1
|
Erasha AM, EL-Gendy H, Aly AS, Fernández-Ortiz M, Sayed RKA. The Role of the Tumor Microenvironment (TME) in Advancing Cancer Therapies: Immune System Interactions, Tumor-Infiltrating Lymphocytes (TILs), and the Role of Exosomes and Inflammasomes. Int J Mol Sci 2025; 26:2716. [PMID: 40141358 PMCID: PMC11942452 DOI: 10.3390/ijms26062716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Understanding how different contributors within the tumor microenvironment (TME) function and communicate is essential for effective cancer detection and treatment. The TME encompasses all the surroundings of a tumor such as blood vessels, fibroblasts, immune cells, signaling molecules, exosomes, and the extracellular matrix (ECM). Subsequently, effective cancer therapy relies on addressing TME alterations, known drivers of tumor progression, immune evasion, and metastasis. Immune cells and other cell types act differently under cancerous conditions, either driving or hindering cancer progression. For instance, tumor-infiltrating lymphocytes (TILs) include lymphocytes of B and T cell types that can invade malignancies, bringing in and enhancing the ability of immune system to recognize and destroy cancer cells. Therefore, TILs display a promising approach to tackling the TME alterations and have the capability to significantly hinder cancer progression. Similarly, exosomes and inflammasomes exhibit a dual effect, resulting in either tumor progression or inhibition depending on the origin of exosomes, type of inflammasome and tumor. This review will explore how cells function in the presence of a tumor, the communication between cancer cells and immune cells, and the role of TILs, exosomes and inflammasomes within the TME. The efforts in this review are aimed at garnering interest in safer and durable therapies for cancer, in addition to providing a promising avenue for advancing cancer therapy and consequently improving survival rates.
Collapse
Affiliation(s)
- Atef M. Erasha
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Hanem EL-Gendy
- Department of Pharmacology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Ahmed S. Aly
- Department of Animal Production, Faculty of Agriculture, Ain Shams University, Cairo 11241, Egypt;
| | - Marisol Fernández-Ortiz
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Ramy K. A. Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
| |
Collapse
|
2
|
Pluetrattanabha N, Direksunthorn T, Ahmad I, Jyothi SR, Shit D, Singh AK, Chauhan AS. Inflammasome activation in melanoma progression: the latest update concerning pathological role and therapeutic value. Arch Dermatol Res 2025; 317:258. [PMID: 39820618 DOI: 10.1007/s00403-025-03802-1] [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: 12/11/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
The progression of melanoma is a complex process influenced by both internal and external cues which encourage the transition of tumour cells, uncontrolled growth, migration, and metastasis. Additionally, inflammation allows tumours to evade the immune system, contributing to cancer development. The inflammasome, a complex of many proteins, is crucial in enhancing immune responses to external and internal triggers. As a critical inflammatory mechanism, it contributes to the development of melanoma. These mechanisms may be triggered via various internal and external stimuli, causing the induction of specific enzymes such as caspase-1, caspase-11, or caspase-8. This, in turn, leads to the release of interleukin (IL)-1β and IL-18 and cell death by apoptosis and pyroptosis. Proper inflammasome stimulation is crucial for the host to deal with invading pathogens or tissue injury. However, inappropriate inflammasome stimulation can result in unregulated tissue reactions, thus easing many diseases, including melanoma. Hence, keeping a delicate equilibrium between the stimulation and prohibition of inflammasomes is crucial, necessitating meticulous control of the assembly and functional aspects of inflammasomes. This review examines the latest advancements in inflammasome studies, specifically focusing on the molecular processes that control inflammasome formation, signalling, and modulation in melanoma.
Collapse
Affiliation(s)
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Health and Medical Research Center, King Khalid University, P.O. Box 960, AlQura'a, Abha, Saudi Arabia
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, JAIN (Deemed to be University) School of Sciences, Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | | | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| |
Collapse
|
3
|
Liu Z, Xu S, Chen L, Gong J, Wang M. The role of pyroptosis in cancer: key components and therapeutic potential. Cell Commun Signal 2024; 22:548. [PMID: 39548573 PMCID: PMC11566483 DOI: 10.1186/s12964-024-01932-z] [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: 09/02/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Pyroptosis is a lytic and inflammatory form of gasdermin protein-mediated programmed cell death that is typically initiated by inflammasomes. The inflammasome response is an effective mechanism for eradicating germs and cancer cells in the event of cellular injury. The gasdermin family is responsible for initiating pyroptosis, a process in which holes are made in the cell membrane to allow inflammatory chemicals to escape. Mounting evidence indicates that pyroptosis is critical for controlling the development of cancer. In this review, we provide a general overview of pyroptosis, examine the relationship between the primary elements of pyroptosis and tumors, and stress the necessity of pyroptosis-targeted therapy in tumors. Furthermore, we explore its dual nature as a double-edged sword capable of both inhibiting and facilitating the growth of cancer, depending on the specific conditions. Ultimately, pyroptosis is a phenomenon that has both positive and negative effects on tumors. Using this dual impact in a reasonable manner may facilitate investigation into the initiation and progression of tumors and offer insights for the development of novel treatments centered on pyroptosis.
Collapse
Affiliation(s)
- Zixi Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Simiao Xu
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lin Chen
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China.
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China.
| |
Collapse
|
4
|
Mecca M, Picerno S, Cortellino S. The Killer's Web: Interconnection between Inflammation, Epigenetics and Nutrition in Cancer. Int J Mol Sci 2024; 25:2750. [PMID: 38473997 DOI: 10.3390/ijms25052750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Inflammation is a key contributor to both the initiation and progression of tumors, and it can be triggered by genetic instability within tumors, as well as by lifestyle and dietary factors. The inflammatory response plays a critical role in the genetic and epigenetic reprogramming of tumor cells, as well as in the cells that comprise the tumor microenvironment. Cells in the microenvironment acquire a phenotype that promotes immune evasion, progression, and metastasis. We will review the mechanisms and pathways involved in the interaction between tumors, inflammation, and nutrition, the limitations of current therapies, and discuss potential future therapeutic approaches.
Collapse
Affiliation(s)
- Marisabel Mecca
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Simona Picerno
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Salvatore Cortellino
- Laboratory of Preclinical and Translational Research, Responsible Research Hospital, 86100 Campobasso, CB, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, 80138 Naples, NA, Italy
- S.H.R.O. Italia Foundation ETS, 10060 Candiolo, TO, Italy
| |
Collapse
|
5
|
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol 2023; 16:24. [PMID: 36932407 PMCID: PMC10022228 DOI: 10.1186/s13045-023-01407-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 03/19/2023] Open
Abstract
Inflammasomes are macromolecular platforms formed in response to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns, whose formation would cause maturation of interleukin-1 (IL-1) family members and gasdermin D (GSDMD), leading to IL-1 secretion and pyroptosis respectively. Several kinds of inflammasomes detecting different types of dangers have been found. The activation of inflammasomes is regulated at both transcription and posttranscription levels, which is crucial in protecting the host from infections and sterile insults. Present findings have illustrated that inflammasomes are involved in not only infection but also the pathology of tumors implying an important link between inflammation and tumor development. Generally, inflammasomes participate in tumorigenesis, cell death, metastasis, immune evasion, chemotherapy, target therapy, and radiotherapy. Inflammasome components are upregulated in some tumors, and inflammasomes can be activated in cancer cells and other stromal cells by DAMPs, chemotherapy agents, and radiation. In some cases, inflammasomes inhibit tumor progression by initiating GSDMD-mediated pyroptosis in cancer cells and stimulating IL-1 signal-mediated anti-tumor immunity. However, IL-1 signal recruits immunosuppressive cell subsets in other cases. We discuss the conflicting results and propose some possible explanations. Additionally, we also summarize interventions targeting inflammasome pathways in both preclinical and clinical stages. Interventions targeting inflammasomes are promising for immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Ziqi Zhang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xue Li
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
6
|
Aghamajidi A, Farhangnia P, Pashangzadeh S, Damavandi AR, Jafari R. Tumor-promoting myeloid cells in the pathogenesis of human oncoviruses: potential targets for immunotherapy. Cancer Cell Int 2022; 22:327. [PMID: 36303138 PMCID: PMC9608890 DOI: 10.1186/s12935-022-02727-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
Oncoviruses, known as cancer-causing viruses, are typically involved in cancer progression by inhibiting tumor suppressor pathways and uncontrolled cell division. Myeloid cells are the most frequent populations recruited to the tumor microenvironment (TME) and play a critical role in cancer development and metastasis of malignant tumors. Tumor-infiltrating myeloid cells, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumor-associated dendritic cells (TADCs), and tumor-associated neutrophils (TANs) exert different states from anti-tumorigenic to pro-tumorigenic phenotypes in TME. Although their role in the anti-tumorigenic state is well introduced, their opposing roles, pro-tumorigenic activities, such as anti-inflammatory cytokine and reactive oxygen species (ROS) production, should not be ignored since they result in inflammation, tumor progression, angiogenesis, and evasion. Since the blockade of these cells had promising results against cancer progression, their inhibition might be helpful in various cancer immunotherapies. This review highlights the promoting role of tumor-associated myeloid cells (TAMCs) in the pathophysiology of human virus tumorigenesis.
Collapse
Affiliation(s)
- Azin Aghamajidi
- grid.411746.10000 0004 4911 7066Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pooya Farhangnia
- grid.411746.10000 0004 4911 7066Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salar Pashangzadeh
- grid.411705.60000 0001 0166 0922Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmasoud Rayati Damavandi
- grid.411705.60000 0001 0166 0922Students’ Scientific Research Center, Exceptional Talents Development Center, Tehran University of Medical Sciences, Tehran, Iran ,grid.411705.60000 0001 0166 0922School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari
- grid.412763.50000 0004 0442 8645Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
7
|
Di Filippo M, Hennig P, Karakaya T, Slaufova M, Beer HD. NLRP1 in Cutaneous SCCs: An Example of the Complex Roles of Inflammasomes in Cancer Development. Int J Mol Sci 2022; 23:12308. [PMID: 36293159 PMCID: PMC9603439 DOI: 10.3390/ijms232012308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
Protein complexes termed inflammasomes ensure tissue protection from pathogenic and sterile stressors by induction of inflammation. This is mediated by different caspase-1-induced downstream pathways, including activation of the pro-inflammatory cytokines proIL-1β and -18, induction of a lytic type of cell death, and regulation of the release of other pro-inflammatory molecules. Aberrant inflammasome activation underlies the pathology of numerous (auto)inflammatory diseases. Furthermore, inflammasomes support or suppress tumor development in a complex cell-type- and stage-dependent manner. In human keratinocytes and skin, NLRP1 is the central inflammasome sensor activated by cellular perturbation induced, for example, by UVB radiation. UVB represents the main inducer of skin cancer, which is the most common type of malignancy in humans. Recent evidence demonstrates that activation of NLRP1 in human skin supports the development of cutaneous squamous cell carcinomas (cSCCs) by inducing skin inflammation. In contrast, the NLRP1 inflammasome pathway is restrained in established cSCCs, suggesting that, at this stage, the protein complex has a tumor suppressor role. A better understanding of the complex functions of NLRP1 in the development of cSCCs and in general of inflammasomes in cancer might pave the way for novel strategies for cancer prevention and therapy. These strategies might include stage-specific modulation of inflammasome activation or its downstream pathways by mono- or combination therapy.
Collapse
Affiliation(s)
- Michela Di Filippo
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Paulina Hennig
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Tugay Karakaya
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Marta Slaufova
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| |
Collapse
|
8
|
Galvão-Lima LJ, Zambuzi FA, Soares LS, Fontanari C, Meireles AFG, Brauer VS, Faccioli LH, Gama L, Figueiredo LTM, Bou-Habib DC, Frantz FG. HIV-1 Gag and Vpr impair the inflammasome activation and contribute to the establishment of chronic infection in human primary macrophages. Mol Immunol 2022; 148:68-80. [PMID: 35659727 DOI: 10.1016/j.molimm.2022.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023]
Abstract
The successful establishment of HIV-1 infection is related to inflammasome blocking or inactivation, which can result in the viral evasion of the immune responses and formation of reservoirs in several tissues. In this sense, we aimed to evaluate the viral and cellular mechanisms activated during HIV-1 infection in human primary macrophages that allow an effective viral replication in these cells. We found that resting HIV-1-infected macrophages, but not those activated in classical or alternative patterns, released IL-1β and other pro-inflammatory cytokines, and showed increased CXCL10 expression, without changes in the NLRP3, AIM2 or RIG-I inflammasome pathways. Also, similar levels of Casp-1, phosphorylated NF-κB (p65) and NLRP3 proteins were found in uninfected and HIV-1-infected macrophages. Likewise, no alterations were detected in ASC specks released in the culture supernatant after HIV-1 infection, suggesting that macrophages remain viable after infection. Using in silico prediction studies, we found that the HIV-1 proteins Gag and Vpr interact with several host proteins. Comparable levels of trans-LTB4 were found in the supernatants of uninfected and HIV-1-infected macrophages, whereas ROS production was impaired in infected cells, which was not reversed after the PMA stimulus. Immunofluorescence analysis showed structural alterations in the mitochondrial architecture and an increase of BIM in the cytoplasm of infected cells. Our data suggest that HIV-1 proteins Gag and Vpr, through interacting with cellular proteins in the early steps of infection, preclude the inflammasome activation and the development of effective immune responses, thus allowing the establishment of the infection.
Collapse
Affiliation(s)
- Leonardo J Galvão-Lima
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil; Laboratory of Technological Innovation in Health (LAIS), Hospital Universitário Onofre Lopes, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | - Fabiana A Zambuzi
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Luana S Soares
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Caroline Fontanari
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Aline F Galvão Meireles
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Verônica S Brauer
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Lúcia H Faccioli
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Lúcio Gama
- Retrovirus Lab, Johns Hopkins University - School of Medicine, Baltimore, MD, USA; Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Luiz T M Figueiredo
- Virology Research Center, Medical School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| | - Fabiani G Frantz
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
9
|
Fidianingsih I, Aryandono T, Widyarini S, Herwiyanti S. Profile of Histopathological Type and Molecular Subtypes of Mammary Cancer of DMBA-induced Rat and its Relevancy to Human Breast Cancer. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Animal models with mammary cancer that closely mimic human breast cancer for treatment development purposes are still required. Induction of 7,12-dimethylbenzanthracene (DMBA) to rats shows the histopathological features and mammary cancer characterization similar to humans. Examinations of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and Ki67 expressions are crucial in deciding the treatment and prognosis of breast cancer.
AIM: This research aimed to view histopathology images of mammary glands and expressions of ER, PR, Ki67, and HER2 of DMBA-induced rats.
METHODS: After 1-week adaptation, 11 5-weeks-old female rats were induced with 20 mg/kg body weight (BW) of DMBA 2 times a week for 5 weeks. On week 29, nodules taken from the mammary gland were examined for hematoxylin-eosin staining and immunohistochemistry with p63, ER, PR, HER2, and Ki67 antibodies. The grading score used the Nottingham Grading System and molecular classifications based on St. Gallen 2013.
RESULTS: Six rats had nodules, but the histopathologic features of one nodule showed normal mammary gland without cancer. The histopathological type of mammary cancer was cribriform carcinoma, comedo carcinoma, lipid-rich carcinoma, adenocarcinoma squamous, and adenomyepithelioma. Histopathological grading showed 60% of grade 3 and 40% of grade 2. P63 expression showed 60% positive and 40% negative. The frequency of ER, PR, HER2, and Ki67 of five nodules showed positivity: 40%, 60%, 60%, and 60%, respectively. Molecular subtypes of Luminal A, B, HER2, and triple-negative were 0%, 60%, 20%, and 20%, respectively.
CONCLUSION: Histopathological features and molecular subtype of mammary cancer on rats induced with 20 mg/kg BW of DMBA showed similarity to human breast cancer.
Collapse
|
10
|
Mittmann LA, Haring F, Schaubächer JB, Hennel R, Smiljanov B, Zuchtriegel G, Canis M, Gires O, Krombach F, Holdt L, Brandau S, Vogl T, Lauber K, Uhl B, Reichel CA. Uncoupled biological and chronological aging of neutrophils in cancer promotes tumor progression. J Immunother Cancer 2021; 9:jitc-2021-003495. [PMID: 34876407 PMCID: PMC8655594 DOI: 10.1136/jitc-2021-003495] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 01/13/2023] Open
Abstract
Background Beyond their fundamental role in homeostasis and host defense, neutrophilic granulocytes (neutrophils) are increasingly recognized to contribute to the pathogenesis of malignant tumors. Recently, aging of mature neutrophils in the systemic circulation has been identified to be critical for these immune cells to properly unfold their homeostatic and anti-infectious functional properties. The role of neutrophil aging in cancer remains largely obscure. Methods Employing advanced in vivo microscopy techniques in different animal models of cancer as well as utilizing pulse-labeling and cell transfer approaches, various ex vivo/in vitro assays, and human data, we sought to define the functional relevance of neutrophil aging in cancer. Results Here, we show that signals released during early tumor growth accelerate biological aging of circulating neutrophils, hence uncoupling biological from chronological aging of these immune cells. This facilitates the accumulation of highly reactive neutrophils in malignant lesions and endows them with potent protumorigenic functions, thus promoting tumor progression. Counteracting uncoupled biological aging of circulating neutrophils by blocking the chemokine receptor CXCR2 effectively suppressed tumor growth. Conclusions Our data uncover a self-sustaining mechanism of malignant neoplasms in fostering protumorigenic phenotypic and functional changes in circulating neutrophils. Interference with this aberrant process might therefore provide a novel, already pharmacologically targetable strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Laura A Mittmann
- Department of Otorhinolaryngology, LMU München, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Florian Haring
- Department of Otorhinolaryngology, LMU München, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Johanna B Schaubächer
- Department of Otorhinolaryngology, LMU München, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Roman Hennel
- Department of Radiotherapy and Radiation Oncology, LMU München, Munich, Germany
| | - Bojan Smiljanov
- Department of Otorhinolaryngology, LMU München, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Gabriele Zuchtriegel
- Department of Otorhinolaryngology, LMU München, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, LMU München, Munich, Germany
| | - Olivier Gires
- Department of Otorhinolaryngology, LMU München, Munich, Germany
| | - Fritz Krombach
- Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Lesca Holdt
- Institute for Laboratory Medicine, LMU München, Munich, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Thomas Vogl
- Institute for Immunology, University of Munster, Munster, Germany
| | - Kirsten Lauber
- Department of Radiotherapy and Radiation Oncology, LMU München, Munich, Germany
| | - Bernd Uhl
- Department of Otorhinolaryngology, LMU München, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, LMU München, Munich, Germany .,Walter Brendel Centre of Experimental Medicine, LMU München, Munich, Germany
| |
Collapse
|
11
|
Lopez-Labady J, Bologna-Molina R, Villarroel-Dorrego M. Expression of Interleukin-1ß and Interleukin-8 in Oral Potentially Malignant Disorders and Carcinomas. FRONTIERS IN ORAL HEALTH 2021; 2:649406. [PMID: 35048001 PMCID: PMC8757690 DOI: 10.3389/froh.2021.649406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/25/2021] [Indexed: 02/05/2023] Open
Abstract
Objective: To evaluate interleukin-1ß (IL-1ß) and interleukin-8 (IL-8) epithelial expressions in potentially malignant disorders of the oral mucosa as malignant predictive markers. Study design: About 55 tissues embedded in paraffin, comprising 15 oral lichen planus (OLP) lesions, 15 leukoplakias, 15 oral squamous cell carcinomas (OSCC), and 10 samples of normal oral mucosa were included in the study. IL-1ß and 8 expressions were assessed by immunohistochemistry using antibodies antihuman IL-1ß human (sc-7884, Santa Cruz® H-153) and antihuman IL-8 (ab7747, abcam®). The number of positive cells was compared using Student's t-test. Any p-value < 0.05 was considered statistically significant. Results: Nuclear and cytoplasmatic keratinocyte staining were positive for both cytokines in all study groups. However, a statistically significant decrease was observed within all cases compared to normal mucosa, both staining for IL-1β and 8. Moreover, IL-8 showed significant differences between OLP and leukoplakia, and when compared to OSCC. Conclusions: Oral epithelial expression of IL-1β and 8 seems to decrease when the malignant transformation of the oral mucosa increases.
Collapse
Affiliation(s)
| | - Ronell Bologna-Molina
- Molecular Pathology Area, School of Dentistry, University of the Republic, Uruguay University of the Republic (UDELAR), Montevideo, Uruguay
| | | |
Collapse
|
12
|
Amôr NG, Santos PSDS, Campanelli AP. The Tumor Microenvironment in SCC: Mechanisms and Therapeutic Opportunities. Front Cell Dev Biol 2021; 9:636544. [PMID: 33634137 PMCID: PMC7900131 DOI: 10.3389/fcell.2021.636544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Squamous cell carcinoma (SCC) is the second most common skin cancer worldwide and, despite the relatively easy visualization of the tumor in the clinic, a sizeable number of SCC patients are diagnosed at advanced stages with local invasion and distant metastatic lesions. In the last decade, immunotherapy has emerged as the fourth pillar in cancer therapy via the targeting of immune checkpoint molecules such as programmed cell-death protein-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). FDA-approved monoclonal antibodies directed against these immune targets have provide survival benefit in a growing list of cancer types. Currently, there are two immunotherapy drugs available for cutaneous SCC: cemiplimab and pembrolizumab; both monoclonal antibodies (mAb) that block PD-1 thereby promoting T-cell activation and/or function. However, the success rate of these checkpoint inhibitors currently remains around 50%, which means that half of the patients with advanced SCC experience no benefit from this treatment. This review will highlight the mechanisms by which the immune checkpoint molecules regulate the tumor microenvironment (TME), as well as the ongoing clinical trials that are employing single or combinatory therapeutic approaches for SCC immunotherapy. We also discuss the regulation of additional pathways that might promote superior therapeutic efficacy, and consequently provide increased survival for those patients that do not benefit from the current checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Nádia Ghinelli Amôr
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Paulo Sérgio da Silva Santos
- Department of Surgery, Stomatology, Pathology, and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| |
Collapse
|
13
|
Magnotti F, Valsesia S, Gupta P, Flechtenmacher C, Contard P, Viarisio D, Venuti A, Wencker M, Tommasino M, Marvel J, Henry T. The Inflammasome Adaptor ASC Delays UV-Induced Skin Tumorigenesis in Beta HPV38 E6 and E7 Transgenic Mice. J Invest Dermatol 2021; 141:236-238.e2. [PMID: 32470340 DOI: 10.1016/j.jid.2020.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/24/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Flora Magnotti
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Univ Lyon, Lyon, France
| | - Séverine Valsesia
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Univ Lyon, Lyon, France
| | - Purnima Gupta
- Infections and Cancer Biology Group, International Agency for Research on Cancer (IARC), Lyon, France
| | | | - Pierre Contard
- Anira-PBES, SFR Biosciences, UMS3444/CNRS, US8/Inserm, ENS de Lyon, Université Claude Bernard Lyon (UCBL), Univ Lyon, Lyon, France
| | - Daniele Viarisio
- Infection and Cancer Epidemiology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Assunta Venuti
- Infections and Cancer Biology Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Mélanie Wencker
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Univ Lyon, Lyon, France
| | - Massimo Tommasino
- Infections and Cancer Biology Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Jacqueline Marvel
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Univ Lyon, Lyon, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Univ Lyon, Lyon, France.
| |
Collapse
|
14
|
Camacho JP, Obaíd M, Bustos C, Calderón W, Lombardi JJ, Subiabre R, Guler K, Correa F. Squamous Cell Carcinoma as a Result of Likely Industrial Grade Ruptured Poly Implant Prosthèse Silicone Buttock Implants. Aesthet Surg J Open Forum 2020; 2:ojaa030. [PMID: 33791653 PMCID: PMC7671286 DOI: 10.1093/asjof/ojaa030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The Poly Implant Prosthèse (PIP) implants were withdrawn from the market in 2010 due to the use of a nonmedical grade silicone filler. In 2012, the French medical authorities and the International Confederation of Societies of Plastic, Reconstructive and Aesthetic Surgery recommended the extraction of PIP implants. However, during the duration of this scandal, each country in the world did not agree with a uniform procedure, and this rule was not implemented in its entirety. Although laboratory test results on PIP implants were negative for cytotoxicity and genotoxicity, there are many reports in the literature of several complications associated with PIP implants, including high rupture rates and the fact that they are 3 to 5 times more likely to produce local tissue reactions. On the other hand, the development of more strange and worse prognosis complications, such as the development of squamous carcinoma associated with the use of silicone implants (not necessarily related to PIP implants), is less known. To date, only 6 cases have been reported, and all are related to breast augmentation. The authors made the first report of primary gluteal squamous cell cancer related to rupture and delayed removal of PIP silicone buttock implants. Level of Evidence: 5
Collapse
Affiliation(s)
- Juan P Camacho
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Miguel Obaíd
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Camilo Bustos
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Wilfredo Calderón
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Juan J Lombardi
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Rodrigo Subiabre
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Kenneth Guler
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| | - Francisca Correa
- Department of Plastic and Reconstructive Surgery and the Department of Pathology, Hospital del Salvador, Santiago, Chile
| |
Collapse
|
15
|
Böhrnsen F, Holzenburg J, Godek F, Kauffmann P, Moser N, Schliephake H. Influence of tumour necrosis factor alpha on epithelial-mesenchymal transition of oral cancer cells in co-culture with mesenchymal stromal cells. Int J Oral Maxillofac Surg 2019; 49:157-165. [PMID: 31345665 DOI: 10.1016/j.ijom.2019.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/11/2019] [Accepted: 06/05/2019] [Indexed: 01/08/2023]
Abstract
Tumour progression in head and neck squamous cell carcinoma (HNSCC) is influenced by the surrounding stroma and inflammatory cytokines such as tumour necrosis factor alpha (TNF-α). The aim of this study was to test the hypothesis that TNF-α modulates the interactions of HNSCC cell line PCI-13 and bone marrow mesenchymal stromal cells (BMSCs) and influences markers of epithelial-mesenchymal transition (EMT). Following induction with TNF-α, mono- and co-cultures of BMSCs and the established HNSCC cell line PCI-13 were analyzed; protein expression of E-cadherin and vimentin and qRT-PCR expression of Snail, Twist, MMP14, vimentin, E-cadherin, and β-catenin were examined, and changes in cellular AKT signalling were analyzed. TNF-α induced a significant decrease in E-cadherin (64.5±6.0%, P=0.002) and vimentin (10.4±3.5%, P=0.04) protein expression in co-cultured PCI-13, while qRT-PCR showed a significant increase in β-catenin (BMSCs P<0.0001; PCI-13 P=0.0005) and Snail (BMSCs P=0.009; PCI-13 P=0.01). TNF-α also resulted in a down-regulation of AKT downstream targets S6 (38.7±20.9%, P=0.01), p70S6 (16.7±12%, P=0.05), RSK1 (23.6±28.8%, P=0.02), and mTOR (27.4±17.5%, P=0.004) in BMSC co-cultures. In summary, while reducing the expression of vimentin and AKT-signalling in PCI-13 and BMSC, respectively, TNF-α introduced an inflammatory-driven tumour-stroma transition, marked by an increased expression of markers of EMT.
Collapse
Affiliation(s)
- F Böhrnsen
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Göttingen, Germany.
| | - J Holzenburg
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Göttingen, Germany
| | - F Godek
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Göttingen, Germany
| | - P Kauffmann
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Göttingen, Germany
| | - N Moser
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Göttingen, Germany
| | - H Schliephake
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|
16
|
LXRα promotes cell metastasis by regulating the NLRP3 inflammasome in renal cell carcinoma. Cell Death Dis 2019; 10:159. [PMID: 30770793 PMCID: PMC6377709 DOI: 10.1038/s41419-019-1345-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 11/25/2022]
Abstract
Notwithstanding the researches on biomarkers and targeted therapies in renal cell carcinomas (RCC) have made progress in the last decades, the application of the biomarkers and targeted therapy agents for RCC in clinic are restricted because of their limitation or side effects. Liver X receptors (LXRs) and the NLRP3 inflammasome have been the research hotspots in recent years. In our study, we integrated bioinformatics analysis, molecular biology experiments and biological function experiments to study the roles of LXRα and the NLRP3 inflammasome in RCC. The study demonstrated that the elevated LXRα expression is correlated with a poor prognosis in RCC. Furthermore, our study revealed the expression levels and roles of the NLRP3 inflammasome in RCC for the first time. This research demonstrated that LXRα could promote the metastasis of RCC cells by suppressing the expression of the NLRP3 inflammasome. In Brief, LXRα had the possibility to be a novel diagnostic and prognostic biomarker and therapeutic target in renal cell cancer and LXRα could regulate the metastasis of renal cell cancer via NLRP3 inflammamsome.
Collapse
|
17
|
Sand J, Fenini G, Grossi S, Hennig P, Di Filippo M, Levesque M, Werner S, French LE, Beer HD. The NLRP1 Inflammasome Pathway Is Silenced in Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2019; 139:1788-1797.e6. [PMID: 30738816 DOI: 10.1016/j.jid.2019.01.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/18/2018] [Accepted: 01/08/2019] [Indexed: 01/27/2023]
Abstract
The inflammasome protein NLRP1 is an important innate immune sensor in human keratinocytes, and, together with ASC and caspase-1, it mediates the activation and secretion of the proinflammatory cytokines IL-1β and IL-18. These cytokines and inflammasomes can have partly opposing roles during tumorigenesis in mice. In contrast, ASC expression is impaired in different types of cancer in humans. In this study, we analyzed inflammasome activation and expression of inflammasome proteins, including their downstream cytokines, in squamous cell carcinomas, a type of nonmelanoma skin cancer derived from keratinocytes. We assessed mRNA and protein levels in human primary keratinocytes and skin carcinoma-derived SCC cell lines and detected a strong down-regulation of expression of NLRP1 inflammasome components, as well as reduced expression of the proinflammatory cytokines proIL-1β and proIL-1α. Protein levels of NLRP1, ASC, caspase-1, and proIL-1β were reduced in patient-derived SCC biopsy samples compared with healthy skin. Furthermore, the results suggest that expression of PYCARD (ASC), CASP1, IL1B, and NLRP1 is silenced by methylation in SCC cell lines. In conclusion, the down-regulation of the inflammasome pathway in SCCs might favor late tumor development in human skin.
Collapse
Affiliation(s)
- Jennifer Sand
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland
| | - Gabriele Fenini
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland
| | - Serena Grossi
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland
| | - Paulina Hennig
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland
| | - Michela Di Filippo
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland
| | - Mitchell Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland
| | - Sabine Werner
- Institute for Molecular Health Sciences, Department of Biology, ETH Zürich, Zurich, Switzerland
| | - Lars E French
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland; Department of Dermatology and Allergology, Klinikum der Universität München, Munich, Germany
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Switzerland.
| |
Collapse
|
18
|
de Oliveira CE, Gasparoto TH, Pinheiro CR, Amôr NG, Nogueira MRS, Kaneno R, Garlet GP, Lara VS, Silva JS, Cavassani KA, Campanelli AP. CCR5-Dependent Homing of T Regulatory Cells to the Tumor Microenvironment Contributes to Skin Squamous Cell Carcinoma Development. Mol Cancer Ther 2017; 16:2871-2880. [DOI: 10.1158/1535-7163.mct-17-0341] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/14/2017] [Accepted: 09/07/2017] [Indexed: 11/16/2022]
|
19
|
Abstract
Inflammasomes are intracellular multiprotein complexes that comprise part of the
innate immune response. Since their definition, inflammasome disorders have been
linked to an increasing number of diseases. Autoinflammatory diseases refer to
disorders in which local factors lead to the activation of innate immune cells,
causing tissue damage when in the absence of autoantigens and autoantibodies.
Skin symptoms include the main features of monogenic inflammasomopathies, such
as Cryopyrin-Associated Periodic Syndromes (CAPS), Familial Mediterranean Fever
(FMF), Schnitzler Syndrome, Hyper-IgD Syndrome (HIDS), PAPA Syndrome, and
Deficiency of IL-1 Receptor Antagonist (DIRA). Concepts from other pathologies
have also been reviewed in recent years, such as psoriasis, after the
recognition of a combined contribution of innate and adaptive immunity in its
pathogenesis. Inflammasomes are also involved in the response to various
infections, malignancies, such as melanoma, autoimmune diseases, including
vitiligo and lupus erythematosus, atopic and contact dermatitis, acne,
hidradenitis suppurativa, among others. Inhibition of the inflammasome pathway
may be a target for future therapies, as already occurs in the handling of CAPS,
through the introduction of IL-1 inhibitors. This study presents a literature
review focusing on the participation of inflammasomes in skin diseases.
Collapse
Affiliation(s)
| | - Cyro Festa
- Universidade de São Paulo (USP) - São Paulo (SP), Brazil
| |
Collapse
|
20
|
Lin C, Zhang J. Inflammasomes in Inflammation-Induced Cancer. Front Immunol 2017; 8:271. [PMID: 28360909 PMCID: PMC5350111 DOI: 10.3389/fimmu.2017.00271] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/24/2017] [Indexed: 12/19/2022] Open
Abstract
The inflammasome is an important multiprotein complex that functions during inflammatory immune responses. The activation of inflammasome will lead to the autoactivation of caspase-1 and subsequent cleavage of proIL-1β and proIL-18, which are key sources of inflammatory manifestations. Recently, the roles of inflammasomes in cancers have been extensively explored, especially in inflammation-induced cancers. In different and specific contexts, inflammasomes exhibit distinct and even contrasting effects in cancer development. In some cases, inflammasomes initiate carcinogenesis through the extrinsic pathway and maintain the malignant cancer microenvironment through the intrinsic pathway. On the contrary, inflammasomes also exert anticancer effects by specialized programmed cell death called pyroptosis and immune regulatory functions. The phases and compartments in which inflammasomes are activated strongly influence the final immune effects. We systemically summarize the functions of inflammasomes in inflammation-induced cancers, especially in gastrointestinal and skin cancers. Besides, information about the current therapeutic use of inflammasome-related products and potential future developing directions are also introduced.
Collapse
Affiliation(s)
- Chu Lin
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology, National Health and Family Planning Commission of the People's Republic of China, Peking University Health Science Center , Beijing , China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology, National Health and Family Planning Commission of the People's Republic of China, Peking University Health Science Center , Beijing , China
| |
Collapse
|
21
|
Talero E, García-Mauriño S, Ávila-Román J, Rodríguez-Luna A, Alcaide A, Motilva V. Bioactive Compounds Isolated from Microalgae in Chronic Inflammation and Cancer. Mar Drugs 2015; 13:6152-209. [PMID: 26437418 PMCID: PMC4626684 DOI: 10.3390/md13106152] [Citation(s) in RCA: 118] [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: 07/30/2015] [Revised: 09/09/2015] [Accepted: 09/15/2015] [Indexed: 12/12/2022] Open
Abstract
The risk of onset of cancer is influenced by poorly controlled chronic inflammatory processes. Inflammatory diseases related to cancer development include inflammatory bowel disease, which can lead to colon cancer, or actinic keratosis, associated with chronic exposure to ultraviolet light, which can progress to squamous cell carcinoma. Chronic inflammatory states expose these patients to a number of signals with tumorigenic effects, including nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) activation, pro-inflammatory cytokines and prostaglandins release and ROS production. In addition, the participation of inflammasomes, autophagy and sirtuins has been demonstrated in pathological processes such as inflammation and cancer. Chemoprevention consists in the use of drugs, vitamins, or nutritional supplements to reduce the risk of developing or having a recurrence of cancer. Numerous in vitro and animal studies have established the potential colon and skin cancer chemopreventive properties of substances from marine environment, including microalgae species and their products (carotenoids, fatty acids, glycolipids, polysaccharides and proteins). This review summarizes the main mechanisms of actions of these compounds in the chemoprevention of these cancers. These actions include suppression of cell proliferation, induction of apoptosis, stimulation of antimetastatic and antiangiogenic responses and increased antioxidant and anti-inflammatory activity.
Collapse
Affiliation(s)
- Elena Talero
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Sofía García-Mauriño
- Department of Plant Biology and Ecology, Faculty of Biology, University of Seville, Seville 41012, Spain.
| | - Javier Ávila-Román
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Azahara Rodríguez-Luna
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Antonio Alcaide
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Virginia Motilva
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| |
Collapse
|