1
|
Qiu Q, Yang X, Li X, Ren Y, Huang N. Viral IL-10 can promote the proliferation, migration and invasion of nasopharyngeal carcinoma cells and inhibit their apoptosis. Discov Oncol 2025; 16:199. [PMID: 39964638 PMCID: PMC11836248 DOI: 10.1007/s12672-025-01937-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
PURPOSE Nasopharyngeal carcinoma is a common clinical malignant tumour in the nasopharynx. The secretion of vIL-10 by EB virus can promote the development of nasopharyngeal carcinoma. The purpose of this study was to provide a theoretical basis for potential targets and mechanisms of vIL-10 in nasopharyngeal carcinoma. METHODS A total of 100 ng/mL vIL-10 was applied to the nasopharyngeal carcinoma CNE-2 cell line for 48 h, and then whole transcriptome sequencing analysis was performed to identify potential targets and signalling pathways of vIL-10 in nasopharyngeal carcinoma. The effects of vIL-10 on the proliferation, migration, invasion and apoptosis of human nasopharyngeal carcinoma cells were determined by ELISA, Ki67 immunofluorescence, colony formation, transwell migration/invasion, Hoechst 33,258 staining, flow cytometry and other experiments, and the potential effects of vIL-10 on nasopharyngeal carcinoma cells were verified at the cellular level. Western blotting was performed to measure the changes in key factors of the JAK1-STAT3 signalling pathway in nasopharyngeal carcinoma cells after vIL-10 treatment. RESULTS The whole transcriptome gene sequencing results showed that vIL-10 could effectively activate the JAK-STAT signalling pathway and upregulate the expression of JAK1 and STAT3. Moreover, vIL-10 inhibited the apoptosis of nasopharyngeal carcinoma cells, enhanced their migration and invasion capabilities, and further promoted the proliferation of nasopharyngeal carcinoma cells. CONCLUSION vIL-10 regulates the JAK1-STAT3 signalling pathway, promotes the proliferation of NPC cells, enhances their migration and invasion capabilities, inhibits tumour cell apoptosis, and participates in the progression of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Qin Qiu
- Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Xingyu Yang
- Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Xiaojiang Li
- Department of Head and Neck Surgery, Third Affiliated Hospital of Kunming Medical University, No.519 Kunzhou Street, Xishan District, Kunming, 650118, People's Republic of China.
| | - Yanxin Ren
- Department of Head and Neck Surgery, Third Affiliated Hospital of Kunming Medical University, No.519 Kunzhou Street, Xishan District, Kunming, 650118, People's Republic of China.
| | - Ning Huang
- Department of Pharmacology, Kunming Medical University, Kunming, Yunnan, People's Republic of China.
| |
Collapse
|
2
|
Zhang H, Cao X, Gui R, Li Y, Zhao X, Mei J, Zhou B, Wang M. Mesenchymal Stem/Stromal cells in solid tumor Microenvironment: Orchestrating NK cell remodeling and therapeutic insights. Int Immunopharmacol 2024; 142:113181. [PMID: 39305890 DOI: 10.1016/j.intimp.2024.113181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs), originating from normal tissues, possess the capacity to home to tumor sites and differentiate into tumor-associated MSCs (TA-MSCs), which are instrumental in shaping an immunosuppressive milieu within tumors. Natural killer (NK) cells, integral to the innate immune system, are endowed with the ability to eradicate target cells autonomously, serving as an immediate defense against neoplastic growths. Nonetheless, within the tumor microenvironment (TME), NK cells often exhibit a decline in both their numerical presence and functionality. TA-MSCs have been shown to exert profound inhibitory effects on the functions of tumor-infiltrating immune cells, notably NK cells. Understanding the mechanisms by which TA-MSCs contribute to NK cell dysfunction is critical for the advancement of immune surveillance and the enhancement of tumoricidal responses. This review summarizes existing literature on NK cell modulation by TA-MSCs within the TME and proposes innovative strategies to augment antitumor immunity.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Xiaoli Cao
- Department of Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226321, China
| | - Rulin Gui
- Laboratory Animal Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Yuanyuan Li
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Xinlan Zhao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Jingyu Mei
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Baocheng Zhou
- Department of Medical Laboratory, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu Province, 222000, China.
| | - Mei Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China.
| |
Collapse
|
3
|
Trugilo KP, Cebinelli GCM, Castilha EP, da Silva MR, Berti FCB, de Oliveira KB. The role of transforming growth factor β in cervical carcinogenesis. Cytokine Growth Factor Rev 2024; 80:12-23. [PMID: 39482191 DOI: 10.1016/j.cytogfr.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024]
Abstract
Human papillomavirus (HPV) is involved in virtually all cases of cervical cancer. However, HPV alone is not sufficient to cause malignant development. The effects of chronic inflammation and the interaction of immune components with the microenvironment infected with the high-risk HPV type (HR) may contribute to cancer development. Transforming growth factor β (TGFB) appears to play an important role in cervical carcinogenesis. Protein and mRNA levels of this cytokine gradually increase as normal tissue develops into malignant tissue and are closely related to the severity of HPV infection. At the onset of infection, TGFB can inhibit the proliferation of infected cells and viral amplification by inhibiting cell growth and downregulating the transcriptional activity of the long control region (LCR) of HPV, thereby reducing the expression of early genes. When infected cells progress to a malignant phenotype, the response to the cell growth inhibitory effect of TGFB1 is lost and the suppression of E6 and E7 expression decreases. Subsequently, TGFB1 expression is upregulated by high levels of E6 and E7 oncoproteins, leading to an increase in TGFB1 in the tumor microenvironment, where this molecule promotes epithelial-to-mesenchymal transition (EMT), cell motility, angiogenesis, and immunosuppression. This interaction between HPV oncoproteins and TGFB1 is an important mechanism promoting the development and progression of cervical cancer.
Collapse
Affiliation(s)
- Kleber Paiva Trugilo
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| | | | - Eliza Pizarro Castilha
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| | - Mariane Ricciardi da Silva
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| | | | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| |
Collapse
|
4
|
Marrero-Rodriguez D, Cortes-Morales VA, Cano-Zaragoza A, Martinez-Mendoza F, Kerbel-Suton J, Vela-Patiño S, Chavez-Santoscoy A, Hinojosa-Alvarez S, Hernandez-Perez J, Gomez-Apo E, Fajardo-Orduña GR, Taniguchi-Ponciano K, Montesinos JJ, Mercado M. Mesenchymal Stem Cells Induce an Immunosuppressive Microenvironment in Pituitary Tumors. J Clin Endocrinol Metab 2024; 109:2943-2955. [PMID: 38589986 DOI: 10.1210/clinem/dgae212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 04/01/2024] [Indexed: 04/10/2024]
Abstract
CONTEXT The tumor microenvironment (TME) includes diverse cellular components such as mesenchymal stem cells (MSCs) and immune cells, among others. MSC have been isolated from different tumors and they favor tumor cell growth; however, their role in pituitary tumors (PTs) remains unknown. OBJECTIVE Herein we report the presence of MSCs in 2 adrenocorticotropin (ACTH)-secreting PTs causing Cushing disease (MCU), 2 nonfunctioning adenomas of gonadotrope differentiation (MNF), and 2 nontumoral pituitary glands (MS). METHODS We have analyzed the transcriptomic profiles by RNA sequencing and compared MSCs in terms of their immunosuppressive effects against lymphoid T-cell and macrophage populations by means of cocultures and flow cytometry. RESULTS Our transcriptomic analysis revealed molecular differences between MSCs derived from nontumoral pituitaries and MSCs derived from PTs. Two distinct subpopulations of MSC emerged: one displaying immunosuppressive properties and the other with increased proproliferative capabilities, regardless of their origin. MSCs derived from ACTH- and nonfunctioning PTs, but not those derived from nontumoral glands, significantly inhibited the proliferation of activated T cells, favored the generation of regulatory T cells, and promoted M2 macrophage polarization. Such immunosuppressive effects were correlated with an upregulation of programmed death ligand 1 and intracellular expression of macrophage colony-stimulating factor (M-CSF) and interleukin-10. Importantly, MSC derived from ACTH-PTs showed a higher immunosuppressive potential than MSC isolated from nonfunctioning tumors. CONCLUSION This study demonstrates the presence of at least 2 MSC subpopulations in the pituitary gland and suggests that immunosuppressive effects of MSCs may have important implications in PT growth.
Collapse
Affiliation(s)
- Daniel Marrero-Rodriguez
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Victor A Cortes-Morales
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
- Laboratorio de Celulas Mesenquimales Troncales, Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Amayrani Cano-Zaragoza
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Florencia Martinez-Mendoza
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Jacobo Kerbel-Suton
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Sandra Vela-Patiño
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Alejandra Chavez-Santoscoy
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Ave. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Silvia Hinojosa-Alvarez
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Ave. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Jesus Hernandez-Perez
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Ave. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Erick Gomez-Apo
- Área de Neuropatología, Servicio de Anatomía Patológica, Hospital General de México Dr. Eduardo Liceaga, Mexico City, 06720, Mexico
| | - Guadalupe R Fajardo-Orduña
- Laboratorio de Celulas Mesenquimales Troncales, Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Keiko Taniguchi-Ponciano
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Juan Jose Montesinos
- Laboratorio de Celulas Mesenquimales Troncales, Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| | - Moises Mercado
- Unidad de Investigación Médica en Enfermedades Endócrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, 06720, Mexico
| |
Collapse
|
5
|
Marín-Aquino LA, Mora-García MDL, Moreno-Lafont MC, García-Rocha R, Montesinos-Montesinos JJ, López-Santiago R, Sánchez-Torres LE, Torres-Pineda DB, Weiss-Steider B, Hernández-Montes J, Don-López CA, Monroy-García A. Adenosine increases PD-L1 expression in mesenchymal stromal cells derived from cervical cancer through its interaction with A 2AR/A 2BR and the production of TGF-β1. Cell Biochem Funct 2024; 42:e4010. [PMID: 38613217 DOI: 10.1002/cbf.4010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024]
Abstract
Mesenchymal stromal cells (MSCs) together with malignant cells present in the tumor microenvironment (TME), participate in the suppression of the antitumor immune response through the production of immunosuppressive factors, such as transforming growth factor beta 1 (TGF-β1). In previous studies, we reported that adenosine (Ado), generated by the adenosinergic activity of cervical cancer (CeCa) cells, induces the production of TGF-β1 by interacting with A2AR/A2BR. In the present study, we provide evidence that Ado induces the production of TGF-β1 in MSCs derived from CeCa tumors (CeCa-MSCs) by interacting with both receptors and that TGF-β1 acts in an autocrine manner to induce the expression of programmed death ligand 1 (PD-L1) in CeCa-MSCs, resulting in an increase in their immunosuppressive capacity on activated CD8+ T lymphocytes. The addition of the antagonists ZM241385 and MRS1754, specific for A2AR and A2BR, respectively, or SB-505124, a selective TGF-β1 receptor inhibitor, in CeCa-MSC cultures significantly inhibited the expression of PD-L1. Compared with CeCa-MSCs, MSCs derived from normal cervical tissue (NCx-MSCs), used as a control and induced with Ado to express PD-L1, showed a lower response to TGF-β1 to increase PD-L1 expression. Those results strongly suggest the presence of a feedback mechanism among the adenosinergic pathway, the production of TGF-β1, and the induction of PD-L1 in CeCa-MSCs to suppress the antitumor response of CD8+ T lymphocytes. The findings of this study suggest that this pathway may have clinical importance as a therapeutic target.
Collapse
Affiliation(s)
- Luis Antonio Marín-Aquino
- Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
- Consejo Nacional de Humanidades Ciencias y Tecnologías, CONAHCyT, Ciudad de México, México
| | - María de Lourdes Mora-García
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Martha C Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Rosario García-Rocha
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Juan José Montesinos-Montesinos
- Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Ruben López-Santiago
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Luvia Enid Sánchez-Torres
- Laboratorio de Inmunología de los microorganismos, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Daniela Berenice Torres-Pineda
- Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Benny Weiss-Steider
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Jorge Hernández-Montes
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Christian Azucena Don-López
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Alberto Monroy-García
- Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer -UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| |
Collapse
|
6
|
Youlin K, Simin L, Jian K, Li Z. Inhibition of miR-20a by pterostilbene facilitates prostate cancer cells killed by NK cells via up-regulation of NKG2D ligands and TGF-β1down-regulation. Heliyon 2023; 9:e14957. [PMID: 37064475 PMCID: PMC10102449 DOI: 10.1016/j.heliyon.2023.e14957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Natural killer (NK) cells play a potent role in antitumor immunity via spontaneously eliminating tumor directly. However, some tumors such as prostate cancer constantly escape this immune response by down-regulating cell surface molecule recognition and/or secreting immune impressive cytokines. Here, we found pterostilbene, a natural agent with potent anticancer activity, could enhance expression of major histocompatibility complex class I chain-related proteins A and B (MICA/B) on prostate cancer cells surface, which are ligands of the natural killer group 2 member D (NKG2D) expressed by NK cells, and inhibit TGF-β1 secretion by prostate cancer cells. Further, we discovered that these effects were caused by inhibition of miR-20a in prostate cancer cells by pterostilbene. MiR-20a could target the 3' untranslated region (UTR) of MICA/B, resulting in their expression down-regulation. Inhibition of TGF-β1 function by its specific antibody attenuated its impairment to NKG2D on NK cells. Finally, we observed that pterostilbene-treated prostate cancer cells were more easily to be killed by NK cells. Taken together, our findings demonstrated inhibition of miR-20a by pterostilbene in prostate cancer cells could increase MICA/B expression and decrease TGF-β1 secretion, which enhanced NK cell-mediated cytotoxicity againt prostate cancer cells, suggesting a potential approach for increasing anti-prostate cancer immune.
Collapse
Affiliation(s)
- Kuang Youlin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liang Simin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kang Jian
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhang Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Corresponding author.
| |
Collapse
|
7
|
Song Y, Li R, Ye M, Pan C, Zheng L, Wang ZW, Zhu X. Differences in chemotaxis of human mesenchymal stem cells and cervical cancer cells. Apoptosis 2022; 27:840-851. [PMID: 35849265 DOI: 10.1007/s10495-022-01749-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 11/30/2022]
Abstract
In the last decade, there has been a rapid expansion in tumor targeted therapy using mesenchymal stem cells (MSCs) based on their unique tropism towards cancer cells. Despite similarities in morphology, immunophenotype, and differential potent in vitro, MSCs originated from different tissues do not necessarily have equivalent biological behaviors. It is important to screen the most chemotactic MSCs to cancer cells. In this study, different MSCs were isolated from various human tissues including adipose, umbilical cord, amniotic membrane, and chorion. The chemotaxis of human MSCs to cervical cancer cells was measured by CCK-8, ELISA and Transwell invasion assays. Western blotting was performed to explore the underlying mechanisms. MSCs derived from distinct sources can be differently recruited to cervical cancer cells, among which chorion-derived MSC (CD-MSC) possessed the strongest tropic capacity. CXCL12 was found to be highly secreted by cervical cancer cells, in parallel with the expression of CXCR4 in all MSCs. CD-MSC displayed the highest level of CXCR4. These results indicated that CXCL12/CXCR4 pathway contributed to the different chemotaxis to cervical cancer cells of each MSCs. This study proposed that CD-MSC with the highest CXCR4 expression is a promising therapeutic vehicle for targeted therapy in cervical cancer.
Collapse
Affiliation(s)
- Yizuo Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Ruyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Miaomiao Ye
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Chunyu Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Lihong Zheng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Zhi-Wei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
8
|
Marvin DL, Spaans VM, de Kroon CD, Slieker RC, Khelil M, Ten Dijke P, Ritsma L, Jordanova ES. Low Transforming Growth Factor-β Pathway Activity in Cervical Adenocarcinomas. Front Oncol 2022; 12:797453. [PMID: 35756604 PMCID: PMC9213724 DOI: 10.3389/fonc.2022.797453] [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: 10/18/2021] [Accepted: 05/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is the fourth most common cancer in women worldwide. Squamous cell carcinoma (SCC) and adenocarcinoma (AC) are the most common histological types, with AC patients having worse prognosis. Over the last two decades, incidence rates of AC have increased, highlighting the importance of further understanding AC tumorigenesis, and the need to investigate new treatment options. The cytokine TGF-β functions as a tumour suppressor in healthy tissue. However, in tumour cells this suppressive function can be overcome. Therefore there is an increasing interest in using TGF-β inhibitors in the treatment of cancer. Here, we hypothesize that TGF-β plays a different role in SCC and AC. Analysis of RNA-seq data from the TCGA, using a TGF-β response signature, resulted in separate clustering of the two subtypes. We further investigated the expression of TGF-β-signalling related proteins (TβR1/2, SMAD4, pSMAD2, PAI-1, αvβ6 and MMP2/9) in a cohort of 62 AC patients. Low TβR2 and SMAD4 expression was associated with worse survival in AC patients and interestingly, high PAI-1 and αvβ6 expression was also correlated with worse survival. Similar correlations of TβR2, PAI-1 and αvβ6 with clinical parameters were found in previously reported SCC analyses. However, when comparing expression levels between SCC and AC patient samples, pSMAD2, SMAD4, PAI-1 and αvβ6 showed lower expression in AC compared to SCC. Because of the low expression of core TβR1/2, (p-)SMAD2 and SMAD4 proteins and the correlation with worse prognosis, TGF-β pathway most likely leads to tumour inhibitory effects in AC and therefore the use of TGF-β inhibitors would not be recommended. However, given the correlation of PAI-1 and αvβ6 with poor prognosis, the use of TGF- β inhibitors might be of interest in SCC and in the subsets of AC patients with high expression of these TGF-β associated proteins.
Collapse
Affiliation(s)
- Dieuwke L Marvin
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Vivian M Spaans
- Department of Gynaecology and Obstetrics, Leiden University Medical Center, Leiden, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Cor D de Kroon
- Department of Gynaecology and Obstetrics, Leiden University Medical Center, Leiden, Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Epidemiology and Data Science, Amsterdam University Medical Center (UMC), location VU University Medical Center (VUmc), Amsterdam, Netherlands
| | - Maryam Khelil
- Department of Gynaecology and Obstetrics, Center Gynaecological Oncology Amsterdam, Amsterdam University Medical Center (UMC), location VU University Medical Center (VUmc), Amsterdam, Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Laila Ritsma
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Ekaterina S Jordanova
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.,Department of Gynaecology and Obstetrics, Center Gynaecological Oncology Amsterdam, Amsterdam University Medical Center (UMC), location VU University Medical Center (VUmc), Amsterdam, Netherlands.,Department of Urology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
9
|
Wang X, Zhang J, Hu B, Qian F. High Expression of CSF-1R Predicts Poor Prognosis and CSF-1R high Tumor-Associated Macrophages Inhibit Anti-Tumor Immunity in Colon Adenocarcinoma. Front Oncol 2022; 12:850767. [PMID: 35444953 PMCID: PMC9014714 DOI: 10.3389/fonc.2022.850767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/10/2022] [Indexed: 11/20/2022] Open
Abstract
Background Colony stimulating factor 1 receptor (CSF-1R) is a single channel III transmembrane receptor tyrosine kinase (RTK) and plays an important role in immune regulation and the development of various cancer types. The expression of CSF-1R in colon adenocarcinoma (COAD) and its prognostic value remain incompletely understood. Therefore, we aim to explore the prognostic value of CSF-1R in COAD and its relationship with tumor immunity. Methods CSF-1R expression in a COAD cohort containing 103 patients was examined using immunohistochemistry (IHC). The relationship between CSF-1R expression and clinicopathological parameters and prognosis was evaluated. Dual immunofluorescence staining was conducted to determine the localization of CSF-1R in COAD tissues. Univariate and multivariate Cox regression analysis were performed to evaluate independent prognostic factors. Transcriptomic profiles of CSF-1Rhigh and CSF-1Rlow tumor-associated macrophages (TAMs) were investigated. Gene enrichment analysis was used to explore the signal pathways related to CSF-1R. In addition, the relationship between CSF-1R in tumor microenvironment (TME) and tumor immunity was also studied. Results IHC analysis showed that CSF-1R was overexpressed in COAD, and higher expression was associated with shorter overall survival (OS). Immunofluorescence staining showed that CSF-1R was co-localized with macrophage marker CD68. Univariate and multivariate Cox regression analysis showed that CSF-1R was an independent prognostic factor for COAD. The results of gene enrichment analysis showed that CSF-1R was involved in tumor immune response and regulation of TME. In addition, CSF-1R was significantly correlated with TME, immune cell infiltration, TMB, MSI, Neoantigen, and immune checkpoint molecules. Conclusion CSF-1R can serve as an independent prognostic factor of COAD and promising immunotherapeutic target of COAD.
Collapse
Affiliation(s)
- Xingchao Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jianfeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Baoying Hu
- Department of Immunology, Medical College, Nantong University, Nantong, China
| | - Fei Qian
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
10
|
Hilgers K, Ibrahim SA, Kiesel L, Greve B, Espinoza-Sánchez NA, Götte M. Differential Impact of Membrane-Bound and Soluble Forms of the Prognostic Marker Syndecan-1 on the Invasiveness, Migration, Apoptosis, and Proliferation of Cervical Cancer Cells. Front Oncol 2022; 12:803899. [PMID: 35155241 PMCID: PMC8828476 DOI: 10.3389/fonc.2022.803899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/03/2022] [Indexed: 12/16/2022] Open
Abstract
Cervical cancer ranks fourth among the most commonly diagnosed malignant tumors in women worldwide. Previously published evidence suggested a possible connection between the expression of the membrane-bound heparan sulfate proteoglycan syndecan-1 (Sdc-1) and the development of cervical carcinoma. Sdc-1 serves as a matrix receptor and coreceptor for receptor tyrosine kinases and additional signaling pathways. It influences cell proliferation, adhesion, and migration and is seen as a modulator of the tumor microenvironment. Following proteolytic cleavage of its extracellular domain in a process called shedding, Sdc-1 can act as a paracrine effector. The loss of Sdc-1 expression is associated with low differentiation of cervical carcinoma and with an increased rate of lymph node metastases. Here, we analyzed the clinical impact of Sdc-1 expression by analysis of public gene expression datasets and studied the effect of an overexpression of Sdc-1 and its membrane-bound and soluble forms on the malignant properties of the human cervical carcinoma cell line HeLa through functional analysis. For this purpose, the HeLa cells were stably transfected with the control plasmid pcDNA3.1 and three different Sdc-1-DNA constructs,encoding wild-type, permanently membrane-bound, and constitutively soluble Sdc-1. In clinical specimens, Sdc-1 mRNA was more highly expressed in local tumor tissues than in normal and metastatic cervical cancer tissues. Moreover, high Sdc-1 expression correlated with a poor prognosis in Kaplan-Meier survival analysis, suggesting the important role of Sdc-1 in the progression of this type of cancer. In vitro, we found that the soluble, as well as the permanently membrane-bound forms of Sdc-1 modulated the proliferation and the cell cycle, while membrane-bound Sdc1 regulated HeLa cell apoptosis. The overexpression of Sdc-1 and its soluble form increased invasiveness. In vitro scratch/wound healing assay, showed reduced Sdc-1-dependent cell motility which was linked to the Rho-GTPase signaling pathway. In conclusion, in cervical cancer Sdc-1 modulates pathogenetically relevant processes, which depend on the membrane-association of Sdc-1.
Collapse
Affiliation(s)
- Katharina Hilgers
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | | | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
11
|
Khorrami S, Zamani H, Hasanzadeh M, Mehramiz M, Soleimani A, Zare Marzouni H, Ferns GA, Esmaeili H, Avan A. Association of a genetic variant in Interleukin-10 gene with increased risk and inflammation associated with cervical cancer. Gene 2022; 807:145933. [PMID: 34464678 DOI: 10.1016/j.gene.2021.145933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/13/2020] [Accepted: 08/26/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Cervical-cancer is among the most commonly diagnosed cancers in women, and infection with human papillomavirus (HPV) is associated with an increased risk of cervical cancer and altered serum concentrations of inflammatory cytokines. We have explored the association between a genetic variation in the Interleukin-10 (IL-10) gene (rs1800896) and cervical cancer risk and its relationship with tissue Interferon gamma (IFN-γ), Transforming growth factor beta (TGF-β), Tumor necrosis factor alpha (TNF-α) concentrations in women with cervical cancer. METHODS A total of 315 women with, or without cervical cancer, were recruited into the study. DNA was extracted from cervical cells, and genotyping was undertaken using Taq-man real-time PCR. The genotype frequency and allele distribution were analyzed together with their association with pathological data. The association of the rs1800896 gene variation with tissue levels of the inflammatory cytokines was also investigated. RESULTS Our data showed a significant association between the A allele of the rs1800896 gene variant and the presence of cervical cancer. In particular, patients with AG/AA genotypes had an increased risk of cervical cancer with an odds ratio of 1.929 (95% confidence interval [CI]: 0.879-4.23, P < 0.001) in a recessive model, compared with the GG genotype. Also, the tissue concentrations of IFN-γ, TGF-β, and TNF-α in cervical tissues were significantly higher in women with cervical cancer (P < 0.001) and were associated with the AA genotype. CONCLUSION We have found an association between the polymorphism rs1800896 in the IL-10 gene and an increased risk of cervical cancer as well as a higher level of tissue inflammatory cytokines. Further investigations are necessary on the value of emerging biomarkers for the risk stratification for the management of cervical cancer patients.
Collapse
Affiliation(s)
- Shadi Khorrami
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hojjatolah Zamani
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehraneh Mehramiz
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atena Soleimani
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Zare Marzouni
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Habibollah Esmaeili
- Department of Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Tumor Escape Phenotype in Bladder Cancer Is Associated with Loss of HLA Class I Expression, T-Cell Exclusion and Stromal Changes. Int J Mol Sci 2021; 22:ijms22147248. [PMID: 34298868 PMCID: PMC8307653 DOI: 10.3390/ijms22147248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer eradication and clinical outcome of immunotherapy depend on tumor cell immunogenicity, including HLA class I (HLA-I) and PD-L1 expression on malignant cells, and on the characteristics of the tumor microenvironment, such as tumor immune infiltration and stromal reaction. Loss of tumor HLA-I is a common mechanism of immune escape from cytotoxic T lymphocytes and is linked to cancer progression and resistance to immunotherapy with the inhibitors of PD-L1/PD-1 signaling. Here we observed that HLA-I loss in bladder tumors is associated with T cell exclusion and tumor encapsulation with stromal elements rich in FAP-positive cells. In addition, PD-L1 upregulation in HLA-I negative tumors demonstrated a correlation with high tumor grade and worse overall- and cancer-specific survival of the patients. These changes define common immuno-morphological signatures compatible with cancer immune escape and acquired resistance to therapeutic interventions across different types of malignancy. They also may contribute to the search of new targets for cancer treatment, such as FAP-expressing cancer-associated fibroblasts, in refractory bladder tumors.
Collapse
|
13
|
Galgaro BC, Beckenkamp LR, van den M Nunnenkamp M, Korb VG, Naasani LIS, Roszek K, Wink MR. The adenosinergic pathway in mesenchymal stem cell fate and functions. Med Res Rev 2021; 41:2316-2349. [PMID: 33645857 DOI: 10.1002/med.21796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) play an important role in tissue homeostasis and damage repair through their ability to differentiate into cells of different tissues, trophic support, and immunomodulation. These properties made them attractive for clinical applications in regenerative medicine, immune disorders, and cell transplantation. However, despite multiple preclinical and clinical studies demonstrating beneficial effects of MSCs, their native identity and mechanisms of action remain inconclusive. Since its discovery, the CD73/ecto-5'-nucleotidase is known as a classic marker for MSCs, but its role goes far beyond a phenotypic characterization antigen. CD73 contributes to adenosine production, therefore, is an essential component of purinergic signaling, a pathway composed of different nucleotides and nucleosides, which concentrations are finely regulated by the ectoenzymes and receptors. Thus, purinergic signaling controls pathophysiological functions such as proliferation, migration, cell fate, and immune responses. Despite the remarkable progress already achieved in considering adenosinergic pathway as a therapeutic target in different pathologies, its role is not fully explored in the context of the therapeutic functions of MSCs. Therefore, in this review, we provide an overview of the role of CD73 and adenosine-mediated signaling in the functions ascribed to MSCs, such as homing and proliferation, cell differentiation, and immunomodulation. Additionally, we will discuss the pathophysiological role of MSCs, via CD73 and adenosine, in different diseases, as well as in tumor development and progression. A better understanding of the adenosinergic pathway in the regulation of MSCs functions will help to provide improved therapeutic strategies applicable in regenerative medicine.
Collapse
Affiliation(s)
- Bruna C Galgaro
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Liziane R Beckenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Martha van den M Nunnenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitória G Korb
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Liliana I S Naasani
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń, Poland
| | - Márcia R Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
14
|
CCL25 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:99-111. [PMID: 34286444 DOI: 10.1007/978-3-030-62658-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multiple checkpoint mechanisms are overridden by cancer cells in order to develop into a tumor. Neoplastic cells, while constantly changing during the course of cancer progression, also craft their surroundings to meet their growing needs. This crafting involves changing cell surface receptors, affecting response to extracellular signals and secretion of signals that affect the nearby cells and extracellular matrix architecture. This chapter briefly comprehends the non-cancer cells facilitating the cancer growth and elaborates on the notable role of the CCR9-CCL25 chemokine axis in shaping the tumor microenvironment (TME), directly and via immune cells. Association of increased CCR9 and CCL25 levels in various tumors has demonstrated the significance of this axis as a tool commonly used by cancer to flourish. It is involved in attracting immune cells in the tumor and determining their fate via various direct and indirect mechanisms and, leaning the TME toward immunosuppressive state. Besides, elevated CCR9-CCL25 signaling allows survival and rapid proliferation of cancer cells in an otherwise repressive environment. It modulates the intra- and extracellular protein matrix to instigate tumor dissemination and creates a supportive metastatic niche at the secondary sites. Lastly, this chapter abridges the latest research efforts and challenges in using the CCR9-CCL25 axis as a cancer-specific target.
Collapse
|
15
|
Yu R, Jin L, Li F, Fujimoto M, Wei Q, Lin Z, Ren X, Jin Q, Li H, Meng F, Jin G. Dihydroartemisinin inhibits melanoma by regulating CTL/Treg anti-tumor immunity and STAT3-mediated apoptosis via IL-10 dependent manner. J Dermatol Sci 2020; 99:193-202. [PMID: 32859456 DOI: 10.1016/j.jdermsci.2020.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND It has been shown that dihydroartemisinin (DHA) is effective in the treatment of malaria. Recently studies have demonstrated that DHA also regulates tumor cell growth, angiogenesis, T cell differentiation and generation. However, how DHA affects melanoma development remains poorly defined. OBJECTIVES To investigate the effects of DHA on the proliferation and migration of melanoma in vivo and in vitro, and to explore its possible mechanism. METHODS B16F10 cells and melanoma-bearing BALB/c mice were used to investigate the effects of DHA on melanoma. RESULTS DHA had inhibitory effect on melanoma proliferation in a time-and dose-dependent manner. Treatment of DHA attenuated melanoma severity and histopathological changes in BALB/c mice. DHA also inhibited melanoma invasion, migration, and community formation in a dose-dependent manner. Flow cytometry revealed a significant increase in IFN-γ+CD8+ T cells in the DHA groups. In tumor microenvironment and spleen, DHA induced expansion of CD8+CTL, while, CD4+CD25+Foxp3+ regulatory T (Treg) cells and IL-10+CD4+CD25+ T cells were normalized by DHA treatment. DHA diminished expression of IL-10 and IL-6, and increased the expression of IFN-γ in the tumor and spleen. Moreover, DHA administration significantly promoted the mitochondrial apoptosis of melanoma by regulating the STAT3 pathway. CONCLUSION DHA induces mitochondrial apoptosis and alters cytokines expression by inhibiting the phosphorylation of STAT3. DHA improves anti-tumor immunity in mice through controlling CD8+CTL function by counteracting IL-10-dependent Treg cells suppression, which promises to be an alternative drug for melanoma.
Collapse
Affiliation(s)
- Ran Yu
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Linbo Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Fangfang Li
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Laboratory of Cutaneous Immunology, Osaka UniversityImmunology Frontier Research Center, Osaka, Japan
| | - Qiang Wei
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Zhenhua Lin
- Department of Pathology, Yanbian University Medical College, Yanji, China
| | - Xiangshan Ren
- Department of Pathology, Yanbian University Medical College, Yanji, China
| | - Quanxin Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Honghua Li
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Fanping Meng
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Guihua Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China.
| |
Collapse
|
16
|
The Effects of Mesenchymal Stem Cells on Antimelanoma Immunity Depend on the Timing of Their Administration. Stem Cells Int 2020; 2020:8842659. [PMID: 32695181 PMCID: PMC7368936 DOI: 10.1155/2020/8842659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
There is still a lively debate about whether mesenchymal stem cells (MSCs) promote or suppress antitumor immune response. Although several possible explanations have been proposed, including different numbers of injected and engrafted MSCs, heterogeneity in phenotype, and function of tumor cells, the exact molecular mechanisms responsible for opposite effects of MSCs in modulation of antitumor immunity are still unknown. Herewith, we used a B16F10 murine melanoma model to investigate whether timing of MSC administration in tumor-bearing mice was crucially important for their effects on antitumor immunity. MSCs, intravenously injected 24 h after melanoma induction (B16F10+MSC1d-treated mice), significantly enhanced natural killer (NK) and T cell-driven antitumor immunity, suppressed tumor growth, and improved survival of melanoma-bearing animals. Significantly higher plasma levels of antitumorigenic cytokines (TNF-α and IFN-γ), remarkably lower plasma levels of immunosuppressive cytokines (TGF-β and IL-10), and a significantly higher number of tumor-infiltrating, IFN-γ-producing, FasL- and granzyme B-expressing NK cells, IL-17-producing CD4+Th17 cells, IFN-γ- and TNF-α-producing CD4+Th1 cells, and CD8+cytotoxic T lymphocytes (CTLs) were observed in B16F10+MSC1d-treated mice. On the contrary, MSCs, injected 14 days after melanoma induction (B16F10+MSC14d-treated mice), promoted tumor growth by suppressing antigen-presenting properties of tumor-infiltrating dendritic cells (DCs) and macrophages and by reducing tumoricidal capacity of NK cells and T lymphocytes. Significantly higher plasma levels of TGF-β and IL-10, remarkably lower plasma levels of TNF-α and IFN-γ, and significantly reduced number of tumor-infiltrating, I-A-expressing, and IL-12-producing macrophages, CD80- and I-A-expressing DCs, granzyme B-expressing CTLs and NK cells, IFN-γ- and IL-17-producing CTLs, CD4+Th1, and Th17 cells were observed in B16F10+MSC14d-treated animals. In summing up, the timing of MSC administration into the tumor microenvironment was crucially important for MSC-dependent modulation of antimelanoma immunity. MSCs transplanted during the initial phase of melanoma growth exerted tumor-suppressive effect, while MSCs injected during the progressive stage of melanoma development suppressed antitumor immunity and enhanced tumor expansion.
Collapse
|
17
|
Torres-Pineda DB, Mora-García MDL, García-Rocha R, Hernández-Montes J, Weiss-Steider B, Montesinos-Montesinos JJ, Don-López CA, Marín-Aquino LA, Muñóz-Godínez R, Ibarra LRÁ, López Romero R, Monroy-García A. Adenosine augments the production of IL-10 in cervical cancer cells through interaction with the A 2B adenosine receptor, resulting in protection against the activity of cytotoxic T cells. Cytokine 2020; 130:155082. [PMID: 32259773 DOI: 10.1016/j.cyto.2020.155082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
Cervical cancer (CeCa) produces large amounts of IL-10, which downregulates the major histocompatibility complex class I molecules (HLA-I) in cancer cells and inhibits the immune response mediated by cytotoxic T lymphocytes (CTLs). In this study, we analyzed the ability of CeCa cells to produce IL-10 through the CD73-adenosine pathway and its effect on the downregulation of HLA-I molecules to evade CTL-mediated immune recognition. CeCa cells cultured in the presence of ≥10 µM AMP or adenosine produced 4.5-6 times as much IL-10 as unstimulated cells. The silencing of CD73 or the blocking of A2BR with the specific antagonist MRS1754 reversed this effect. In addition, IL-10 decreased the expression of HLA-I molecules, resulting in the protection of CeCa cells against the cytotoxic activity of CTLs. The addition of MRS1754 or anti-IL-10 reversed the decrease in HLA-I molecules and favored the cytotoxic activity of CTLs. These results strongly suggest the presence of a feedback loop encompassing the adenosinergic pathway, the production of IL-10, and the downregulation of HLA-I molecules in CeCa cells that favors immune evasion and thus tumor progression. This pathway may have clinical importance as a therapeutic target.
Collapse
Affiliation(s)
- Daniela Berenice Torres-Pineda
- Laboratorio de Inmunología y Cáncer, UIMEO, H Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico; Programa de Posgrado en Ciencias Biológicas, UNAM, Ciudad de México, Mexico.
| | | | - Rosario García-Rocha
- Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, Mexico
| | - Jorge Hernández-Montes
- Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, Mexico
| | - Benny Weiss-Steider
- Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, Mexico
| | - Juan José Montesinos-Montesinos
- Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | | | - Luis Antonio Marín-Aquino
- Laboratorio de Inmunología y Cáncer, UIMEO, H Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Ricardo Muñóz-Godínez
- Laboratorio de Inmunología y Cáncer, UIMEO, H Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | | | - Ricardo López Romero
- Laboratorio de Oncología Genómica, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Alberto Monroy-García
- Laboratorio de Inmunología y Cáncer, UIMEO, H Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico; Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, Mexico.
| |
Collapse
|
18
|
Onyedibe KI, Wang M, Sintim HO. ENPP1, an Old Enzyme with New Functions, and Small Molecule Inhibitors-A STING in the Tale of ENPP1. Molecules 2019; 24:molecules24224192. [PMID: 31752288 PMCID: PMC6891441 DOI: 10.3390/molecules24224192] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022] Open
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase I (ENPP1) was identified several decades ago as a type II transmembrane glycoprotein with nucleotide pyrophosphatase and phosphodiesterase enzymatic activities, critical for purinergic signaling. Recently, ENPP1 has emerged as a critical phosphodiesterase that degrades the stimulator of interferon genes (STING) ligand, cyclic GMP-AMP (cGAMP). cGAMP or analogs thereof have emerged as potent immunostimulatory agents, which have potential applications in immunotherapy. This emerging role of ENPP1 has placed this "old" enzyme at the frontier of immunotherapy. This review highlights the roles played by ENPP1, the mechanism of cGAMP hydrolysis by ENPP1, and small molecule inhibitors of ENPP1 with potential applications in diverse disease states, including cancer.
Collapse
Affiliation(s)
- Kenneth I. Onyedibe
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; (K.I.O.); (M.W.)
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, IN 47907, USA
| | - Modi Wang
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; (K.I.O.); (M.W.)
| | - Herman O. Sintim
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; (K.I.O.); (M.W.)
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, IN 47907, USA
- Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
- Correspondence: ; Tel.: +1-(765)-496-6078; Fax: +1-(765)-494-0239
| |
Collapse
|
19
|
Knobloch TJ, Peng J, Hade EM, Cohn DE, Ruffin MT, Schiano MA, Calhoun BC, McBee WC, Lesnock JL, Gallion HH, Pollock J, Lu B, Oghumu S, Zhang Z, Sears MT, Ogbemudia BE, Perrault JT, Weghorst LC, Strawser E, DeGraffinreid CR, Paskett ED, Weghorst CM. Inherited alterations of TGF beta signaling components in Appalachian cervical cancers. Cancer Causes Control 2019; 30:1087-1100. [PMID: 31435875 PMCID: PMC6768402 DOI: 10.1007/s10552-019-01221-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE This study examined targeted genomic variants of transforming growth factor beta (TGFB) signaling in Appalachian women. Appalachian women with cervical cancer were compared to healthy Appalachian counterparts to determine whether these polymorphic alleles were over-represented within this high-risk cancer population, and whether lifestyle or environmental factors modified the aggregate genetic risk in these Appalachian women. METHODS Appalachian women's survey data and blood samples from the Community Awareness, Resources, and Education (CARE) CARE I and CARE II studies (n = 163 invasive cervical cancer cases, 842 controls) were used to assess gene-environment interactions and cancer risk. Polymorphic allele frequencies and socio-behavioral demographic measurements were compared using t tests and χ2 tests. Multivariable logistic regression was used to evaluate interaction effects between genomic variance and demographic, behavioral, and environmental characteristics. RESULTS Several alleles demonstrated significant interaction with smoking (TP53 rs1042522, TGFB1 rs1800469), alcohol consumption (NQO1 rs1800566), and sexual intercourse before the age of 18 (TGFBR1 rs11466445, TGFBR1 rs7034462, TGFBR1 rs11568785). Interestingly, we noted a significant interaction between "Appalachian self-identity" variables and NQO1 rs1800566. Multivariable logistic regression of cancer status in an over-dominant TGFB1 rs1800469/TGFBR1 rs11568785 model demonstrated a 3.03-fold reduction in cervical cancer odds. Similar decreased odds (2.78-fold) were observed in an over-dominant TGFB1 rs1800469/TGFBR1 rs7034462 model in subjects who had no sexual intercourse before age 18. CONCLUSIONS This study reports novel associations between common low-penetrance alleles in the TGFB signaling cascade and modified risk of cervical cancer in Appalachian women. Furthermore, our unexpected findings associating Appalachian identity and NQO1 rs1800566 suggests that the complex environmental exposures that contribute to Appalachian self-identity in Appalachian cervical cancer patients represent an emerging avenue of scientific exploration.
Collapse
Affiliation(s)
- Thomas J Knobloch
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA.
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| | - Juan Peng
- Department of Biomedical Informatics, Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Erinn M Hade
- Department of Biomedical Informatics, Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - David E Cohn
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wexner Medical Center, College of Medicine, The Ohio State University Columbus, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Mack T Ruffin
- Department of Family and Community Medicine, Milton S. Hershey Medical Center, Penn State University, Hersey, PA, 17033, USA
| | - Michael A Schiano
- Department of Obstetrics & Gynecology, West Virginia University, Charleston, WV, 26505, USA
- Charleston Area Medical Center Health System, Charleston, WV, 25302, USA
| | - Byron C Calhoun
- Department of Obstetrics & Gynecology, West Virginia University, Charleston, WV, 26505, USA
- Charleston Area Medical Center Health System, Charleston, WV, 25302, USA
| | | | | | | | - Jondavid Pollock
- Wheeling Hospital, Schiffler Cancer Center, Wheeling, WV, 26003, USA
| | - Bo Lu
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Steve Oghumu
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Zhaoxia Zhang
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Marta T Sears
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | | | - Joseph T Perrault
- Division of Cancer Prevention and Control, Wexner Medical Center, College of Medicine, The Ohio State University Columbus, Columbus, OH, 43210, USA
| | - Logan C Weghorst
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Erin Strawser
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Cecilia R DeGraffinreid
- Division of Cancer Prevention and Control, Wexner Medical Center, College of Medicine, The Ohio State University Columbus, Columbus, OH, 43210, USA
| | - Electra D Paskett
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
- Division of Cancer Prevention and Control, Wexner Medical Center, College of Medicine, The Ohio State University Columbus, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Christopher M Weghorst
- College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| |
Collapse
|
20
|
He P, Zhou W, Liu M, Chen Y. Recent Advances of Small Molecular Regulators Targeting G Protein- Coupled Receptors Family for Oncology Immunotherapy. Curr Top Med Chem 2019; 19:1464-1483. [PMID: 31264549 DOI: 10.2174/1568026619666190628115644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022]
Abstract
The great clinical success of chimeric antigen receptor T cell (CAR-T) and PD-1/PDL-1 inhibitor therapies suggests the drawing of a cancer immunotherapy age. However, a considerable proportion of cancer patients currently receive little benefit from these treatment modalities, indicating that multiple immunosuppressive mechanisms exist in the tumor microenvironment. In this review, we mainly discuss recent advances in small molecular regulators targeting G Protein-Coupled Receptors (GPCRs) that are associated with oncology immunomodulation, including chemokine receptors, purinergic receptors, prostaglandin E receptor EP4 and opioid receptors. Moreover, we outline how they affect tumor immunity and neoplasia by regulating immune cell recruitment and modulating tumor stromal cell biology. We also summarize the data from recent clinical advances in small molecular regulators targeting these GPCRs, in combination with immune checkpoints blockers, such as PD-1/PDL-1 and CTLA4 inhibitors, for cancer treatments.
Collapse
Affiliation(s)
- Peng He
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenbo Zhou
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
21
|
Silva JR, Sales NS, Silva MO, Aps LRMM, Moreno ACR, Rodrigues EG, Ferreira LCS, Diniz MO. Expression of a soluble IL-10 receptor enhances the therapeutic effects of a papillomavirus-associated antitumor vaccine in a murine model. Cancer Immunol Immunother 2019; 68:753-763. [PMID: 30806747 PMCID: PMC11028134 DOI: 10.1007/s00262-018-02297-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/28/2018] [Indexed: 12/25/2022]
Abstract
The presence of IL-10, produced either by tumor cells or immunosuppressive cells, is frequently associated with a poor prognosis for cancer progression. It may also negatively impact anticancer treatments, such as immunotherapies, that otherwise would promote the activation of cytotoxic T cells capable of detecting and destroying malignant cells. In the present study, we evaluated a new adjuvant approach for anticancer immunotherapy using a plasmid vector encoding a soluble form of the IL-10 receptor (pIL-10R). pIL-10R was coadministered to mice with a DNA vaccine encoding the type 16 human papillomavirus (HPV-16) E7 oncoprotein genetically fused with glycoprotein D of herpes simplex virus (HSV) (pgDE7h). Immunization regimens based on the coadministration of pIL-10R and pgDE7h enhanced the antitumor immunity elicited in mice injected with TC-1 cells, which express HPV-16 oncoproteins. The administration of the DNA vaccines by in vivo electroporation further enhanced the anticancer effects of the vaccines, leading to the activation of tumor-infiltrating polyfunctional E7-specific cytotoxic CD8+ T cells and control of the expansion of immunosuppressive cells. In addition, the combination of immunotherapy and pIL-10R allowed the control of tumors in more advanced growth stages that otherwise would not be treatable by the pgDE7h vaccine. In conclusion, the proposed treatment involving the expression of IL-10R enhanced the antitumor protective immunity induced by pgDE7h administration and may contribute to the development of more efficient clinical interventions against HPV-induced tumors.
Collapse
Affiliation(s)
- Jamile R Silva
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Natiely S Sales
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Mariângela O Silva
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Luana R M M Aps
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Ana C R Moreno
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Elaine G Rodrigues
- Tumor Immunobiology Laboratory, Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Luís C S Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil.
| | - Mariana O Diniz
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
- Division of Infection and Immunity, University College London, 5 University St, Bloomsbury, London, WC1E 6JF, UK
| |
Collapse
|
22
|
Stress responses in stromal cells and tumor homeostasis. Pharmacol Ther 2019; 200:55-68. [PMID: 30998941 DOI: 10.1016/j.pharmthera.2019.04.004] [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: 03/06/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023]
Abstract
In most (if not all) solid tumors, malignant cells are outnumbered by their non-malignant counterparts, including immune, endothelial and stromal cells. However, while the mechanisms whereby cancer cells adapt to microenvironmental perturbations have been studied in great detail, relatively little is known on stress responses in non-malignant compartments of the tumor microenvironment. Here, we discuss the mechanisms whereby cancer-associated fibroblasts and other cellular components of the tumor stroma react to stress in the context of an intimate crosstalk with malignant, endothelial and immune cells, and how such crosstalk influences disease progression and response to treatment.
Collapse
|
23
|
Ávila-Ibarra LR, Mora-García MDL, García-Rocha R, Hernández-Montes J, Weiss-Steider B, Montesinos JJ, Lizano Soberon M, García-López P, López CAD, Torres-Pineda DB, Chacón-Salinas R, Vallejo-Castillo L, Pérez-Tapia SM, Monroy-García A. Mesenchymal Stromal Cells Derived from Normal Cervix and Cervical Cancer Tumors Increase CD73 Expression in Cervical Cancer Cells Through TGF-β1 Production. Stem Cells Dev 2019; 28:477-488. [PMID: 30696359 DOI: 10.1089/scd.2018.0183] [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] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) in the tumor microenvironment (TME) participate together with tumor cells to suppress antitumor effector cells through the production of immunosuppressive factors, such as transforming growth factor-beta 1 (TGF-β1). Furthermore, TGF-β1 can induce 5'-nucleotidase (CD73) expression in various cell types; this functional activity is associated with the production of adenosine (Ado), which is an immunosuppressive nucleoside. In this study, we provide evidence that coculture of MSCs derived from cervical tumors (CeCa-MSC) with CeCa tumor cells increases CD73 expression in tumor cells and the capacity of these cells to generate Ado in a MSC ratio-dependent manner. Interestingly, the increase in CD73 in the CeCa cell membrane corresponded to an increase in the TGF-β1 expression level in the tumor cells and the TGF-β1 content in the supernatants of the CeCa/CeCa-MSC cocultures. The addition of anti-hTGF-β neutralizing antibodies strongly reversed CD73 expression in the tumor cells. This phenomenon was not exclusive to CeCa-MSCs; coculture of MSCs derived from the normal cervix with CeCa cells produced similar results. These results suggest that the interaction of MSCs with CeCa tumor cells in the TME may condition higher TGF-β1 production to maintain an immunosuppressive status not only through the activity of this cytokine per se but also through its ability to induce CD73 expression in tumor cells and generate an immunosuppressive microenvironment rich in Ado.
Collapse
Affiliation(s)
- Luis Roberto Ávila-Ibarra
- 1 Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.,2 Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México.,3 Programa de Posgrado en Ciencias Biológicas, UNAM, Ciudad de México, México
| | | | - Rosario García-Rocha
- 2 Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Jorge Hernández-Montes
- 2 Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Benny Weiss-Steider
- 2 Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| | - Juan José Montesinos
- 4 Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Marcela Lizano Soberon
- 5 Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, UNAM, Ciudad de México, México
| | - Patricia García-López
- 6 Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, México
| | | | - Daniela Berenice Torres-Pineda
- 1 Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.,3 Programa de Posgrado en Ciencias Biológicas, UNAM, Ciudad de México, México
| | - Rommel Chacón-Salinas
- 7 Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Instituto Politécnico Nacional, Ciudad de México, México.,8 Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Ciudad de México, México
| | - Luis Vallejo-Castillo
- 7 Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Instituto Politécnico Nacional, Ciudad de México, México.,9 Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Ciudad de México, México
| | - Sonia Mayra Pérez-Tapia
- 7 Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Instituto Politécnico Nacional, Ciudad de México, México.,8 Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, ENCB-IPN, Ciudad de México, México
| | - Alberto Monroy-García
- 1 Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.,2 Laboratorio de Inmunobiología, UIDCC-UMIEZ, FES-Zaragoza, UNAM, Ciudad de México, México
| |
Collapse
|
24
|
Ma T, Wang X, Jiao Y, Wang H, Qi Y, Gong H, Zhang L, Jiang D. Interleukin 17 (IL-17)-Induced Mesenchymal Stem Cells Prolong the Survival of Allogeneic Skin Grafts. Ann Transplant 2018; 23:615-621. [PMID: 30166501 PMCID: PMC6248056 DOI: 10.12659/aot.909381] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have the potential of self-renewal and multi-differentiation and have a wide application prospect in organ transplantation for the effect of inducing immune tolerance. It has found that interleukin 17 (IL-17) could enhance the inhibition effect of MSCs on T cell proliferation and increase the immunosuppressive effect of MSCs. In this study, we aimed to investigate the effect of IL-17-induced MSCs on allograft survival time after transplantation. MATERIAL AND METHODS BMSCs were characterized by differential staining. The allogenic skin transplantations were performed and the BMSCs pre-treated by IL-17 were injected. To assess the immunosuppressive function of IL-17-induced BMSCs, the morphology of the grafts, the homing ability of the BMSCs, and the survival time of the grafts were analyzed. RESULTS BMSCs from BALB/c have multidirectional differentiation potential to differentiate into osteogenic, chondrogenic, and adipogenic lineage cells. IL-17-induced BMSCs prolonged the survival time of allogeneic skin grafts dramatically. We found that there were more labeled MSCs in the skin grafts, and the Treg subpopulations percentage, IL-10, and TGF-β were significantly increased, while the IFN-γ level was decreased compared to the control group and MSCs group. In conclusion, IL-17 can enhance the homing ability of MSCs and regulate the immunosuppressive function of MSC. CONCLUSIONS Our data demonstrate that IL-17 plays the crucial role in MSC homing behaviors and promotes immunosuppression of MSCs during transplantation procedures, suggesting that IL-17-pre-treated MSCs have potential to prolong graft survival and reduce transplant rejection.
Collapse
Affiliation(s)
- Tengxiao Ma
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland).,School of Medicine, Shandong University, Jinan, Shandong, China (mainland).,Department of Plastic Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, China (mainland)
| | - Xiao Wang
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland).,School of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Ya Jiao
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland).,School of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Haitao Wang
- School of Medicine, Shandong University, Jinan, Shandong, China (mainland).,Department of Pathology, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Yongjun Qi
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland).,School of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Hongmin Gong
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland).,School of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Longxiao Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Duyin Jiang
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland).,School of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
25
|
Bashaw AA, Leggatt GR, Chandra J, Tuong ZK, Frazer IH. Modulation of antigen presenting cell functions during chronic HPV infection. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2017; 4:58-65. [PMID: 29179871 PMCID: PMC5883240 DOI: 10.1016/j.pvr.2017.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022]
Abstract
High-risk human papillomaviruses (HR-HPV) infect basal keratinocytes, where in some individuals they evade host immune responses and persist. Persistent HR-HPV infection of the cervix causes precancerous neoplasia that can eventuate in cervical cancer. Dendritic cells (DCs) are efficient in priming/cross-priming antigen-specific T cells and generating antiviral and antitumor cytotoxic CD8+ T cells. However, HR-HPV have adopted various immunosuppressive strategies, with modulation of DC function crucial to escape from the host adaptive immune response. HPV E6 and E7 oncoproteins alter recruitment and localization of epidermal DCs, while soluble regulatory factors derived from HPV-induced hyperplastic epithelium change DC development and influence initiation of specific cellular immune responses. This review focuses on current evidence for HR-HPV manipulation of antigen presentation in dendritic cells and escape from host immunity.
Collapse
Affiliation(s)
- Abate Assefa Bashaw
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
26
|
Reconstruction of Craniomaxillofacial Bone Defects Using Tissue-Engineering Strategies with Injectable and Non-Injectable Scaffolds. J Funct Biomater 2017; 8:jfb8040049. [PMID: 29156629 PMCID: PMC5748556 DOI: 10.3390/jfb8040049] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023] Open
Abstract
Engineering craniofacial bone tissues is challenging due to their complex structures. Current standard autografts and allografts have many drawbacks for craniofacial bone tissue reconstruction; including donor site morbidity and the ability to reinstate the aesthetic characteristics of the host tissue. To overcome these problems; tissue engineering and regenerative medicine strategies have been developed as a potential way to reconstruct damaged bone tissue. Different types of new biomaterials; including natural polymers; synthetic polymers and bioceramics; have emerged to treat these damaged craniofacial bone tissues in the form of injectable and non-injectable scaffolds; which are examined in this review. Injectable scaffolds can be considered a better approach to craniofacial tissue engineering as they can be inserted with minimally invasive surgery; thus protecting the aesthetic characteristics. In this review; we also focus on recent research innovations with different types of stem-cell sources harvested from oral tissue and growth factors used to develop craniofacial bone tissue-engineering strategies.
Collapse
|
27
|
Espinoza-Sánchez NA, Vadillo E, Balandrán JC, Monroy-García A, Pelayo R, Fuentes-Pananá EM. Evidence of lateral transmission of aggressive features between different types of breast cancer cells. Int J Oncol 2017; 51:1482-1496. [PMID: 29048610 PMCID: PMC5643070 DOI: 10.3892/ijo.2017.4128] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BrC) is a major public health problem worldwide. The intra-tumoral heterogeneity and tumor cell plasticity importantly contribute to disease progression and treatment failure. However, the dynamic interactions between different tumor clones, as well as their contribution to tumor aggressiveness are still poorly understood. In this study, we provide evidence of a lateral transmission of aggressive features between aggressive and non-aggressive tumor cells, consisting of gain of expression of cancer stem cell markers, increased expression of CXCL12 receptors CXCR4 and CXCR7 and increased invasiveness in response to CXCL12, which correlated with high levels of secretion of pro-inflammatory mediators G-CSF, GM-CSF, MCP-1, IL-8 and metalloproteinases 1 and 2 by the aggressive cells. Noteworthy, we found no evidence of a TGF-β participation in the inducible-invasive phenotype. Altogether, our results provide evidence of communication between tumor cells with different potentials for aggressiveness, which could influence intra-tumoral population dynamics promoting the emergence of clones with novel functions. Understanding these interactions will provide better targets for diagnosis, prognosis and therapeutic strategies.
Collapse
Affiliation(s)
- Nancy Adriana Espinoza-Sánchez
- PhD Program in Biomedical Science, Medicine Faculty, National Autonomous University of Mexico, University City, Mexico City 04510, Mexico
| | - Eduardo Vadillo
- Department of Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Juan Carlos Balandrán
- Department of Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Alberto Monroy-García
- Oncology Research Unit, Oncology Hospital, Mexican Institute for Social Security, Mexico City 06720, Mexico
| | - Rosana Pelayo
- Oncology Research Unit, Oncology Hospital, Mexican Institute for Social Security, Mexico City 06720, Mexico
| | - Ezequiel M Fuentes-Pananá
- Virology and Cancer Research Unit, Children's Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
28
|
Mesiano G, Zini R, Montagner G, Bianchi N, Manfredini R, Chillemi A, Aglietta M, Grignani G, Lampronti I, Fiorino E, Malavasi F, Sangiolo D, Gambari R, Ferrari D. Analytic and Dynamic Secretory Profile of Patient-Derived Cytokine-Induced Killer Cells. Mol Med 2017; 23:235-246. [PMID: 28805233 DOI: 10.2119/molmed.2017.00084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/01/2017] [Indexed: 12/18/2022] Open
Abstract
Adoptive immunotherapy with Cytokine Induced Killer (CIK) cells has shown antitumor activity against several kinds of cancers in preclinical models and clinical trials. CIK cells are a subset of ex vivo expanded T lymphocytes with T-NK phenotype and MHC-unrestricted antitumor activity. Literature provides scanty information on cytokines, chemokines and growth factors secreted by CIK cells. Therefore, we investigated the secretory profile of CIK cells generated from tumor patients. The secretome analysis was performed at specific time points (day 1, day 14 and day 21) of CIK cells expansion. Mature CIK cells (day 21) produce a great variety of interleukins and secreted proteins that can be divided into 3 groups based on their secretion quantity: high (IL-13, RANTES, MIP-1α and 1β), medium (IL-1Ra, IL-5, IL-8, IL-10, IL-17, IP-10, INF-γ, VEGF and GMCSF) and low (IL-1β, IL-4, IL-6, IL-7, IL-9, IL-12, IL-15, Eotaxin, PDGF-bb, FGF basic, G-CSF and MCP-1) secreted. Moreover, comparing PBMC (day 1) and mature CIK cells (day 14 and 21) secretome, we observed that IL-5, IL-10, IL-13, GM-CSF, VEGF resulted greatly up-regulated, while IL-1β, IL-6, IL-8, IL-15, IL-17, eotaxin, MCP-1, and RANTES were down-regulated. We also performed a gene expression profile analysis of patient-derived CIK cells showing that mRNA for the different cytokines and secreted proteins were modulated during PBMC to CIK differentiation. We highlighted previously unknown secretory properties and provided for the first time a comprehensive molecular characterization of CIK cells. Our findings provide rationale to explore the functional implications and possible therapeutic modulation of CIK secretome.
Collapse
Affiliation(s)
- Giulia Mesiano
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Montagner
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonella Chillemi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Massimo Aglietta
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Giovanni Grignani
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Ilaria Lampronti
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Erika Fiorino
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Fabio Malavasi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Dario Sangiolo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Roberto Gambari
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Davide Ferrari
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy.,Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
29
|
Yang X, Yang J, Li X, Ma W, Zou H. Bone marrow-derived mesenchymal stem cells inhibit T follicular helper cell in lupus-prone mice. Lupus 2017; 27:49-59. [PMID: 28537524 DOI: 10.1177/0961203317711013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The objective of this paper is to analyze the role of bone marrow-derived mesenchymal stem cells (BM-MSCs) on the differentiation of T follicular helper (Tfh) cells in lupus-prone mice. Methods Bone marrow cells were isolated from C57BL/6 (B6) mice and cultured in vitro, and surface markers were identified by flow cytometry. Naïve CD4+ T cells, splenocytes and Tfh cells were isolated from B6 mice spleens and co-cultured with BM-MSCs. The proliferation and the differentiation of CD4+ T cells and Tfh cells were analyzed by flow cytometry. Lupus-prone MRL/Mp-lpr/lpr (MRL/lpr) mice were treated via intravenous injection with expanded BM-MSCs, the differentiation of Tfh cells was detected, and the relief of lupus nephritis was analyzed. Results MSCs could be successfully induced from bone marrow cells, and cultured BM-MSCs could inhibit T cell proliferation dose-dependently. BM-MSCs could prevent Tfh cell development from naïve CD4+ T cells and splenocytes. BM-MSCs could inhibit IL-21 gene expression and cytokine production and inhibit isolated Tfh cells and STAT3 phosphorylation. In vivo study proved that BM-MSCs intravenous injection could effectively inhibit Tfh cell expansion and IL-21 production, alleviate lupus nephritis, and prolong the survival rate of lupus-prone mice. Conclusions BM-MSCs could effectively inhibit the differentiation of Tfh cells both in vitro and in vivo. BM-MSC treatment could relieve lupus nephritis, which indicates that BM-MSCs might be a promising therapeutic method for the treatment of SLE.
Collapse
Affiliation(s)
- X Yang
- 1 Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,2 Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - J Yang
- 3 Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - X Li
- 4 Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - W Ma
- 5 Central Laboratory, Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - H Zou
- 1 Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,2 Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Sepúlveda C, Palomo I, Fuentes E. Role of adenosine A2b receptor overexpression in tumor progression. Life Sci 2016; 166:92-99. [DOI: 10.1016/j.lfs.2016.10.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 10/01/2016] [Accepted: 10/07/2016] [Indexed: 02/07/2023]
|
31
|
Otter S, Franklin A, Ajaz M, Stewart A. Improving the efficiency of image guided brachytherapy in cervical cancer. J Contemp Brachytherapy 2016; 8:557-565. [PMID: 28115963 PMCID: PMC5241377 DOI: 10.5114/jcb.2016.64452] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/16/2016] [Indexed: 11/21/2022] Open
Abstract
Brachytherapy is an essential component of the treatment of locally advanced cervical cancers. It enables the dose to the tumor to be boosted whilst allowing relative sparing of the normal tissues. Traditionally, cervical brachytherapy was prescribed to point A but since the GEC-ESTRO guidelines were published in 2005, there has been a move towards prescribing the dose to a 3D volume. Image guided brachytherapy has been shown to reduce local recurrence, and improve survival and is optimally predicated on magnetic resonance imaging. Radiological studies, patient workflow, operative parameters, and intensive therapy planning can represent a challenge to clinical resources. This article explores the ways, in which 3D conformal brachytherapy can be implemented and draws findings from recent literature and a well-developed hospital practice in order to suggest ways to improve the efficiency and efficacy of a brachytherapy service. Finally, we discuss relatively underexploited translational research opportunities.
Collapse
Affiliation(s)
- Sophie Otter
- St Luke’s Cancer Centre, Royal Surrey County Hospital, Guildford
| | - Adrian Franklin
- St Luke’s Cancer Centre, Royal Surrey County Hospital, Guildford
| | - Mazhar Ajaz
- St Luke’s Cancer Centre, Royal Surrey County Hospital, Guildford
- University of Surrey, Guildford, United Kingdom
| | - Alexandra Stewart
- St Luke’s Cancer Centre, Royal Surrey County Hospital, Guildford
- University of Surrey, Guildford, United Kingdom
| |
Collapse
|
32
|
de Lourdes Mora-García M, García-Rocha R, Morales-Ramírez O, Montesinos JJ, Weiss-Steider B, Hernández-Montes J, Ávila-Ibarra LR, Don-López CA, Velasco-Velázquez MA, Gutiérrez-Serrano V, Monroy-García A. Mesenchymal stromal cells derived from cervical cancer produce high amounts of adenosine to suppress cytotoxic T lymphocyte functions. J Transl Med 2016; 14:302. [PMID: 27782859 PMCID: PMC5080842 DOI: 10.1186/s12967-016-1057-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 10/10/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In recent years, immunomodulatory mechanisms of mesenchymal stem/stromal cells (MSCs) from bone marrow and other "classic" sources have been described. However, the phenotypic and functional properties of tumor MSCs are poorly understood. The aim of this study was to analyze the immunosuppressive capacity of cervical cancer-derived MSCs (CeCa-MSCs) on effector T lymphocytes through the purinergic pathway. METHODS We determined the expression and functional activity of the membrane-associated ectonucleotidases CD39 and CD73 on CeCa-MSCs and normal cervical tissue-derived MSCs (NCx-MSCs). We also analyzed their immunosuppressive capacity to decrease proliferation, activation and effector cytotoxic T (CD8+) lymphocyte function through the generation of adenosine (Ado). RESULTS We detected that CeCa-MSCs express higher levels of CD39 and CD73 ectonucleotidases in cell membranes compared to NCx-MSCs, and that this feature was associated with the ability to strongly suppress the proliferation, activation and effector functions of cytotoxic T-cells through the generation of large amounts of Ado from the hydrolysis of ATP, ADP and AMP nucleotides. CONCLUSIONS This study suggests that CeCa-MSCs play an important role in the suppression of the anti-tumor immune response in CeCa through the purinergic pathway.
Collapse
Affiliation(s)
| | - Rosario García-Rocha
- Immunobiology Laboratory, Cellular Differentiation and Cancer Unit, FES-Zaragoza, UNAM, Mexico City, Mexico.,Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Omar Morales-Ramírez
- Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Juan José Montesinos
- Mesenchymal Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Benny Weiss-Steider
- Immunobiology Laboratory, Cellular Differentiation and Cancer Unit, FES-Zaragoza, UNAM, Mexico City, Mexico
| | - Jorge Hernández-Montes
- Immunobiology Laboratory, Cellular Differentiation and Cancer Unit, FES-Zaragoza, UNAM, Mexico City, Mexico
| | - Luis Roberto Ávila-Ibarra
- Immunobiology Laboratory, Cellular Differentiation and Cancer Unit, FES-Zaragoza, UNAM, Mexico City, Mexico.,Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | | | | | - Vianey Gutiérrez-Serrano
- Immunobiology Laboratory, Cellular Differentiation and Cancer Unit, FES-Zaragoza, UNAM, Mexico City, Mexico.,Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Alberto Monroy-García
- Immunobiology Laboratory, Cellular Differentiation and Cancer Unit, FES-Zaragoza, UNAM, Mexico City, Mexico. .,Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico. .,, Oriente 170 No. 160 Colonia Moctezuma 2a Sección Delegación Venustiano Carranza, 15530, Mexico City, Mexico.
| |
Collapse
|