotype, in EMT program and potential to migrate were performed to determine the possible role of WF on the migration of breast cancer cells. Our findings show that wound fluids stimulate the CSC phenotype and EMT program in breast cancer cell lines. This effect was partially abrogated when the cells were incubated in wound fluids collected from patients after breast-conserving surgery followed by IORT. Additionally, we confirmed the role of radiation-induced bystander effect in altering the properties of the WF to induce the CSC phenotype and EMT program.
Collapse
|
47
|
Bahiraee A, Ebrahimi R, Halabian R, Aghabozorgi AS, Amani J. The role of inflammation and its related microRNAs in breast cancer: A narrative review. J Cell Physiol 2019; 234:19480-19493. [PMID: 31025369 DOI: 10.1002/jcp.28742] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/27/2019] [Accepted: 04/10/2019] [Indexed: 12/21/2022]
Abstract
Breast cancer is recognized as the most common type of cancer among women with a high rate of mortality all over the world. Over the past years, growing attention has been regarded to realize more about the mechanisms underlying the disease process. It is revealed that the progression of breast cancer may be strongly linked to chronic inflammation owing to the role of inflammatory factors in genetic instability and subsequent cancer predisposition. Although the association between breast cancer and inflammatory pathways has been well-defined now, only recent evidence pointed towards the inflammation-related microRNAs (miRNAs) as potential biomarkers and therapeutic targets involved in the crosstalk of multiple pathways during breast cancer development. Moreover, the practical interactions between these miRNAs and inflammatory factors are also a little characterized. In this review, we intended to describe the effects of predominant inflammatory pathways such as cytokines, phosphoinositide 3-kinase/protein kinase B, and nuclear factor kappa B in association with tumor promoting and tumor suppressing miRNAs on breast cancer progression. Providing new studies in the field of combining biomarkers for early diagnosis, prognosis, and monitoring breast cancer are very important. Notably, understanding the underlying mechanisms of miRNAs as a possible link between inflammation and tumorigenesis may offer a novel insight for combating this epidemic.
Collapse
Affiliation(s)
- Alireza Bahiraee
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirsaeed Sabeti Aghabozorgi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Cytokine Modulation in Breast Cancer Patients Undergoing Radiotherapy: A Revision of the Most Recent Studies. Int J Mol Sci 2019; 20:ijms20020382. [PMID: 30658426 PMCID: PMC6359111 DOI: 10.3390/ijms20020382] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/13/2022] Open
Abstract
Breast cancer (BC) is the most common tumor and the second cause for cancer-related death in women worldwide, although combined treatments are well-established interventions. Several effects seem to be responsible for poor outcomes in advanced or triple-negative BC patients. Focusing on the interaction of ionizing radiation with tumor and normal tissues, the role of cytokine modulation as a surrogate of immunomodulation must still be explored. In this work, we carried out an overview of studies published in the last five years involving the cytokine profile in BC patients undergoing radiotherapy. The goal of this review was to evaluate the profile and modulation of major cytokines and interleukins as potential biomarkers of survival, treatment response, and toxicity in BC patient undergoing radiotherapy. Out of 47 retrieved papers selected using PubMed search, 15 fulfilled the inclusion criteria. Different studies reported that the modulation of specific cytokines was time- and treatment-dependent. Radiotherapy (RT) induces the modulation of inflammatory cytokines up to 6 months for most of the analyzed cytokines, which in some cases can persist up to several years post-treatment. The role of specific cytokines as prognostic and predictive of radiotherapy outcome is critically discussed.
Collapse
|
49
|
Shu L, Wang Z, Wang Q, Wang Y, Zhang X. Signature miRNAs in peripheral blood monocytes of patients with gastric or breast cancers. Open Biol 2018; 8:180051. [PMID: 30381359 PMCID: PMC6223219 DOI: 10.1098/rsob.180051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
The dysregulation of microRNAs (miRNAs), key posttranscriptional regulators of gene expression, is closely associated with cancer development. However, the miRNAs of monocytes, important cells of tumour immunity, have not been extensively explored. In the present study, the differentially expressed miRNAs of blood monocytes derived from gastric and breast cancer patients and healthy donors were characterized. The results indicated that 74 miRNAs were upregulated and 46 miRNAs were downregulated in monocytes of patients with breast or gastric cancers compared with the healthy donors, suggesting that these 120 miRNAs from transformed monocytes were associated with cancers. The differentially expressed miRNAs, 38 of which were novel, were further validated using quantitative real-time PCR. As an example, the results showed that miR-150-5p downregulated the CCR2 expression in monocytes by targeting Notch 3, thus leading to the suppression of tumorigenesis. The target gene analysis showed that 36 of the 120 miRNAs targeted cancer-related genes. KEGG pathway analysis indicated that the cancer-associated miRNAs were involved in pathways related to cancers, such as the HIF-1 signalling and the mTOR signalling pathways. Thus, our study provided new clues to comprehensively understand the relationship between miRNAs and cancers.
Collapse
Affiliation(s)
- Le Shu
- Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology and College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Zhe Wang
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, People's Republic of China
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, People's Republic of China
| | - Yumeng Wang
- Department of Gastroenterology, Chaohu Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Xiaobo Zhang
- Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology and College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| |
Collapse
|
50
|
Chemotherapy-Exacerbated Breast Cancer Metastasis: A Paradox Explainable by Dysregulated Adaptive-Response. Int J Mol Sci 2018; 19:ijms19113333. [PMID: 30373101 PMCID: PMC6274941 DOI: 10.3390/ijms19113333] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023] Open
Abstract
An emerging picture in cancer biology is that, paradoxically, chemotherapy can actively induce changes that favor cancer progression. These pro-cancer changes can be either inside (intrinsic) or outside (extrinsic) the cancer cells. In this review, we will discuss the extrinsic pro-cancer effect of chemotherapy; that is, the effect of chemotherapy on the non-cancer host cells to promote cancer progression. We will focus on metastasis, and will first discuss recent data from mouse models of breast cancer. Despite reducing the size of primary tumors, chemotherapy changes the tumor microenvironment, resulting in an increased escape of cancer cells into the blood stream. Furthermore, chemotherapry changes the tissue microenvironment at the distant sites, making it more hospitable to cancer cells upon their arrival. We will then discuss the idea and evidence that these devastating pro-metastatic effects of chemotherapy can be explained in the context of adaptive-response. At the end, we will discuss the potential relevance of these mouse data to human breast cancer and their implication on chemotherapy in the clinic.
Collapse
|