1
|
Warrick KA, Vallez CN, Meibers HE, Pasare C. Bidirectional Communication Between the Innate and Adaptive Immune Systems. Annu Rev Immunol 2025; 43:489-514. [PMID: 40279312 DOI: 10.1146/annurev-immunol-083122-040624] [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] [Indexed: 04/27/2025]
Abstract
Effective bidirectional communication between the innate and adaptive immune systems is crucial for tissue homeostasis and protective immunity against infections. The innate immune system is responsible for the early sensing of and initial response to threats, including microbial ligands, toxins, and tissue damage. Pathogen-related information, detected primarily by the innate immune system via dendritic cells, is relayed to adaptive immune cells, leading to the priming and differentiation of naive T cells into effector and memory lineages. Memory T cells that persist long after pathogen clearance are integral for durable protective immunity. In addition to rapidly responding to reinfections, memory T cells also directly instruct the interacting myeloid cells to induce innate inflammation, which resembles microbial inflammation. As such, memory T cells act as newly emerging activators of the innate immune system and function independently of direct microbial recognition. While T cell-mediated activation of the innate immune system likely evolved as a protective mechanism to combat reinfections by virulent pathogens, the detrimental outcomes of this mechanism manifest in the forms of autoimmunity and other T cell-driven pathologies. Here, we review the complexities and layers of regulation at the interface between the innate and adaptive immune systems to highlight the implications of adaptive instruction of innate immunity in health and disease.
Collapse
Affiliation(s)
- Kathrynne A Warrick
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Charles N Vallez
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Hannah E Meibers
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| |
Collapse
|
2
|
Xu G, Li Y, Lu G, Xie D. Tissue-resident memory T cells in urinary tract diseases. Front Immunol 2025; 16:1535930. [PMID: 40066439 PMCID: PMC11891219 DOI: 10.3389/fimmu.2025.1535930] [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: 11/28/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are a specialized subset of memory T cells that permanently reside in non-lymphoid tissues, providing localized and long-lasting immune protection. In the urinary tract, TRM cells play critical roles in defending against infections, mediating tumor immunity, and influencing the pathogenesis of chronic inflammatory diseases. Their therapeutic potential is immense, with promising avenues for vaccine development, enhanced cancer immunotherapy, and targeted treatments for chronic inflammation. However, challenges remain in harnessing their protective roles while minimizing their pathological effects, particularly in immunosuppressive or inflammatory microenvironments. This review explores the diverse roles of TRM cells in urinary tract diseases, including infections, cancer, and chronic inflammation, and discusses therapeutic strategies and future directions for leveraging TRM cells to improve clinical outcomes. By advancing our understanding of TRM cell biology, we can develop innovative interventions that balance their immune-protective and regulatory functions.
Collapse
Affiliation(s)
- Guofeng Xu
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuying Li
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Respiratory Critical Care, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guanting Lu
- Laboratory of Translational Medicine Research, Deyang People’s Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Daoyuan Xie
- Laboratory of Translational Medicine Research, Deyang People’s Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| |
Collapse
|
3
|
Salomão NG, Araújo L, de Souza LJ, Luiza Young A, Basílio-de-Oliveira C, Basílio-de-Oliveira RP, de Carvalho JJ, Nunes PCG, da Silva Amorim JF, Barbosa DVDS, Paes MV, Rabelo K, Dos Santos F. Chikungunya virus infection in the skin: histopathology and cutaneous immunological response. Front Microbiol 2025; 16:1497354. [PMID: 39935638 PMCID: PMC11811090 DOI: 10.3389/fmicb.2025.1497354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Alphavirus chikungunya virus (CHIKV) is an arbovirus, belonging to the Togaviridae family. The disease caused by CHIKV generally evolves with spontaneous resolution in a few weeks; however, progression to a chronic disease may occur. The most common symptoms are fever, myalgia, and arthralgia; however, skin manifestations may occur in 40 to 80% of infected individuals. Morbilliform and maculopapular erythematous eruptions, vesiculobullous lesions, generalized erythema, maculopapular eruption and skin peeling, hypermelanosis, painful oral lesions, and urticarial lesions have been reported. Usually, these manifestations disappear, but they can become sequelae. Since the skin is the first line of defense against CHIKV infection, in this study, we aimed to investigate the immunohistopathological aspects of the skin of infected individuals during the acute phase of the disease by performing histopathological and ultrastructural analysis, detection and quantification of the viral genome, detection of viral antigen and immune cells, and cytokines/chemokines' characterization. The main histopathological findings were perivascular and inflammatory infiltrates, blood capillary ectasia, and interstitial edema. The immunohistochemistry revealed CHIKV antigen in the epidermis, endothelial cells, fibroblasts, and macrophages in the reticular and papillary dermis; inflammatory cells infiltrate; arrector pili muscle; sweat and sebaceous glands; and hair follicle. Moreover, inflammatory infiltrates were composed of lymphocytes (CD4+ and CD8+) and macrophages (CD68+) in the dermis and perivascular infiltrate. TNF-α, IL-6, RANTES, and VEGFR2 were expressed in the epidermis, blood vessels, sweat glands, and migrating cells. Loss of contact among adjacent keratinocytes, epidermis presenting necrotic cells, and fibroblasts with dilated cisternae in the endoplasmic reticulum and mitochondria with few cristae was observed by transmission electron microscopy. Studies involving skin immunopathogenesis during CHIKV infection are still scarce; therefore, the findings presented here can contribute to a better understanding of the disease immunopathogenesis.
Collapse
Affiliation(s)
- Natália Gedeão Salomão
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| | - Luciana Araújo
- Departamento de Anatomia Patológica, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | | | | | - Carlos Basílio-de-Oliveira
- Departamento de Anatomia Patológica, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | | | - Jorge José de Carvalho
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Priscilla Conrado Guerra Nunes
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| | | | | | - Marciano Viana Paes
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| | - Kíssila Rabelo
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Flavia Dos Santos
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Lin Q, Olkowski CP, Choyke PL, Sato N. Tumor growth suppression in adoptive T cell therapy via IFN-γ targeting of tumor vascular endothelial cells. Theranostics 2024; 14:6897-6912. [PMID: 39629126 PMCID: PMC11610145 DOI: 10.7150/thno.101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 12/06/2024] Open
Abstract
Rationale: In adoptive T cell therapy (ACT), the direct cytotoxic effects of CD8 T cells on tumor cells, including the release of interferon-gamma (IFN-γ), are considered the primary mechanism for tumor eradication. Cancer antigen escape diminishes the T cell responses, thereby limiting the therapeutic success. The impacts of IFN-γ targeting non-tumor cells in ACT, on the other hand, remains under-investigated. We hypothesized that IFN-γ action on non-tumor cells, particularly tumor vascular endothelial cells within the physiological tumor microenvironment, could influence therapeutic efficacy. Methods: ACT was performed against ovalbumin (OVA)- or OVA-peptide SIINFEKL-expressing syngeneic mouse tumors, MCA-205-OVA-GFP fibrosarcoma or MOC2-SIINFEKL oral squamous cell carcinoma, using ex vivo-activated OT-1 CD8 T cells expressing the T cell receptor against OVA. Efficacy was examined in wild-type mice, mice deficient for IFN-γ receptor 1 (IFN-γR1KO), and bone marrow chimeras lacking IFN-γR1 expression in endothelial cells. To exclude direct IFN-γ action against tumor cells, IFN-γR1KO-MCA-205-OVA-GFP tumors were used. IFN-γ production, STAT1 induction in its targets, and subsequent changes, especially in vasculatures in the tumor, were examined. Results: ACT suppressed the growth of MCA-205-OVA-GFP and MOC2-SIINFEKL tumors in wild-type mice but failed in IFNγR1KO mice. Furthermore, in the bone marrow chimeras lacking endothelial cell IFN-γR1, ACT efficacy was lost, thus implicating a vital role of IFN-γ action on the endothelium. IFN-γR1KO-MCA-205-OVA-GFP tumor growth was successfully suppressed by ACT in wild-type mice, suggesting that IFN-γ targeting of tumor cells may not be essential for ACT efficacy. OT-1 CD8 T cells interacted with endothelial cells or localized in proximity to the vessels on Day 1.5 after transfer, as observed by intravital microscopy. The OT-1 T cells found in tumors were limited in number but produced high levels of IFN-γ on Day 1.5, while their number peaked on Day 5.5 with negligible IFN-γ production. Together with IFN-γ production by endogenous lymphocytes, IFN-γ levels in the whole tumor peaked on Day 1.5, inducing IFN-γ/STAT1 signaling in endothelial cells. Early targeting of tumor vascular endothelial cells by IFN-γ led to endothelial regression, reduced perfusion, and tumor hypoxia/necrosis (Day 4.5-7). Conclusions: These findings highlight the critical role of T cell-derived IFN-γ action on endothelial cells early in ACT, emphasizing its dynamic influence on the tumor microenvironment, and offering insights into addressing antigen escape.
Collapse
Affiliation(s)
| | | | | | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Li Y, Li J, Li W, Liang S, Wei W, Chu J, Lai J, Lin Y, Chen H, Su J, Hu X, Wang G, Meng J, Jiang J, Ye L, An S. Scm6A: A Fast and Low-cost Method for Quantifying m6A Modifications at the Single-cell Level. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae039. [PMID: 39436235 PMCID: PMC12016562 DOI: 10.1093/gpbjnl/qzae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/03/2024] [Accepted: 05/24/2024] [Indexed: 10/23/2024]
Abstract
It is widely accepted that N6-methyladenosine (m6A) exhibits significant intercellular specificity, which poses challenges for its detection using existing m6A quantitative methods. In this study, we introduced Single-cell m6A Analysis (Scm6A), a machine learning-based approach for single-cell m6A quantification. Scm6A leverages input features derived from the expression levels of m6A trans regulators and cis sequence features, and offers remarkable prediction efficiency and reliability. To further validate the robustness and precision of Scm6A, we first applied Scm6A to single-cell RNA sequencing (scRNA-seq) data from peripheral blood mononuclear cells (PBMCs) and calculated the m6A levels in CD4+ and CD8+ T cells. We also applied a winscore-based m6A calculation method to conduct N6-methyladenosine sequencing (m6A-seq) analysis on CD4+ and CD8+ T cells isolated through magnetic-activated cell sorting (MACS) from the same samples. Notably, the m6A levels calculated by Scm6A exhibited a significant positive correlation with those quantified through m6A-seq in different cells isolated by MACS, providing compelling evidence for Scm6A's reliability. Additionally, we performed single-cell-level m6A analysis on lung cancer tissues as well as blood samples from patients with coronavirus disease 2019 (COVID-19), and demonstrated the landscape and regulatory mechanisms of m6A in different T cell subtypes from these diseases. In summary, Scm6A is a novel, dependable, and accurate method for single-cell m6A detection and has broad applications in the realm of m6A-related research.
Collapse
Affiliation(s)
- Yueqi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Jingyi Li
- Department of Pathology, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Wenxing Li
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Shuaiyi Liang
- Department of Bioinformatics, Anjin Biotechnology Co., Ltd., Guangzhou 510000, China
| | - Wudi Wei
- Life Sciences Institute & Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Guangxi Medical University, Nanning 530021, China
| | - Jiemei Chu
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jingzhen Lai
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Yao Lin
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Hubin Chen
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jinming Su
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Xiaopeng Hu
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Gang Wang
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jun Meng
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Junjun Jiang
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Li Ye
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Sanqi An
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
6
|
Salazar FC, Martinez MS, Paira DA, Chocobar YA, Olivera C, Godoy GJ, Acosta-Rodriguez EV, Rivero VE, Motrich RD. CD8 T cells are dispensable for experimental autoimmune prostatitis induction and chronic pelvic pain development. Front Immunol 2024; 15:1387142. [PMID: 38807587 PMCID: PMC11130463 DOI: 10.3389/fimmu.2024.1387142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction Chronic Pelvic Pain Syndrome or Chronic Prostatitis (CPPS/CP) is the most prevalent urologic affliction among young adult men. It is a challenging condition to treat, which significantly decreases patient quality of life, mostly because of its still uncertain aetiology. In that regard, an autoimmune origin is a prominent supported theory. Indeed, studies in patients and in rodent models of Experimental Autoimmune Prostatitis (EAP) have provided compelling evidence suggesting a key role of CD4 Th1 cells in disease pathogenesis. However, the implication of other prominent effectors of the immune system, such as CD8 T cells, has yet to be studied. Methods We herein analyzed the induction of prostatitis and the development of chronic pelvic pain in EAP using CD8 T cell-deficient animals. Results We found similarly elevated PA-specific immune responses, with high frequencies of specific IFNg+CD4+ and IL17+CD4+ T cells in prostate draining lymph nodes from PA-immunized either CD8 KO or wild type animals with respect to controls. Moreover, these peripheral immune responses were paralleled by the development of significant chronic pelvic pain, and accompanied by prostate histological lesions, characterized by hemorrhage, epithelial cell desquamation, marked periglandular leukocyte infiltration, and increased collagen deposition in both, PA-immunized CD8 KO and wild type animals. As expected, control animals did not develop prostate histological lesions. Discussion Our results indicate that CD8 T cells do not play a major role in EAP pathogenesis and chronic pelvic pain development. Moreover, our results corroborate the previous notion that a CD4 Th1 associated immune response drives the induction of prostate tissue inflammation and the development of chronic pelvic pain.
Collapse
Affiliation(s)
- Florencia C. Salazar
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Maria S. Martinez
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
- Federation of Clinical Immunology Societies (FOCIS) Center of Excellence Centro de Inmunologia Clinica de Cordoba (CICC), Cordoba, Argentina
| | - Daniela A. Paira
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
- Federation of Clinical Immunology Societies (FOCIS) Center of Excellence Centro de Inmunologia Clinica de Cordoba (CICC), Cordoba, Argentina
| | - Yair A. Chocobar
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
- Federation of Clinical Immunology Societies (FOCIS) Center of Excellence Centro de Inmunologia Clinica de Cordoba (CICC), Cordoba, Argentina
| | - Carolina Olivera
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
- Federation of Clinical Immunology Societies (FOCIS) Center of Excellence Centro de Inmunologia Clinica de Cordoba (CICC), Cordoba, Argentina
| | - Gloria J. Godoy
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Eva V. Acosta-Rodriguez
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Virginia E. Rivero
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
- Federation of Clinical Immunology Societies (FOCIS) Center of Excellence Centro de Inmunologia Clinica de Cordoba (CICC), Cordoba, Argentina
| | - Ruben D. Motrich
- Centro de Investigaciones en Bioquimica Clinica e Inmunologia (CIBICI)-CONICET, Departamento de Bioquimica Clinica, Facultad de Ciencias Quimicas, Universidad Nacional de Cordoba, Cordoba, Argentina
- Federation of Clinical Immunology Societies (FOCIS) Center of Excellence Centro de Inmunologia Clinica de Cordoba (CICC), Cordoba, Argentina
| |
Collapse
|
7
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [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: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
8
|
Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, Luo P, Bai Y. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer 2024; 23:58. [PMID: 38515134 PMCID: PMC10956324 DOI: 10.1186/s12943-024-01972-6] [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: 12/25/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play critical antitumor roles, encompassing diverse subsets including CD4+, NK, and γδ T cells beyond conventional CD8+ CTLs. However, definitive CTLs biomarkers remain elusive, as cytotoxicity-molecule expression does not necessarily confer cytotoxic capacity. CTLs differentiation involves transcriptional regulation by factors such as T-bet and Blimp-1, although epigenetic regulation of CTLs is less clear. CTLs promote tumor killing through cytotoxic granules and death receptor pathways, but may also stimulate tumorigenesis in some contexts. Given that CTLs cytotoxicity varies across tumors, enhancing this function is critical. This review summarizes current knowledge on CTLs subsets, biomarkers, differentiation mechanisms, cancer-related functions, and strategies for improving cytotoxicity. Key outstanding questions include refining the CTLs definition, characterizing subtype diversity, elucidating differentiation and senescence pathways, delineating CTL-microbe relationships, and enabling multi-omics profiling. A more comprehensive understanding of CTLs biology will facilitate optimization of their immunotherapy applications. Overall, this review synthesizes the heterogeneity, regulation, functional roles, and enhancement strategies of CTLs in antitumor immunity, highlighting gaps in our knowledge of subtype diversity, definitive biomarkers, epigenetic control, microbial interactions, and multi-omics characterization. Addressing these questions will refine our understanding of CTLs immunology to better leverage cytotoxic functions against cancer.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and ImmunologySchool of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South University, Hunan, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
9
|
Cadenas-De Miguel S, Lucianer G, Elia I. The metabolic cross-talk between cancer and T cells. Trends Biochem Sci 2023; 48:597-609. [PMID: 37080875 DOI: 10.1016/j.tibs.2023.03.004] [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: 11/10/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 04/22/2023]
Abstract
The metabolic cross-talk between cancer cells and T cells dictates cancer formation and progression. These cells possess metabolic plasticity. Thus, they adapt their metabolic profile to meet their phenotypic requirements. However, the nutrient microenvironment of a tumor is a very hostile niche in which these cells are forced to compete for the available nutrients. The hyperactive metabolism of tumor cells often outcompetes the antitumorigenic CD8+ T cells while promoting the protumorigenic exhausted CD8+ T cells and T regulatory (Treg) cells. Thus, cancer cells elude the immune response and spread in an uncontrolled manner. Identifying the metabolic pathways necessary to shift the balance from a protumorigenic to an antitumorigenic immune phenotype is essential to potentiate antitumor immunity.
Collapse
Affiliation(s)
| | - Giulia Lucianer
- Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Ilaria Elia
- Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
10
|
Teque F, Wegehaupt A, Roufs E, Killian MS. CD8+ Lymphocytes from Healthy Blood Donors Secrete Antiviral Levels of Interferon-Alpha. Viruses 2023; 15:v15040894. [PMID: 37112874 PMCID: PMC10144965 DOI: 10.3390/v15040894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
The adaptive immune response to viral infections features the antigen-driven expansion of CD8+ T cells. These cells are widely recognized for their cytolytic activity that is mediated through the secretion of cytokines such as perforin and granzymes. Less appreciated is their ability to secrete soluble factors that restrict virus replication without killing the infected cells. In this study we measured the ability of primary anti-CD3/28-stimulated CD8+ T cells from healthy blood donors to secrete interferon-alpha. Supernatants collected from CD8+ T cell cultures were screened for their ability to suppress HIV-1 replication in vitro and their interferon-alpha concentrations were measured by ELISA. Interferon-alpha concentrations in the CD8+ T cell culture supernatants ranged from undetectable to 28.6 pg/mL. The anti-HIV-1 activity of the cell culture supernatants was observed to be dependent on the presence of interferon-alpha. Appreciable increases in the expression levels of type 1 interferon transcripts were observed following T cell receptor stimulation, suggesting that the secretion of interferon-alpha by CD8+ T cells is an antigen-driven response. In 42-plex cytokine assays, the cultures containing interferon-alpha were also found to contain elevated levels of GM-CSF, IL-10, IL-13, and TNF-alpha. Together, these results demonstrate that the secretion of anti-viral levels of interferon-alpha is a common function of CD8+ T cells. Furthermore, this CD8+ T cell function likely plays broader roles in health and disease.
Collapse
|