1
|
Gorvel L, Panouillot M, Rouvière MS, Billon E, Fattori S, Sonongbua J, Boucherit N, Ben Amara A, Quilichini O, Granjeaud S, Degos C, Nunès JA, Carcopino X, Lambaudie E, Chrétien AS, Sabatier R, Dieu-Nosjean MC, Olive D. Tertiary Lymphoid Structures Are Associated with Enhanced Macrophage Activation and Immune Checkpoint Expression and Predict Outcome in Cervical Cancer. Cancer Immunol Res 2025; 13:712-728. [PMID: 39888676 DOI: 10.1158/2326-6066.cir-24-0979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/23/2024] [Accepted: 01/31/2025] [Indexed: 02/02/2025]
Abstract
Cervical tumors are usually treated using surgery, chemotherapy, and radiotherapy and would benefit from immunotherapies. However, the immune microenvironment in cervical cancer remains poorly described. Tertiary lymphoid structures (TLS) were recently described as markers for better immunotherapy response and overall better prognosis in patients with cancer. We evaluated the cervical tumor immune microenvironment, specifically focusing on TLS, using combined high-throughput phenotyping, soluble factor concentration dosage in the tumor microenvironment, and spatial interaction analyses. We found that TLS presence was associated with a more inflammatory soluble microenvironment, with the presence of B cells as well as more activated macrophages and dendritic cells (DC). Furthermore, this myeloid cell activation was associated with the expression of immune checkpoints, such as PD-L1 and CD40, and the proximity of activated conventional type 2 DCs to CD8+ T cells, indicating better immune interactions and tumor control. Finally, we associated TLS presence, greater B-cell density, and activated DC density with improved progression-free survival, substantiating TLS presence as a potential prognostic marker. Our results provide evidence that TLS presence denotes cell activation and immunotherapy target expression.
Collapse
Affiliation(s)
- Laurent Gorvel
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Marylou Panouillot
- UMRS1135 Sorbonne University, INSERM U1135, Centre of Immunology and Microbial Infections (Cimi), Immune Microenvironment and Immunotherapy Team, Paris, France
| | - Marie-Sarah Rouvière
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Emilien Billon
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Stéphane Fattori
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Jumaporn Sonongbua
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Nicolas Boucherit
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Amira Ben Amara
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Olivia Quilichini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Samuel Granjeaud
- CRCM Integrative Bioinformatics Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Clara Degos
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Jacques A Nunès
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Xavier Carcopino
- Department of Obstetrics and Gynecology, Hôpital Nord, APHM, Aix-Marseille University (AMU), CNRS, IRD, IMBE UMR 7263, 13397, Marseille, France
| | - Eric Lambaudie
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Anne-Sophie Chrétien
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Renaud Sabatier
- Predictive Oncology Laboratory, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Marie-Caroline Dieu-Nosjean
- UMRS1135 Sorbonne University, INSERM U1135, Centre of Immunology and Microbial Infections (Cimi), Immune Microenvironment and Immunotherapy Team, Paris, France
| | - Daniel Olive
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| |
Collapse
|
2
|
Gonzalez CE, Vaidya RS, Clayton SW, Tang SY. Secreted chemokines and transcriptomic analyses reveal diverse inflammatory and degenerative processes in the intervertebral disc of the STZ-HFD mouse model of Type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.31.605332. [PMID: 39131361 PMCID: PMC11312574 DOI: 10.1101/2024.07.31.605332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The chronic inflammation resultant from type 2 diabetes (T2D) is also associated with spinal pathologies, including intervertebral disc (IVD) degeneration and chronic neck and back pain. Although confounding factors, such as increased weight gain in obesity, studies have shown that even after adjusting age, body mass index, and genetics (e.g. twins), patients with T2D suffer from disproportionately more IVD degeneration and back pain. We hypothesize that chronic T2D fosters a proinflammatory microenvironment within the IVD that promotes degeneration and disrupts disc homeostasis. To test this hypothesis, we evaluated two commonly used mouse models of T2D - the leptin-receptor deficient mouse (db/db) and the chronic high-fat diet in mice with impaired beta-cell function (STZ-HFD). STZ-HFD IVDs were more degenerated and showed differential expression of chemokines from the db/db models. Moreover, the RNAseq analysis revealed vast transcriptional dysregulation of many pathways in the STZ-HFD but not in the db/db tissues. Leptin signaling may be essential to mediating the inflammation in T2D. Taken together, the STZ-HFD may better recapitulates the complexities of the chronic inflammatory processes in the IVD during T2D.
Collapse
Affiliation(s)
- Christian E. Gonzalez
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
| | - Rachana S. Vaidya
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| | - Sade W. Clayton
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| | - Simon Y. Tang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
- Institute of Material Science and Engineering, Washington University in St. Louis, St. Louis, MO
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
3
|
Yang X, Yang J, Zhou Q, Kang L, Li X, Guo W, Li F, Deng Y. Protocatechuic aldehyde ameliorates psoriasis-like skin inflammation and represses keratinocyte-derived IL-1α and CXCL9 via inhibiting STAT3 activation. Int Immunopharmacol 2025; 147:114037. [PMID: 39793224 DOI: 10.1016/j.intimp.2025.114037] [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: 11/11/2024] [Revised: 12/27/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease. Consistent activation of Signal Transducer and Activator of Transcription 3 (STAT3) in epidermal keratinocyte transactivates various keratinocyte-derived pro-inflammatory cytokines and elicits spontaneous psoriasis-like skin inflammation. In the current study, we first report that topical application of protocatechuic aldehyde (PA), the bioactive compound from Salvia miltiorrhiza (Danshen), significantly improved psoriasis-like skin symptoms and reduced immune cell infiltration in psoriatic lesions. Further molecular mechanism studies demonstrated that PA inactivated STAT3 and inhibited STAT3-mediated transactivation of interleukin-1α (IL-1α) and C-X-C motif chemokine ligand 9 (CXCL9) in epidermal keratinocyte both in vivo and in vitro. Knockdown of STAT3 attenuated the repression effect on IL-1α and CXCL9 by PA. Our results suggested that PA repressed the transactivation of IL-1α and CXCL9 through inhibiting STAT3 in keratinocyte. PA could be potentially used for psoriasis topical treatment or be as a lead compound for drug development.
Collapse
Affiliation(s)
- Xiaoxuan Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106 China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106 China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106 China
| | - Jie Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106 China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106 China; School of Pharmacy, Chengdu University, Chengdu 610106 China
| | - Qian Zhou
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106 China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106 China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106 China
| | - Liang Kang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106 China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106 China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106 China
| | - Xiaoya Li
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106 China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106 China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106 China
| | - Wanjun Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437 China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437 China.
| | - Yu Deng
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106 China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106 China; School of Basic Medical Sciences, Chengdu University, Chengdu 610106 China.
| |
Collapse
|
4
|
Faraji F, Ramirez SI, Clubb LM, Sato K, Burghi V, Hoang TS, Officer A, Anguiano Quiroz PY, Galloway WMG, Mikulski Z, Medetgul-Ernar K, Marangoni P, Jones KB, Cao Y, Molinolo AA, Kim K, Sakaguchi K, Califano JA, Smith Q, Goren A, Klein OD, Tamayo P, Gutkind JS. YAP-driven malignant reprogramming of oral epithelial stem cells at single cell resolution. Nat Commun 2025; 16:498. [PMID: 39779672 PMCID: PMC11711616 DOI: 10.1038/s41467-024-55660-6] [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: 08/24/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Tumor initiation represents the first step in tumorigenesis during which normal progenitor cells undergo cell fate transition to cancer. Capturing this process as it occurs in vivo, however, remains elusive. Here we employ spatiotemporally controlled oncogene activation and tumor suppressor inhibition together with multiomics to unveil the processes underlying oral epithelial progenitor cell reprogramming into tumor initiating cells at single cell resolution. Tumor initiating cells displayed a distinct stem-like state, defined by aberrant proliferative, hypoxic, squamous differentiation, and partial epithelial to mesenchymal invasive gene programs. YAP-mediated tumor initiating cell programs included activation of oncogenic transcriptional networks and mTOR signaling, and recruitment of myeloid cells to the invasive front contributing to tumor infiltration. Tumor initiating cell transcriptional programs are conserved in human head and neck cancer and associated with poor patient survival. These findings illuminate processes underlying cancer initiation at single cell resolution, and identify candidate targets for early cancer detection and prevention.
Collapse
Affiliation(s)
- Farhoud Faraji
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego Health, La Jolla, CA, 92037, USA.
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA.
| | - Sydney I Ramirez
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego Health, La Jolla, CA, 92093, USA
- La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Lauren M Clubb
- University of California San Diego, Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | - Kuniaki Sato
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Valeria Burghi
- Department of Pharmacology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Thomas S Hoang
- University of California San Diego, Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | - Adam Officer
- University of California San Diego, Bioinformatics and Systems Biology Graduate Program, La Jolla, CA, 92093, USA
| | - Paola Y Anguiano Quiroz
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - William M G Galloway
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA, 92697, USA
| | | | - Kate Medetgul-Ernar
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Pauline Marangoni
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kyle B Jones
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Yuwei Cao
- University of California San Diego, Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | - Alfredo A Molinolo
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Joseph A Califano
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego Health, La Jolla, CA, 92037, USA
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Alon Goren
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, 90048, USA
| | - Pablo Tamayo
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, CA, 92037, USA
| | - J Silvio Gutkind
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA.
- Department of Pharmacology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
5
|
Guo Q, Wei X, Qi J, Li C, Xie F. FGFR3 Upregulates Interferon-Stimulated Genes Via the JAK1-STAT1 Signaling Pathway in HPV2 E2 Stable Expressing Keratinocytes. J Med Virol 2025; 97:e70147. [PMID: 39739565 DOI: 10.1002/jmv.70147] [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: 08/24/2024] [Revised: 11/14/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Human papillomavirus (HPV) infections are prevalent skin infectious diseases. While there are no specific anti-HPV drugs available, understanding the viral mechanisms could lead to novel therapeutic strategies. Verruca vulgaris, a common HPV infection, is frequently encountered in dermatological clinics. The HPV E2 protein, an early viral protein, has been implicated in high-risk HPV infections by interacting with fibroblast growth factor receptor 3 (FGFR3) to inhibit viral DNA replication. However, the role of HPV E2 and FGFR3 in low-risk HPV infections remains elusive. Our study takes HPV2, a common subtype of verruca vulgaris, to explore the proliferation and immune regulatory effects of HPV2 E2 on keratinocytes. By overexpressing FGFR3 in HPV2 E2 stable expressing keratinocytes, we assessed changes in interferon-stimulated genes (ISGs) level and cell proliferation. Our findings revealed that HPV2 E2 induced phosphorylation of FGFR3 could activate JAK1-STAT1 pathway, thereby enhancing antiviral immunity through the upregulation of ISGs. Furthermore, we observed co-localization and interaction between FGFR3 and HPV2 E2 in keratinocytes. In conclusion, our study underscores the crucial role of FGFR3 in innate antiviral immunity against HPV2 infection in keratinocytes. These findings may provide a potential therapeutic target for HPV infections.
Collapse
Affiliation(s)
- Qingqing Guo
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Science, Jinan, China
| | - Xuanjin Wei
- Department of Dermatology, Air Force Medical Center, PLA, Beijing, China
| | - Jiayue Qi
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chengxin Li
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fang Xie
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
6
|
Faraji F, Ramirez SI, Clubb L, Sato K, Burghi V, Hoang TS, Officer A, Anguiano Quiroz PY, Galloway WM, Mikulski Z, Medetgul-Ernar K, Marangoni P, Jones KB, Molinolo AA, Kim K, Sakaguchi K, Califano JA, Smith Q, Goren A, Klein OD, Tamayo P, Gutkind JS. YAP-Driven Oral Epithelial Stem Cell Malignant Reprogramming at Single Cell Resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550427. [PMID: 37546810 PMCID: PMC10402053 DOI: 10.1101/2023.07.24.550427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Tumor initiation represents the first step in tumorigenesis during which normal progenitor cells undergo cell fate transition to cancer. Capturing this process as it occurs in vivo, however, remains elusive. Here we employ spatiotemporally controlled oncogene activation and tumor suppressor inhibition together with multiomics to unveil the processes underlying oral epithelial progenitor cell reprogramming into tumor initiating cells (TIC) at single cell resolution. TIC displayed a distinct stem-like state, defined by aberrant proliferative, hypoxic, squamous differentiation, and partial epithelial to mesenchymal (pEMT) invasive gene programs. YAP-mediated TIC programs included the activation of oncogenic transcriptional networks and mTOR signaling, and the recruitment of myeloid cells to the invasive front contributing to tumor infiltration. TIC transcriptional programs are conserved in human head and neck cancer and associated with poor patient survival. These findings illuminate processes underlying cancer initiation at single cell resolution, and identify candidate targets for early cancer detection and prevention.
Collapse
|
7
|
Gutkind JS, Faraji F, Ramirez S, Clubb L, Sato K, Quiroz PA, Galloway W, Mikulski Z, Hoang T, Medetgul-Ernar K, Marangoni P, Jones K, Officer A, Molinolo A, Kim K, Sakaguchi K, Califano J, Smith Q, Klein O, Tamayo P. YAP-Driven Malignant Reprogramming of Epithelial Stem Cells at Single Cell Resolution. RESEARCH SQUARE 2023:rs.3.rs-3426301. [PMID: 37961717 PMCID: PMC10635308 DOI: 10.21203/rs.3.rs-3426301/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Tumor initiation represents the first step in tumorigenesis during which normal progenitor cells undergo cell fate transition to cancer. Capturing this process as it occurs in vivo, however, remains elusive. Here we employ cell tracing approaches with spatiotemporally controlled oncogene activation and tumor suppressor inhibition to unveil the processes underlying oral epithelial progenitor cell reprogramming into cancer stem cells (CSCs) at single cell resolution. This revealed the rapid emergence of a distinct stem-like cell state, defined by aberrant proliferative, hypoxic, squamous differentiation, and partial epithelial to mesenchymal (pEMT) invasive gene programs. Interestingly, CSCs harbor limited cell autonomous invasive capacity, but instead recruit myeloid cells to remodel the basement membrane and ultimately initiate tumor invasion. CSC transcriptional programs are conserved in human carcinomas and associated with poor patient survival. These findings illuminate the process of cancer initiation at single cell resolution, thus identifying candidate targets for early cancer detection and prevention.
Collapse
Affiliation(s)
| | - Farhoud Faraji
- University of California San Diego Health Department of Otolaryngology-Head and Neck Surgery and Moores Cancer Center
| | | | - Lauren Clubb
- University of California San Diego Health Moores Cancer Center
| | - Kuniaki Sato
- University of California San Diego Health Moores Cancer Center
| | | | - William Galloway
- University of California Irvine Department of Chemical and Biomolecular Engineering
| | | | - Thomas Hoang
- University of California San Diego Health Moores Cancer Center
| | | | - Pauline Marangoni
- Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco
| | - Kyle Jones
- University of California San Francisco (UCSF)
| | - Adam Officer
- University of California San Diego Health Moores Cancer Center
| | | | | | | | | | - Quinton Smith
- University of California Irvine Department of Chemical and Biomolecular Engineering
| | | | | |
Collapse
|
8
|
Alfaro R, Llorente S, Gonzalez-Martínez G, Jimenez-Coll V, Martínez-Banaclocha H, Galián JA, Botella C, Moya-Quiles MR, de la Peña-Moral J, Minguela A, Legaz I, Muro M. Clinical Significance of the Pre-Transplant CXCR3 and CCR6 Expression on T Cells In Kidney Graft Recipients. Transplant Proc 2023; 55:66-71. [PMID: 36621349 DOI: 10.1016/j.transproceed.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND T cells play a fundamental role in the processes that mediate graft rejection, tolerance, and defense against infections. The CXCR3 and CCR6 receptors, highly expressed in Th1 (type 1 T helper cells)/Tc1 (T cytotoxic cells, type 1), Th1-Tc1, and Th17-Tc17 lymphocytes, respectively, participate in cell migration toward inflamed tissues. The altered expression level of CXCR3 and CCR6 has been associated with different clinical events after renal transplantation, such as acute rejection (AR) and chronic graft dysfunction, but data are still limited. In this study, we evaluated the expression of the receptor CXCR3 and CCR6 in peripheral blood T lymphocytes from kidney transplant recipients (KTR) and their association with viral infections, AR, and allograft function. METHODS Through flow cytometry, the peripheral blood expression of CXCR3 and CCR6 in T cells was evaluated in a pretransplant collection of KTR. The levels of these T subpopulations and their association with the incidence of AR, kidney graft function, viral infections, cytomegalovirus, and BK virus were studied. Adverse clinical events and graft function were monitored during the first year post transplant. RESULTS KTRs with low pretransplantation levels of Th17 (CD4+CXCR3-CCR6+) (tertile 1, Th17<16.4%) had a higher risk of suffering AR during the first year post transplantation (P = .033). KTRs with viral infections or reactivations during the first 3 months post transplantation had significantly lower levels of Tc17 (CD8+CXCR3-CCR6+) and higher levels of Th1 (CD4+CXCR3+CCR6-). In patients with cytomegalovirus reactivations, the viral peak correlates negatively with the pretransplant levels of Th1 (r = -0.606, P = .037). CONCLUSIONS Pretransplantation assessment of Th1-Th17 and Tc1-Tc17 levels may help predict post-transplant clinical events such as AR and reactivation of viral infections.
Collapse
Affiliation(s)
- Rafael Alfaro
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Santiago Llorente
- Nephrology Services; University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Gema Gonzalez-Martínez
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Víctor Jimenez-Coll
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Helios Martínez-Banaclocha
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - José Antonio Galián
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Carmen Botella
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - María Rosa Moya-Quiles
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Jesús de la Peña-Moral
- Pathology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Alfredo Minguela
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Isabel Legaz
- Department of Legal and Forensic Medicine, Biomedical Research Institute (IMIB), Regional Campus of International Excellence "Campus Mare Nostrum", Faculty of Medicine, University of Murcia, Murcia, Spain.
| | - Manuel Muro
- Immunology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain.
| |
Collapse
|
9
|
Valdés N, Cortés M, Barraza F, Reyes-López FE, Imarai M. CXCL9-11 chemokines and CXCR3 receptor in teleost fish species. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2022; 3:100068. [PMID: 36569039 PMCID: PMC9782732 DOI: 10.1016/j.fsirep.2022.100068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 12/27/2022] Open
Abstract
The coordinated migration of immune cells from lymphoid organs to in or out of the bloodstream, and towards the site of infection or tissue damage is fundamental for an efficient innate and adaptive immune response. Interestingly, an essential part of this movement is mediated by chemoattractant cytokines called chemokines. Although the nature and function of chemokines and their receptors are well documented in mammals, much research is needed to accomplish a similar level of understanding of the role of chemokines in fish immunity. The first chemokine gene identified in teleosts (rainbow trout, Oncorhynchus mykiss) was CK1 in 1998. Since then, the identification of fish chemokine orthologue genes and characterization of their role has been more complex than expected, primarily because of the whole genome duplication processes occurring in fish, and because chemokines evolve faster than other immune genes. Some of the most studied chemokines are CXCL9, CXCL10, CXCL11, and the CXCR3 receptor, all involved in T cell migration and in the induction of the T helper 1 (Th1) immune response. Data from the zebrafish and rainbow trout CXCL9-11/CXCR3 axis suggest that these chemokines and the receptor arose early in evolution and must be present in most teleost fish. However, the pieces of knowledge also indicate that different numbers of gene copies can be present in different species, with distinct regulatory expression mechanisms and probably, also with different roles, as the differential expression in fish tissues suggest. Here, we revised the current knowledge of the CXCL9-11/CXCR3 axis in teleost fishes, identifying the gaps in knowledge, and raising some hypotheses for the role of CXCL9, CXCL10 CXCL11, and CXCR3 receptor axis in fish, which can encourage further studies in the field.
Collapse
Affiliation(s)
- Natalia Valdés
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología. Universidad de Santiago de Chile, Chile,Corresponding author.
| | - Marcos Cortés
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología. Universidad de Santiago de Chile, Chile
| | - Felipe Barraza
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología. Universidad de Santiago de Chile, Chile
| | - Felipe E. Reyes-López
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología. Universidad de Santiago de Chile, Chile,Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain,Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago, Chile
| | - Mónica Imarai
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología. Universidad de Santiago de Chile, Chile
| |
Collapse
|
10
|
Deng ZL, Zhou DZ, Cao SJ, Li Q, Zhang JF, Xie H. Development and Validation of an Inflammatory Response-Related Gene Signature for Predicting the Prognosis of Pancreatic Adenocarcinoma. Inflammation 2022; 45:1732-1751. [PMID: 35322324 DOI: 10.1007/s10753-022-01657-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 11/05/2022]
Abstract
Pancreatic adenocarcinoma (PAAD) is a highly dangerous malignant tumor of the digestive tract, and difficult to diagnose, treat, and predict the prognosis. As we all know, tumor and inflammation can affect each other, and thus the inflammatory response in the microenvironment can be used to affect the prognosis. So far, the prognostic value of inflammatory response-related genes in PAAD is still unclear. Therefore, this study aimed to explore the inflammatory response-related genes for predicting the prognosis of PAAD. In this study, the mRNA expression profiles of PAAD patients and the corresponding clinical characteristics data of PAAD patients were downloaded from the public database. The least absolute shrinkage and selection operator (LASSO) Cox analysis model was used to identify and construct the prognostic gene signature in The Cancer Genome Atlas (TCGA) cohort. The PAAD patients used for verification are from the International Cancer Genome Consortium (ICGC) cohort. The Kaplan-Meier method was used to compare the overall survival (OS) between the high- and low-risk groups. Univariate and multivariate Cox analyses were performed to identify the independent predictors of OS. Gene set enrichment analysis (GSEA) was performed to obtain gene ontology (GO) terms and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and the correlation between gene expression and immune infiltrates was investigated via single sample gene set enrichment analysis (ssGSEA). The GEPIA database was performed to examine prognostic genes in PAAD. LASSO Cox regression analysis was used to construct a model of inflammatory response-related gene signature. Compared with the low-risk group, patients in the high-risk group had significantly lower OS. The receiver operating characteristic curve (ROC) analysis confirmed the signature's predictive capacity. Multivariate Cox analysis showed that risk score is an independent predictor of OS. Functional analysis shows that the immune status between the two risk groups is significantly different, and the cancer-related pathways were abundant in the high-risk group. Moreover, the risk score is significantly related to tumor grade, stage, and immune infiltration types. It was also obtained that the expression level of prognostic genes was significantly correlated with the sensitivity of cancer cells to anti-tumor drugs. In addition, there are significant differences in the expression of PAAD tissues and adjacent non-tumor tissues. The novel signature constructed from five inflammatory response-related genes can be used to predict prognosis and affect the immune status of PAAD. In addition, suppressing these genes may be a treatment option.
Collapse
Affiliation(s)
- Zu-Liang Deng
- Department of Radiation Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, People's Republic of China
| | - Ding-Zhong Zhou
- Department of Interventional Vascular Surgery, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, People's Republic of China
| | - Su-Juan Cao
- Department of Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, People's Republic of China
| | - Qing Li
- Department of Interventional Vascular Surgery, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, People's Republic of China
| | - Jian-Fang Zhang
- Department of Physical Examination, Beihu Centers for Disease Control and Prevention, Chenzhou, 423000, People's Republic of China
| | - Hui Xie
- Department of Radiation Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| |
Collapse
|
11
|
Saluzzo S, Pandey RV, Gail LM, Dingelmaier-Hovorka R, Kleissl L, Shaw L, Reininger B, Atzmüller D, Strobl J, Touzeau-Römer V, Beer A, Staud C, Rieger A, Farlik M, Weninger W, Stingl G, Stary G. Delayed antiretroviral therapy in HIV-infected individuals leads to irreversible depletion of skin- and mucosa-resident memory T cells. Immunity 2021; 54:2842-2858.e5. [PMID: 34813775 DOI: 10.1016/j.immuni.2021.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/20/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023]
Abstract
People living with HIV (PLWH) are at increased risk for developing skin and mucosal malignancies despite systemic reconstitution of CD4+ T cells upon antiretroviral therapy (ART). The underlying mechanism of chronic tissue-related immunodeficiency in HIV is unclear. We found that skin CD4+ tissue-resident memory T (Trm) cells were depleted after HIV infection and replenished only upon early ART initiation. TCR clonal analysis following early ART suggested a systemic origin for reconstituting CD4+ Trm cells. Single-cell RNA sequencing in PLWH that received late ART treatment revealed a loss of CXCR3+ Trm cells and a tolerogenic skin immune environment. Human papilloma virus-induced precancerous lesion biopsies showed reduced CXCR3+ Trm cell frequencies in the mucosa in PLWH versus HIV- individuals. These results reveal an irreversible loss of CXCR3+ Trm cells confined to skin and mucosa in PLWH who received late ART treatment, which may be a precipitating factor in the development of HPV-related cancer.
Collapse
Affiliation(s)
- Simona Saluzzo
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria.
| | - Ram Vinay Pandey
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Laura Marie Gail
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | | | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
| | - Lisa Shaw
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Bärbel Reininger
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Denise Atzmüller
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | | | - Andrea Beer
- Department of Pathology, Medical University of Vienna, Vienna 1090, Austria
| | - Clement Staud
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna 1090, Austria
| | - Armin Rieger
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria.
| |
Collapse
|
12
|
Tuong ZK, Lukowski SW, Nguyen QH, Chandra J, Zhou C, Gillinder K, Bashaw AA, Ferdinand JR, Stewart BJ, Teoh SM, Hanson SJ, Devitt K, Clatworthy MR, Powell JE, Frazer IH. A model of impaired Langerhans cell maturation associated with HPV induced epithelial hyperplasia. iScience 2021; 24:103326. [PMID: 34805788 PMCID: PMC8586807 DOI: 10.1016/j.isci.2021.103326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Langerhans cells (LC) are skin-resident antigen-presenting cells that regulate immune responses to epithelial microorganisms. Human papillomavirus (HPV) infection can promote malignant epithelial transformation. As LCs are considered important for controlling HPV infection, we compared the transcriptome of murine LCs from skin transformed by K14E7 oncoprotein and from healthy skin. We identified transcriptome heterogeneity at the single cell level amongst LCs in normal skin, associated with ontogeny, cell cycle, and maturation. We identified a balanced co-existence of immune-stimulatory and immune-inhibitory LC cell states in normal skin that was significantly disturbed in HPV16 E7-transformed skin. Hyperplastic skin was depleted of immune-stimulatory LCs and enriched for LCs with an immune-inhibitory gene signature, and LC-keratinocyte crosstalk was dysregulated. We identified reduced expression of interleukin (IL)-34, a critical molecule for LC homeostasis. Enrichment of an immune-inhibitory LC gene signature and reduced levels of epithelial IL-34 were also found in human HPV-associated cervical epithelial cancers. Single cell atlas of Langerhans cells in cutaneous skin Stimulatory and inhibitory Langerhans cell states are in balance Inhibitory Langerhans cell states dominate HPV-transformed hyperplastic skin Langerhans cell imbalance is associated with disrupted IL-34 signaling
Collapse
Affiliation(s)
- Zewen K Tuong
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia.,Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Samuel W Lukowski
- Australia Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Quan H Nguyen
- Australia Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Janin Chandra
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Chenhao Zhou
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Kevin Gillinder
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Abate A Bashaw
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - John R Ferdinand
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Siok Min Teoh
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Sarah J Hanson
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Katharina Devitt
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Menna R Clatworthy
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK.,Wellcome Trust Sanger Institute, Hinxton, UK
| | - Joseph E Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
13
|
Lin F, Hu W, Xu W, Zhou M, Xu AE. CXCL9 as a key biomarker of vitiligo activity and prediction of the success of cultured melanocyte transplantation. Sci Rep 2021; 11:18298. [PMID: 34521889 PMCID: PMC8440594 DOI: 10.1038/s41598-021-97296-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/11/2021] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate the potential biomarkers of vitiligo by evaluating the disease activity and curative effect of autologous cultured pure melanocyte transplantation (CMT) on patients. Altogether, 36patients with stable vitiligo were treated with CMT. Blister fluid samples were collected from patients with stable vitiligo. Patients with active vitiligo were matched with healthy controls. The chemokine levels in the serum and blister fluid samples were measured using Luminex. The curative effect on patients with stable vitiligo was evaluated 6 months after treatment. Treatment responses were defined according to the extent of repigmentation as effective (if 50% or more repigmentation was achieved) or ineffective (if less than 50% or worse repigmentation was achieved). Patients received re-transplantation if the initial treatment was ineffective. The levels of C-X-C motif chemokine ligand (CXCL)9 and CXCL10 in blister fluid samples were significantly lower in stable patients than in active participants. Receiver operating characteristic analysis revealed that the levels of CXCL9 and CXCL10 were sensitive and specific in diagnosing active vitiligo. Further, 65.6% (21/32) of patients who received CMT had effective treatment responses. The high CXCL9 level in the blister fluid was a significant predictor of ineffective treatment responses. The treatment response was significantly enhanced after treatment. Four patients with ineffective treatment responses received anti-inflammatory treatment and re-transplantation. The CXCL9 and CXCL10 levels in the blister fluid were related to the presence of active vitiligo. Also, the CXCL9 level was a predictor of the effectiveness of CMT in treating vitiligo.
Collapse
Affiliation(s)
- Fuquan Lin
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenting Hu
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Xu
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Miaoni Zhou
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Ai-E Xu
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
14
|
Chen X, He H, Xiao Y, Hasim A, Yuan J, Ye M, Li X, Hao Y, Guo X. CXCL10 Produced by HPV-Positive Cervical Cancer Cells Stimulates Exosomal PDL1 Expression by Fibroblasts via CXCR3 and JAK-STAT Pathways. Front Oncol 2021; 11:629350. [PMID: 34422627 PMCID: PMC8377428 DOI: 10.3389/fonc.2021.629350] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 05/26/2021] [Indexed: 11/13/2022] Open
Abstract
Persistent infection with human papillomavirus (HPV) and immune surveillance failure may be the initiating factors for the carcinogenesis of cervical squamous cell carcinoma (CSCC). HPV infection might affect the innate immune pathway of cervical epithelial cells that constitute the "microenvironment" for tumor cells. Programmed death-ligand 1 (PD-L1) has been reported to be an immunosuppressor that helps cancer cells escape the actions of T cells. In the present study, CXCL10 was substantially upregulated both in cervical tissues of HPV infected patients with cervical intraepithelial neoplasia (CIN) or CSCC, as well as in HPV16 E6/E7 transgenic murine cervix. The HPV-positive (HPV+) cervical cancer cell lines SiHa and Caski secreted increased levels of CXCL10 compared to human foreskin fibroblasts (HFF-1), and its receptor CXCR3 was overexpressed in HFF-1. After co-culture with SiHa or Caski, the JAK-STAT signaling pathway and exosomal PD-L1 expression were both upregulated in HFF-1. Recombinant human CXCL10 induced JAK-STAT and PD-L1, while the CXCL10-CXCR3 and JAK-STAT inhibitors AMG487 or ruxolitinib reduced the expression of PD-L1 in HFF-1 cells. Furthermore, the upregulated expression of PD-L1 was verified in HPV+ but not HPV-negative (HPV-) patients with cervical cancers by analysis of tissue microarray cores in 25 cervical lesion patients (P < 0.05). The results indicate that HPV infection can induce cervical cancer cells to secrete CXCL10, which binds to CXCR3 in the surrounding fibroblast cells,leading to JAK-STAT pathway activation and the subsequent upregulated expression of exosomal PD-L1. These mechanisms may help HPV to escape immune response attack, leading to carcinogenesis.
Collapse
Affiliation(s)
- Xiaona Chen
- Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Shenzhen, China
- Clinical Medical Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Hui He
- Department of Pathology, Shenzhen Hospital, The University of Hong Kong, Shenzhen, China
| | - Yue Xiao
- Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Shenzhen, China
- Clinical Medical Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Ayshamgul Hasim
- Department of Pathology, Basic College, Xinjiang Medical University, Urumqi, China
| | - Jianlin Yuan
- Department of Gynecology, Affiliated Cancer Hospital, Xinjiang Medical University, Urumqi, China
| | - Min Ye
- Department of Pathology, Affiliated Cancer Hospital, Xinjiang Medical University, Urumqi, China
| | - Xin Li
- Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Shenzhen, China
- Clinical Medical Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yi Hao
- Department of Ultrasound, South China Hospital of Shenzhen University, Shenzhen, China
| | - Xia Guo
- Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Shenzhen, China
- Clinical Medical Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
15
|
Lin Y, Lu R, Hou J, Zhou GG, Fu W. IFNgamma-inducible CXCL10/CXCR3 axis alters the sensitivity of HEp-2 cells to ionizing radiation. Exp Cell Res 2020; 398:112382. [PMID: 33253709 DOI: 10.1016/j.yexcr.2020.112382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 01/06/2023]
Abstract
Radiotherapy is a conventional approach for anti-cancer treatment, killing tumor cells through damaging cellular DNA. While increasing studies have demonstrated that tumors generated the tolerance to radiation and tumor immune system was found to be correlated to radiotherapy resistance. Therefore, it is critical to identify potential immune factors associated with the efficacy of radiotherapy. Here in this study, we evaluated the sensitivities of different tumor cells to radiation and determined HEp-2 cells as the radio-resistant tumor cells for further investigation. IFNgamma as a key regulator of host immune response showed the potential to sensitize tumors to ionizing radiation (IR). Besides, IFNgamma-induced CXC chemokine ligand 10 (CXCL10) was found to be necessary for effective IR-induced killing of cultured HEp-2 cells. Increased clonogenic survival was observed in CXCL10-depleted HEp-2 cells and CXCL10-KO cells. Additionally, the loss of CXCL10 in HEp-2 cells showed less progression of the G0/G1 phase to G2/M when exposed to IR (8 Gy). Local IR (20 Gy) to nude mice bearing HEp-2 tumors significantly reduced tumor burden, while fewer effects on tumor burden in mice carrying CXCL10-KO tumors were observed. We furtherly evaluated the possible roles the chemokine receptor CXCR3 plays in mediating the sensitivity of cultured HEp-2 cells to IR. Altered expression of CXCR3 in HEp-2 cells affected IR-induced killing of HEp-2 cells. Our data suggest the IFNgamma-activated CXCL10/CXCR3 axis may contribute to the effective radiation-induced killing of HEp-2 cells in vitro.
Collapse
Affiliation(s)
- Yunting Lin
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Ruitao Lu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Jingmei Hou
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Grace Guoying Zhou
- Shenzhen International Institute for Biomedical Research, 1301 Guanguang Rd. 3F Building 1-B, Silver Star Hi-tech Park Longhua District, Shenzhen, Guangdong, 518116, China.
| | - Wenmin Fu
- Shenzhen International Institute for Biomedical Research, 1301 Guanguang Rd. 3F Building 1-B, Silver Star Hi-tech Park Longhua District, Shenzhen, Guangdong, 518116, China.
| |
Collapse
|
16
|
Wang W, Uberoi A, Spurgeon M, Gronski E, Majerciak V, Lobanov A, Hayes M, Loke A, Zheng ZM, Lambert PF. Stress keratin 17 enhances papillomavirus infection-induced disease by downregulating T cell recruitment. PLoS Pathog 2020; 16:e1008206. [PMID: 31968015 PMCID: PMC6975545 DOI: 10.1371/journal.ppat.1008206] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022] Open
Abstract
High-risk human papillomaviruses (HPVs) cause 5% of human cancers. Despite the availability of HPV vaccines, there remains a strong urgency to find ways to treat persistent HPV infections, as current HPV vaccines are not therapeutic for individuals already infected. We used a mouse papillomavirus infection model to characterize virus-host interactions. We found that mouse papillomavirus (MmuPV1) suppresses host immune responses via overexpression of stress keratins. In mice deficient for stress keratin K17 (K17KO), we observed rapid regression of papillomas dependent on T cells. Cellular genes involved in immune response were differentially expressed in the papillomas arising on the K17KO mice correlating with increased numbers of infiltrating CD8+ T cells and upregulation of IFNγ-related genes, including CXCL9 and CXCL10, prior to complete regression. Blocking the receptor for CXCL9/CXCL10 prevented early regression. Our data provide a novel mechanism by which papillomavirus-infected cells evade host immunity and defines new therapeutic targets for treating persistent papillomavirus infections.
Collapse
Affiliation(s)
- Wei Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Aayushi Uberoi
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Megan Spurgeon
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Ellery Gronski
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Vladimir Majerciak
- Tumor Virus RNA Biology Section, National Cancer Institute, Frederick, MD, United States of America
| | - Alexei Lobanov
- CCR Collaborative Bioinformatics Resource (CCBR), National Cancer Institute, Bethesda, MD, United States of America
| | - Mitchell Hayes
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Amanda Loke
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, National Cancer Institute, Frederick, MD, United States of America
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, United States of America
- * E-mail:
| |
Collapse
|
17
|
Cytokine/chemokine profiles in squamous cell carcinoma correlate with precancerous and cancerous disease stage. Sci Rep 2019; 9:17754. [PMID: 31780824 PMCID: PMC6882799 DOI: 10.1038/s41598-019-54435-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 11/14/2019] [Indexed: 11/20/2022] Open
Abstract
Actinic Keratosis (AK), Intraepidermal Carcinoma (IEC), and Squamous Cell Carcinoma (SCC) are generally considered to be advancing stages of the same disease spectrum. However, while AK often regress spontaneously, and IEC often regress in response to immune-activating treatments, SCC typically do not regress. Therefore, it is vital to define whether fundamental immunological changes occur during progression to SCC. Here we show that proinflammatory cytokine expression, chemokine expression, and immune cell infiltration density change during progression to SCC. Our findings suggest a switch from predominantly proinflammatory cytokine production to chemokine production is a key feature of progression from precancer to cancer. Together, these observations propose a model that can underpin current research and open new avenues of exploration into the clinical significance of these profiles with respect to immunotherapeutic or other treatment outcomes.
Collapse
|
18
|
Zhou C, Tuong ZK, Frazer IH. Papillomavirus Immune Evasion Strategies Target the Infected Cell and the Local Immune System. Front Oncol 2019; 9:682. [PMID: 31428574 PMCID: PMC6688195 DOI: 10.3389/fonc.2019.00682] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
Persistent infection with human papillomavirus (HPV) initiates ~5% of all human cancers, and particularly cervical and oropharyngeal cancers. HPV vaccines prevent HPV infection, but do not eliminate existing HPV infections. Papillomaviruses induce hyperproliferation of epithelial cells. In this review we discuss how hyperproliferation renders epithelial cells less sensitive to immune attack, and impacts upon the efficiency of the local immune system. These observations have significance for the design of therapeutic HPV cancer immunotherapies.
Collapse
Affiliation(s)
- Chenhao Zhou
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Zewen Kelvin Tuong
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia.,Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ian Hector Frazer
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
19
|
Frazer IH, Chandra J. Immunotherapy for HPV associated cancer. PAPILLOMAVIRUS RESEARCH 2019; 8:100176. [PMID: 31310819 PMCID: PMC6639647 DOI: 10.1016/j.pvr.2019.100176] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 02/01/2023]
Affiliation(s)
- Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, 4102, Australia.
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
20
|
Bashaw AA, Teoh SM, Tuong ZK, Leggatt GR, Frazer IH, Chandra J. HPV16 E7-Driven Epithelial Hyperplasia Promotes Impaired Antigen Presentation and Regulatory T-Cell Development. J Invest Dermatol 2019; 139:2467-2476.e3. [PMID: 31207230 DOI: 10.1016/j.jid.2019.03.1162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/15/2019] [Accepted: 03/31/2019] [Indexed: 01/21/2023]
Abstract
Human papillomaviruses infect keratinocytes and can lead to hyperproliferative dysplasia and malignant transformation if not cleared by the immune system. Human papillomavirus has evolved an array of mechanisms to evade and manipulate the immune system to improve replication efficiency and promote persistent infection. We here demonstrate that hyperproliferative skin expressing the high-risk human papillomavirus 16 E7 oncogene as a transgene drives immunomodulation of dendritic cells (DCs), resulting in reduced capacity to take up antigen and prime effector CD4+ T cell responses. The phenotype of DCs in the E7-expressing hyperproliferative skin was not reversible by activation through intradermal immunization. Naïve CD4+ T cells primed by E7-driven hyperproliferative skin acquired FoxP3 expression and an anergic phenotype. DC and T help modulation was dependent on E7-retinoblastoma protein interaction-driven epithelial hyperproliferation, rather than on expression of E7. Inhibition of binding of E7 to retinoblastoma protein, and of consequent epithelial hyperplasia, was associated with normal skin DC phenotype, and T helper type 1 effector responses to immunization were restored. We conclude that human papillomavirus-induced epithelial hyperplasia modulates epithelial DCs and inhibits T helper type 1 immunity while polarizing T-cell differentiation to a regulatory or anergic phenotype.
Collapse
Affiliation(s)
- Abate Assefa Bashaw
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Siok M Teoh
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia.
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
21
|
Luo H, Hu L, Ma B, Zhao M, Luo M, Deng Q, Deng S, Ye H, Lin T, Chen J, Wang T, Zhu J, Lu H. Molecular dynamics based improvement of the solubilizing self-cleavable tag Z basic-ΔI-CM application in the preparation of recombinant proteins in Escherichia coli. Biochem Biophys Res Commun 2019; 513:412-418. [PMID: 30967267 DOI: 10.1016/j.bbrc.2019.03.205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 10/27/2022]
Abstract
Zbasic-ΔI-CM is a novel intein-based self-cleavable tag we developed to accelerate the soluble expression of recombinant proteins in Escherichia coli (E. coli). Previously we found that intein activity could be interfered by its flanking exteins, and thus reducing the production efficiency and final yield. In this work, we used CXC-chemokine 9 (CXCL9) as a model C-extein, which fusion with Zbasic-ΔI-CM showed high intein activity. When the fusion protein got soluble expression, CXCL9 was released immediately and purified directly from cell lysis supernatant. The results demonstrated that Zbasic-ΔI-CM tag had successfully mediated the efficient production of high-quality CXCL9 with reduced time and resources consumption in comparison with inclusion bodies expression. Molecular dynamics simulations suggested that the improved cleavage activity of Zbasic-ΔI-CM upon fusion with CXCL9 may be due to the higher dynamics of the first half loop and stabilization of the second half loop of intein. Our results proved that the self-cleavable Zbasic-ΔI-CM mediated soluble expression could be a feasible process for cytokines like CXCL9, thus of attractive potentials for production of therapeutic proteins using E. coli expression system.
Collapse
Affiliation(s)
- Han Luo
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Lifu Hu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Research Institute at Frederick, Frederick, MD, 21702, USA
| | - Meiqi Zhao
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Manyu Luo
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qing Deng
- Regeneromics Laboratory, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Shaorong Deng
- Regeneromics Laboratory, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Ye
- Regeneromics Laboratory, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Tong Lin
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Junsheng Chen
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Tao Wang
- Jecho Biopharmaceuticals Co. Ltd., 2633 Zhongbin Road, Sino-Singapore Tianjin Eco-City, Tianjin, 300467, China
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China; Jecho Biopharmaceuticals Co. Ltd., 2633 Zhongbin Road, Sino-Singapore Tianjin Eco-City, Tianjin, 300467, China; Jecho Laboratories, Inc., 7320 Executive Way, Frederick, MD, 21704, USA.
| | - Huili Lu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
22
|
Kuo P, Teoh SM, Tuong ZK, Leggatt GR, Mattarollo SR, Frazer IH. Recruitment of Antigen Presenting Cells to Skin Draining Lymph Node From HPV16E7-Expressing Skin Requires E7-Rb Interaction. Front Immunol 2018; 9:2896. [PMID: 30619266 PMCID: PMC6305623 DOI: 10.3389/fimmu.2018.02896] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/26/2018] [Indexed: 12/27/2022] Open
Abstract
“High-risk” human papillomaviruses (HPV) infect keratinocytes of squamous epithelia. The HPV16E7 protein induces epithelial hyperplasia by binding Rb family proteins and disrupting cell cycle termination. Murine skin expressing HPV16E7 as a transgene from a keratin 14 promoter (K14.E7) demonstrates epithelial hyperplasia, dysfunctional antigen presenting cells, ineffective antigen presentation by keratinocytes, and production of immunoregulatory cytokines. Furthermore, grafted K14.E7 skin is not rejected from immunocompetent non-transgenic recipient animals. To establish the contributions of E7, of E7-Rb interaction and of epithelial hyperplasia to altered local skin immunity, K14.E7 skin was compared with skin from K14.E7 mice heterozygous for a mutant Rb unable to bind E7 (K14.E7xRbΔL/ΔL mice), that have normoplastic epithelium. Previously, we demonstrated that E7-speicfic T cells do not accumulate in K14.E7xRbΔL/ΔL skin grafts. Here, we further show that K14.E7xRbΔL/ΔL skin, like K14.E7 skin, is not rejected by immunocompetent non-transgenic animals. There were fewer CD11b+ antigen presenting cells in skin draining lymph nodes from animals recipient of K14.E7xRbΔL/ΔL grafts, when compared with animals receiving K14.E7 grafts or K5mOVA grafts. Maturation of migratory DCs derived from K14.E7xRbΔL/ΔL grafts found in the draining lymph nodes is significantly lower than that of K14.E7 grafts. Surprisingly, K14.E7xRbΔL/ΔL keratinocytes, unlike K14.E7 keratinocytes, are susceptible to E7 directed CTL-mediated lysis in vitro. We conclude that E7-Rb interaction and its associated epithelial hyperplasia partially contribute to the suppressive local immune responses in area affected by HPV16E7 expression.
Collapse
Affiliation(s)
- Paula Kuo
- Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Siok Min Teoh
- Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Zewen K Tuong
- Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Graham R Leggatt
- Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Stephen R Mattarollo
- Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Ian H Frazer
- Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| |
Collapse
|
23
|
Kuo PT, Zeng Z, Salim N, Mattarollo S, Wells JW, Leggatt GR. The Role of CXCR3 and Its Chemokine Ligands in Skin Disease and Cancer. Front Med (Lausanne) 2018; 5:271. [PMID: 30320116 PMCID: PMC6167486 DOI: 10.3389/fmed.2018.00271] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Chemokines and their receptors play an important role in the recruitment, activation and differentiation of immune cells. The chemokine receptor, CXCR3, and its ligands, CXCL9, CXCL10, and CXCL11 are key immune chemoattractants during interferon-induced inflammatory responses. Inflammation of the skin resulting from infections or autoimmune disease drives expression of CXCL9/10/11 and the subsequent recruitment of effector, CXCR3+ T cells from the circulation. The relative contributions of the different CXCR3 chemokines and the three variant isoforms of CXCR3 (CXCR3A, CXCR3B, CXCR3alt) to the inflammatory process in human skin requires further investigation. In skin cancers, the CXCR3 receptor can play a dual role whereby expression on tumor cells can lead to cancer metastasis to systemic sites while receptor expression on immune cells can frequently promote anti-tumor immune responses. This review will discuss the biology of CXCR3 and its associated ligands with particular emphasis on the skin during inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Paula T Kuo
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Zhen Zeng
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Nazhifah Salim
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Stephen Mattarollo
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - James W Wells
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Graham R Leggatt
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|