1
|
Zhang M, Ma J, Edwards R, Li M. The dynamics of CD4+ T cell proliferation and regulation. JOURNAL OF BIOLOGICAL DYNAMICS 2025; 19:2458867. [PMID: 39881560 DOI: 10.1080/17513758.2025.2458867] [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: 04/21/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
We use mathematical modeling to study the proliferation dynamics of CD4+ T cells within an immune response. This proliferation is driven by the autocrine reaction of helper T cells and interleukin-2 (IL-2), and regulated by natural regulatory T cells (nTregs). Previous studies suggested that a fratricidal mechanism is necessary to eliminate helper T cells post-infection. Contrary to this, our mathematical analysis establishes that the depletion of these cells is due to two pivotal factors: the saturation in the proliferation rate of helper CD4+ T cells at high IL-2 concentrations, and the activation rate of nTregs outpacing their death rate. This yields an excitable process, such that the proliferation starts once the helper T cell population passes a threshold. Additionally, we find that when the proliferation of nTregs lags behind their mortality, induced regulatory T cells (iTregs) are crucial to curbing the proliferation of helper CD4+ T cells.
Collapse
Affiliation(s)
- Mingran Zhang
- College of Information Science and Technology, Donghua University, Shanghai, People's Republic of China
| | - Junling Ma
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada
| | - Roderick Edwards
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada
| | - Meili Li
- School of Mathematics and Statistics, Donghua University, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Phan NM, Nguyen TL, Min DK, Kim J. Mesoporous polydopamine nanoparticle-based tolerogenic vaccine induces antigen-specific immune tolerance to prevent and treat autoimmune multiple sclerosis. Biomaterials 2025; 316:122997. [PMID: 39662275 DOI: 10.1016/j.biomaterials.2024.122997] [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: 12/21/2023] [Revised: 10/24/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Multiple sclerosis (MS) is a chronic neurological disorder derived from autoreactive immune system attacking the protective myelin sheath that surrounds nerves in the central nervous system (CNS). Here, a tolerogenic nanovaccine for generating an antigen-specific immune tolerance for treating MS is proposed. It consisted of a mesoporous polydopamine (mPDA) nanoparticle, characterized by high reactive oxygen species (ROS)-scavenging property, loaded with MS-derived autoantigen. Intravenous vaccination of autoantigen-loaded mPDA could induce tolerogenic dendritic cells (DCs) with low expression of co-stimulatory molecules while presenting peptide epitopes. The tolerogenic DCs induced peripheral regulatory T-cells (Tregs), thereby reducing infiltration of autoreactive CD4+ T-cells and inflammatory antigen-presenting cells (APCs) into the CNS. In MS-mimicking mouse model, the tolerogenic nanovaccine prevented MS development in the early therapeutic setup and exhibited an enhanced recovery from complete paralysis in the late therapeutic model. The current platform could be exploited to treat other autoimmune diseases where disease-dependent autoantigen peptides are delivered.
Collapse
Affiliation(s)
- Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Dong Kwang Min
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Department of MetaBioHealth, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Institute of Quantum Biophysics (IQB), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
3
|
Surico PL, Barone V, Singh RB, Coassin M, Blanco T, Dohlman TH, Basu S, Chauhan SK, Dana R, Di Zazzo A. Potential applications of mesenchymal stem cells in ocular surface immune-mediated disorders. Surv Ophthalmol 2025; 70:467-479. [PMID: 39097173 DOI: 10.1016/j.survophthal.2024.07.008] [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: 05/17/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
We explore the interaction between corneal immunity and mesenchymal stem/stromal cells (MSCs) and their potential in treating corneal and ocular surface disorders. We outline the cornea's immune privilege mechanisms and the immunomodulatory substances involved. In this realm, MSCs are characterized by their immunomodulatory properties and regenerative potential, making them promising for therapeutic application. Therefore, we focus on the role of MSCs in immune-mediated corneal diseases such as dry eye disease, corneal transplantation rejection, limbal stem cell deficiency, and ocular graft-versus-host disease. Preclinical and clinical studies demonstrate MSCs' efficacy in promoting corneal healing and reducing inflammation in these conditions. Overall, we emphasize the potential of MSCs as innovative therapies in ophthalmology, offering promising solutions for managing various ocular surface pathologies.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Vincenzo Barone
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Marco Coassin
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Thomas H Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sayan Basu
- Brien Holden Eye Research Centre (BHERC), L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Antonio Di Zazzo
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy.
| |
Collapse
|
4
|
Maes M, Vasupanrajit A, Jirakran K, Zhou B, Tunvirachaisakul C, Almulla AF. Simple dysmood disorder, a mild subtype of major depression, is not an inflammatory condition: Depletion of the compensatory immunoregulatory system. J Affect Disord 2025; 375:75-85. [PMID: 39848470 DOI: 10.1016/j.jad.2025.01.101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/13/2024] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND A recent study conducted by the laboratory of the first author revealed that major depression is composed of two distinct subtypes: major dysmood disorder (MDMD) and simple dysmood disorder (SDMD). The latter is a less severe phenotype with fewer aberrant biological pathways. MDMD, but not SDMD, patients were identified to have highly sensitized cytokine/growth factor networks using stimulated whole blood cultures. However, no information regarding serum cytokines/chemokines/growth factors in SDMD is available. OBJECTIVES This case-control study compares 48 serum cytokines/chemokines/growth factors in academic students with SDMD (n = 64) and first episode (FE)-SDMD (n = 47) to those of control students (n = 44) using a multiplex assay. FINDINGS Both FE-SDMD and SDMD exhibited a notable inhibition of immune profiles, such as the compensatory immunoregulatory response system (CIRS) and alternative M2 macrophage and T helper-2 (Th-2) profiles. We observed a substantial reduction in the serum concentrations of five proteins: interleukin (IL)-4, IL-10, soluble IL-2 receptor (sIL-2R), IL-12p40, and macrophage colony-stimulating factor. A considerable proportion of the variability observed in suicidal behaviors (26.7 %) can be accounted for by serum IL-4, IL-10, and sIL-2R (all decreased), CCL11 (eotaxin) and granulocyte CSF (both increased). The same biomarkers (except for IL-10), accounted for 25.5 % of the variance in SDMS severity. A significant correlation exists between decreased levels of IL-4 and elevated ratings of the brooding type of rumination. CONCLUSIONS SDMD is characterized by the suppression of the CIRS profile, which signifies a disruption of immune homeostasis and tolerance, rather than the presence of an inflammatory response.
Collapse
Affiliation(s)
- Michael Maes
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Fitness and Biopsychological Technology Research Unit, Faculty of Medicine Chulalongkorn University, Bangkok 10330, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Asara Vasupanrajit
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ketsupar Jirakran
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Center of Excellence for Maximizing Children's Developmental Potential, Department of Pediatric, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
| | - Chavit Tunvirachaisakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Abbas F Almulla
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq.
| |
Collapse
|
5
|
Ziogas DC, Theocharopoulos C, Aravantinou K, Boukouris AE, Stefanou D, Anastasopoulou A, Lialios PP, Lyrarakis G, Gogas H. Clinical benefit of immune checkpoint inhibitors in elderly cancer patients: Current evidence from immunosenescence pathophysiology to clinical trial results. Crit Rev Oncol Hematol 2025; 208:104635. [PMID: 39889861 DOI: 10.1016/j.critrevonc.2025.104635] [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: 06/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
The age-related decline in immunity appears to be associated not only with cancer development but also with differential responses to immune checkpoint inhibitors (ICIs). Despite their increasing utility across various malignancies and therapeutic settings, limited data -derived primarily from subgroup analyses of randomized controlled trials (RCTs), pooled meta-analyses, and retrospective studies- are available on the effects of aging on their efficacy and toxicity. Immunosenescence, characterized by the progressive decline of the function of the immune system, and inflammaging, a state of persistent low-grade sterile inflammation, may influence ICI outcomes. Additionally, the incidence, severity, and subtypes of immune-related adverse events (irAEs) may differ between older and younger individuals due to loss of immunotolerance. In the current review, starting from a a comprehensive discussion of the pathophysiology of immunosenescence, we proceed to critically review age-related retrospective and randomized evidence supporting FDA-approved ICIs. We highlight similarities or differences across age groups and the clinical benefit of ICIs in elderly versus younger cancer patients. The optimal integration of ICIs in geriatric oncology necessitates greater inclusion of this patient demographic in RCTs along with real-world data in order to acquire robust data which will guide evidence-based treatment decisions for this population.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Katerina Aravantinou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Aristeidis E Boukouris
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Dimitra Stefanou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Amalia Anastasopoulou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - George Lyrarakis
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
6
|
Eggenhuizen PJ, Ng BH, Lo C, Chang J, Snelgrove SL, Cheong RMY, Shen C, Lim S, Zhong Y, Gan PY, Ooi JD. Engineered antigen-specific T regulatory cells suppress autoreactivity to the anti-glomerular basement membrane disease antigen. Kidney Int 2025; 107:751-756. [PMID: 39842638 DOI: 10.1016/j.kint.2025.01.005] [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: 05/17/2024] [Revised: 12/15/2024] [Accepted: 01/02/2025] [Indexed: 01/24/2025]
Abstract
Anti-glomerular basement membrane (anti-GBM) disease is accompanied by insufficient antigen-specific regulatory T cells (Tregs) and clonally expanded antigen-specific conventional T cells. In particular, this applied to the immunodominant T cell autoepitope of type IV collagen, α3(IV)NC1135-145, presented by human leukocyte antigen-DRB1∗1501. Here, we investigated whether Tregs engineered to express GBM-T cell receptors (TCR) specific for α3(IV)NC1135-145 better suppress autoimmunity. The GBM-TCR Treg cell product exhibited a phenotypically stable Treg phenotype, produced α3(IV)NC1135-145-specific functional responses, and were superior suppressors of autoreactive conventional T cells and bystander conventional T cells compared to polyclonal Tregs or irrelevant TCR-transduced Tregs. We also found that GBM-TCR Tregs modulate other immune cells like dendritic cells and B cells to a more tolerogenic phenotype. Importantly, our findings support the development of GBM-TCR Tregs as a promising cell-based therapy for anti-GBM disease.
Collapse
Affiliation(s)
- Peter J Eggenhuizen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Boaz H Ng
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Cecilia Lo
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Janet Chang
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Sarah L Snelgrove
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Rachel M Y Cheong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Chanjuan Shen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; Department of Hematology, the Affiliated Zhuzhou Hospital of Xiangya Medical College, Central South University, Zhuzhou, China
| | - Steven Lim
- Alfred Research Alliance Flow Cytometry Core Facility, Melbourne, Victoria, Australia
| | - Yong Zhong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Poh-Yi Gan
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Joshua D Ooi
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
7
|
Johnson B, Guo Q, Chaludiya K, Kim S. The Proimmunomodulatory and Anti-immunomodulatory Effects of Radiotherapy in Oncologic Care. Hematol Oncol Clin North Am 2025; 39:399-411. [PMID: 39827043 PMCID: PMC11932133 DOI: 10.1016/j.hoc.2024.11.009] [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] [Indexed: 01/22/2025]
Abstract
The abscopal effect in radiotherapy (RT) refers to the phenomenon where localized radiation treatment causes regression of distant, nonirradiated tumors. Although rare, recent research shows that combining radiation with immunotherapies, such as immune checkpoint inhibitors, can enhance this effect. The interaction between radiation-induced cell death, immune responses, and the tumor microenvironment manifests in competing biologic mechanisms resulting in complex immunologic outcomes. In order to maximize the therapeutic advantages of the immunogenic effect of RT in the future, further studies are needed to fully understand its biologic underpinnings.
Collapse
Affiliation(s)
- Bryan Johnson
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Qianyu Guo
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA; Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Keyur Chaludiya
- Department of Laboratory Medicine, Mayo Clinic Minnesota, 150 3rd Street SW, Rochester, MN 55902, USA
| | - Sungjune Kim
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA.
| |
Collapse
|
8
|
Yang L, Zheng SG. Role of regulatory T cells in inflammatory liver diseases. Autoimmun Rev 2025:103806. [PMID: 40139456 DOI: 10.1016/j.autrev.2025.103806] [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: 07/08/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The liver is the human body's largest digestive gland, which can participate in digestion, metabolism, excretion, detoxification and immunity. Chronic liver diseases such as metabolic dysfunction-associated fatty liver disease (MAFLD) or viral hepatitis involve ongoing inflammation and resulting liver fibrosis may ultimately lead to the development of hepatobiliary cancers (HCC). Inflammation is the coordinated reaction of different liver cell types to cell signals and death of inflammation, which are linked to injury pathways within the liver or external agents from the gut-liver axis and the circulation. Regulatory T (Treg) cells play a crucial role in controlling inflammation and are essential for maintaining immune tolerance and balance. In this review, we highlight the recent discoveries related to the function of immune systems in liver inflammation and discuss the role of Treg cells in the different liver diseases (including MAFLD, autoimmune hepatitis and others).
Collapse
Affiliation(s)
- Linjie Yang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Song Guo Zheng
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China; Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; State Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 201600, China.
| |
Collapse
|
9
|
Enzler T, Frankel TL. Pancreatic Cancer Precursor Lesions - Can Immunotherapy Prevent Progression into Pancreatic Ductal Adenocarcinoma? Cancer Lett 2025:217662. [PMID: 40127814 DOI: 10.1016/j.canlet.2025.217662] [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: 11/12/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, with a 5-year survival rate of only 12.5%. Early detection of PDAC or addressing risk factors for PDAC development are ways to improve outcomes. PDAC can arise from precursor lesions, including pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasm (IPMN), and less frequent, mucinous cystic neoplasm (MCN), and other rare precursor variants. High-risk precursor lesions harbor a substantial chance of evolving into PDAC. Such lesions can often be found in resected PDAC specimens adjacent to the cancer. Unfortunately, recognizing precursor lesions that need to be resected is often tricky, and resections frequently end in major surgical interventions. Thus, better ways to handle precursor lesions are desperately needed. We mapped the immune microenvironments (IMEs) of PanINs, IPMNs, and MCNs on a cellular level using multiplex immunofluorescence and computational imaging technology and compared the findings to PDACs and normal pancreatic tissues. We found distinct and potentially targetable mechanisms of immunosuppression between the two main precursor lesions, PanIN and IMPN. Immunosuppression in IPMNs seems partly mediated by programmed cell death protein 1 ligand (PD-L1) expression on antigen-presenting cells (APCs). By contrast, elevated numbers of regulatory T cells (Tregs) seem to be key players in the immunosuppression of PanINs. Thus, treating high-risk IPMNs with anti-PD-1 and high-risk PanINs with agents targeting Tregs, such as anti-lymphocyte associated protein 4 (anti-CTLA-4) antibodies, could reverse their immunosuppressive state. Reversal of immunosuppression will restore immunosurveillance and eventually prevent progression into PDAC. We also review relevant published and ongoing non-surgical treatment approaches for high-risk IPMNs and PanINs.
Collapse
Affiliation(s)
- Thomas Enzler
- Department of Medicine, University of Michigan, Ann Arbor, MI 40109.
| | | |
Collapse
|
10
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
11
|
Deguchi H, Nagata K, Inaba T, Aoki T, Kitano H, Sotozono C. Novel diagnostic method for B cell vitreoretinal lymphoma by identification of regulatory T cells and PD-1 + cytotoxic T lymphocytes in the vitreous via flow cytometry. Br J Ophthalmol 2025; 109:510-515. [PMID: 39401866 DOI: 10.1136/bjo-2024-326240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/23/2024] [Indexed: 03/22/2025]
Abstract
AIMS To investigate the significance of regulatory T cells (Tregs) and programmed cell death 1 (PD-1)+ cytotoxic T lymphocytes (CTLs) in the vitreous of patients with vitreoretinal lymphoma (VRL) and uveitis. METHODS This study involved 51 patients with VRL and uveitis, 15 males and 36 females (mean age: 72 years, range: 51-86 years), who underwent vitrectomy at the Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan, from December 2019 to February 2024. All patients underwent lymphocyte surface antigen analysis via flow cytometry, and the proportion of Tregs in CD4+ T lymphocytes and PD-1+ CTLs in CD8+ T lymphocytes was measured. RESULTS This study involved B cell VRL (B-VRL, n=16), sarcoidosis (n=8), human herpesvirus (HHV)-associated uveitis (n=7), human T cell lymphotropic virus type 1 associated uveitis (HAU, n=3) and unclassifiable uveitis (n=17) cases. The median proportions of Tregs were significantly lower in B-VRL (2.2%) compared with sarcoidosis (8.5%), HHV-associated uveitis (16.4%) and unclassifiable uveitis (10.1%) (p<0.05). Conversely, a significantly higher proportion of PD-1+ CTLs was found in B-VRL (95.6%) compared with sarcoidosis (61.1%), HHV-associated uveitis (67.1%) and unclassifiable (64.8%) (p<0.05). Receiver operating characteristic analysis of Tregs and PD-1+ CTLs proportions in B-VRL revealed high area under the curve values of 0.913 and 0.940, respectively. CONCLUSIONS Our findings indicate that analysis of the ratio of Tregs and PD-1+ CTLs via flow cytometry is helpful in diagnosing B-VRL.
Collapse
Affiliation(s)
- Hideto Deguchi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Buck Institute for Research on Aging, Novato, California, USA
| | - Kenji Nagata
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tohru Inaba
- Department of Infection Control & Laboratory Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takanori Aoki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hikaru Kitano
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Ophthalmology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
Luo S, Larson JH, Blazar BR, Abdi R, Bromberg JS. Foxp3 +CD8 + regulatory T cells: bona fide Tregs with cytotoxic function. Trends Immunol 2025:S1471-4906(25)00053-5. [PMID: 40113537 DOI: 10.1016/j.it.2025.02.010] [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: 01/03/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Various mammalian CD8+ T cell subsets with regulatory properties are either formed through a thymus-dependent mechanism or induced under various experimental protocols and referred to as CD8+ regulatory T cells (Tregs). CD8+ Tregs maintain distinct functions in the presence of CD4+ Tregs. This review focuses on the Foxp3+CD8+ Treg subset, which is typically extremely rare in unmanipulated mice and healthy humans under steady-state conditions. However, they can be induced and expanded for transplantation, autoimmune diseases, cancer, viral infections, and T cell receptor transgenic adoptive cell transfer models. Here, we summarize recent research progress related to this population, including the identification of phenotypic markers, induction determinants, and functional activities. Additionally, we discuss advances in manipulating Foxp3+CD8+ Tregs in autoimmunity and transplantation.
Collapse
Affiliation(s)
- Shunqun Luo
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Jemma H Larson
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN 55455
| | - Bruce R Blazar
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN 55455
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201.
| |
Collapse
|
13
|
Paul P, Choong C, Heinemann J, Al-Hallaf R, Agha Z, Ganatra S, Abdulrahman L, Sinha A, Kumar H, Nourbakhsh B, Hamad ARA. The Lasting Impact of IL-2: Approaching 50 Years of Advancing Immune Tolerance, Cancer Immunotherapies, and Autoimmune Diseases. Immunol Invest 2025:1-15. [PMID: 40094273 DOI: 10.1080/08820139.2025.2479609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BACKGROUND The discovery of interleukin-2 (IL-2) and its receptor (IL-2R) almost 50 years ago revolutionized immunology, marking a pivotal moment in understanding T cell biology and immune regulation. Initially identified as a T cell growth factor, IL-2 unveiled critical insights into cytokine-mediated immune cell proliferation and differentiation. METHODS This review highlighted the characterization of IL-2R as a multi-chain receptor complex set a precedent for decoding cytokine receptor signaling. The unique interplay between IL-2 and its high-affinity receptor component, IL-2Rα, epitomizes the principle of specificity and efficiency in cytokine signaling, enabling precise immune modulation. Regulatory T cells (Tregs) exploit IL-2Rα high affinity to outcompete effector T cells for IL-2, ensuring immune tolerance and preventing autoimmunity. RESULTS Despite its foundational role in immune homeostasis, leveraging IL-2 for therapeutic purposes has proven challenging. CONCLUSION IL-2-based therapies hold transformative potential in autoimmunity, cancer immunology, and transplantation, yet they remain elusive due to the complex balance between immunostimulatory and immunosuppressive effects. This review explores the milestones in IL-2 biology, its dualistic functions, and the ongoing quest to harness its therapeutic promise.
Collapse
Affiliation(s)
- Prajita Paul
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cherry Choong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph Heinemann
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rafid Al-Hallaf
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zainab Agha
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shaan Ganatra
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lina Abdulrahman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Agastya Sinha
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Harrsha Kumar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Abdel Rahim A Hamad
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Wen Y, Zhao J, Zhang Z. Heterogeneity and longitudinal transcriptomic characteristics of Tregs in COVID-19 patients. Front Immunol 2025; 16:1548173. [PMID: 40114921 PMCID: PMC11922936 DOI: 10.3389/fimmu.2025.1548173] [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: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance by suppressing immune responses against pathogens. The fluctuation of Treg proportions in COVID-19 remains a topic of debate, and the mechanisms triggering Treg activation in COVID-19 are still unclear. Understanding these issues is essential for better managing immune responses in COVID-19 patients. Methods We collected a cohort of COVID-19 patients with varying disease severity and stage to explore the transcriptomic and functional traits of Tregs in these individuals. Using transcriptomic analysis, we evaluated the proportion and functionality of different Treg subsets, specifically HLA_DR+ Tregs, across different stages of COVID-19 patients. Results Our analysis revealed that the proportion of CCR7 + Tregs decreased as the disease advanced, while the cell proportion of HLA_DR+ regs escalated with the severity of the disease. Moreover, the transcription actor CARHSP1 exhibited apositive correlation with the proportion of HLA_DR+ Tregs. Notably, the heightened suppressive function of HLA_DR+ Tregs in severe COVID-19 patients, with interactions between PF4 and CXCR3, contributed to the homeostasis of HLA_DR+ Tregs in severe COVID-19 patients. Furthermore, we observed that Tregs in COVID-19 patients exhibited weakened TCR clonotype expansion, and the suppression of HLA_DR+ Tregs with expanded TCR clonotypes in severe COVID-19 cases did not show a significant increase compared to asymptomatic and mild COVID-19 groups. The findings indicate that Tregs may be activated through the bystander effect, as evidenced by the analysis of TCR clonotype characteristics. Discussion Our research delineates the diversity of dynamic alterations in Tregs and sheds light on potential mechanisms underlying Treg activation, providing a theoretical foundation and offering treatment strategies for managing COVID-19 patients.
Collapse
Affiliation(s)
- Yanling Wen
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Juanjuan Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Cui Y, David M, Bouchareychas L, Rouquier S, Sajuthi S, Ayrault M, Navarin C, Lara G, Lafon A, Saviane G, Boulakirba S, Menardi A, Demory A, Frikeche J, de la Forest Divonne Beghelli S, Lu HH, Dumont C, Abel T, Fenard D, de la Rosa M, Gertner-Dardenne J. IL23R-Specific CAR Tregs for the Treatment of Crohn's Disease. J Crohns Colitis 2025; 19:jjae135. [PMID: 39252592 DOI: 10.1093/ecco-jcc/jjae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/18/2024] [Accepted: 09/09/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND AND AIMS Regulatory T cells (Tregs) are key regulators in maintaining tissue homeostasis. Disrupted immune homeostasis is associated with Crohn's disease (CD) pathogenesis. Thus, Treg therapy represents a promising long-acting treatment to restore immune balance in the diseased intestine. Chimeric antigen receptor (CAR) T-cell therapy has revolutionized cancer treatment. This innovative approach also provides the opportunity to improve therapy for CD. By targeting a disease-relevant protein, interleukin-23 receptor (IL23R), we engineered Tregs expressing IL23R-CAR for treating active CD. METHODS Intestinal IL23R expression from active CD was verified by immunohistochemical analysis. Phenotypic and functional characteristics of IL23R-CAR Tregs were assessed using in vitro assays and their migration capacity was monitored in a xenograft tumor model. Transcriptomic and proteomic analyses were performed to associate molecular profiles with IL23R-CAR Treg activation against colon biopsy-derived cells from active CD patients. RESULTS Our study showed that IL23R-CAR displayed negligible tonic signaling and a strong signal-to-noise ratio. IL23R-CAR Tregs maintained regulatory phenotype during in vitro expansion, even when chronically exposed to proinflammatory cytokines and target antigen. IL23R engagement on IL23R-CAR Tregs triggered CAR-specific activation and significantly enhanced their suppressive activity. Also, IL23R-CAR Tregs migrated to IL23R-expressing tissue in humanized mice. Finally, IL23R-CAR Tregs elicited a specific activation against colon biopsy-derived cells from active CD, suggesting an efficient CAR engagement in active CD. Molecular profiling of CD patient biopsies also revealed transcriptomic and proteomic patterns associated with IL23R-CAR activation. CONCLUSIONS Overall, our results demonstrate that IL23R-CAR Tregs represent a promising therapy for active CD.
Collapse
Affiliation(s)
- Yue Cui
- Research, Sangamo Therapeutics, Valbonne, France
| | - Marion David
- Research, Sangamo Therapeutics, Valbonne, France
| | | | | | | | | | | | - Gregory Lara
- Research, Sangamo Therapeutics, Valbonne, France
| | - Audrey Lafon
- Research, Sangamo Therapeutics, Valbonne, France
| | | | | | | | | | | | | | | | | | - Tobias Abel
- Research, Sangamo Therapeutics, Valbonne, France
| | - David Fenard
- Research, Sangamo Therapeutics, Valbonne, France
| | | | | |
Collapse
|
16
|
Poorva P, Mast J, Cao B, Shah MV, Pollok KE, Shen J. Killing the killers: Natural killer cell therapy targeting glioma stem cells in high-grade glioma. Mol Ther 2025:S1525-0016(25)00168-6. [PMID: 40040281 DOI: 10.1016/j.ymthe.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
High-grade gliomas (HGGs), including glioblastoma (GBM) in adults and diffuse intrinsic pontine glioma (DIPG) in children, are among the most aggressive and deadly brain tumors. A key factor in their resilience is the presence of glioma stem cells (GSCs), which drive tumor initiation, progression, and resistance to treatment. Targeting and eradicating GSCs holds potential for curing both GBM and DIPG. Natural killer (NK) cells, as part of the innate immune system, naturally recognize and destroy malignant cells. Recent advances in NK cell-based therapies, such as chimeric antigen receptor (CAR)-NK cells, NK cell engagers, and NK cell-derived exosomes, offer promising approaches for treating GBM and DIPG, particularly by addressing the persistence of GSCs. This review highlights these advancements, explores challenges such as the blood-brain barrier and the immunosuppressive tumor microenvironment, and proposes future directions for improving and clinically advancing these NK cell-based therapies for HGGs.
Collapse
Affiliation(s)
- Poorva Poorva
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Jensen Mast
- Biochemistry Graduate Program, Indiana University, Bloomington, IN 47405, USA
| | - Bihui Cao
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Mitesh V Shah
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen E Pollok
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
17
|
Chen X, Zuo Z, Li L, Liu L, Bao X, Song R, Wang Y, Wang L, Zhu M, Wang Y. The effect of Time-Acupoints-Space Acupuncture on fatigue in postoperative chemotherapy patients with breast cancer: a randomized controlled trial. Front Oncol 2025; 15:1518278. [PMID: 40098703 PMCID: PMC11911492 DOI: 10.3389/fonc.2025.1518278] [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: 10/28/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Background Breast cancer (BC) is a common malignant tumor in women, and cancer-related fatigue (CRF) is prevalent among breast cancer patients. Time-Acupoints-Space Acupuncture (ATAS) is an acupuncture method different from traditional acupuncture. It combines time acupoints with space acupoints, proposing a new treatment approach. This randomized controlled trial aims to evaluate whether ATAS can improve fatigue in postoperative chemotherapy patients with breast cancer. Objective This randomized controlled trial focuses on survivors of postoperative chemotherapy for breast cancer, primarily assessing whether ATAS can reduce fatigue in these patients. Additionally, it reports on the effects of ATAS on sleep, anxiety, depression, and inflammatory factors. Methods The researchers randomly assigned 90 postoperative breast cancer patients to the ATAS group (n=30), the sham acupuncture group (n=30), and the waitlist control group (n=30). The primary outcome was the Piper Fatigue Scale (PFS), and the secondary outcomes were the Insomnia Severity Index (ISI), Hospital Anxiety and Depression Scale (HADS), Interleukin-2 (IL-2), Interleukin-6 (IL-6), CD3+T, and CD4+T. Data analysis was performed using the statistical software SPSS, utilizing descriptive statistics and analytic statistics. The significance level was set at less than 0.05. Results The baseline differences in PFS scores among the three groups were not statistically significant (P > 0.05). ATAS treatment is superior to sham acupuncture and the waitlist control in improving fatigue (mean difference 4.98, 95% CI 3.96 to 6.00, P<0.05). Additionally, secondary outcome analysis shows that the ATAS group has positive effects on ISI, HADS, and inflammatory factors. After the treatment ended, ISI (mean difference 15.17, 95% CI 12.28 to 18.06, P<0.05), HADS-A (mean difference 8.63, 95% CI 5.18 to 12.08, P<0.05), HADS-D (mean difference 7.80, 95% CI 4.73 to 10.87, P<0.05). IL-2(mean difference 20.18, 95% CI 11.51 to 28.85, P<0.05), IL-6(mean difference 24.56, 95% CI 7.57 to 41.55, P<0.05), CD3+T(mean difference 79.03, 95% CI 68.56 to 89.50, P<0.05), CD4+T(mean difference 42.89, 95% CI 35.14 to 50.64, P<0.05). Conclusions Our preliminary findings indicate that ATAS effectively improves fatigue in postoperative chemotherapy patients with breast cancer. It also has positive effects on sleep, anxiety, depression, and inflammatory factors. These results suggest that ATAS intervention may be an effective method for alleviating fatigue in breast cancer patients. Clinical Trial Registration https://www.chictr.org.cn/showproj.html?proj=21999, identifier ChiCTR17013652.
Collapse
Affiliation(s)
- Xin Chen
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zheng Zuo
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Li
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Liangxian Liu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiongying Bao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Ran Song
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Yinghao Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | | | - Miansheng Zhu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
- ARIATAS, Association Pour la Recherche et I’Information de I’Acupuncture Time-Acupoints-Space, College of Acupuncture, Paris, France
| | - Yan Wang
- Second Department of Acupuncture and Moxibustion, Dali Bai Autonomous Prefecture Chinese Medicine Hospital, Dali, China
| |
Collapse
|
18
|
Xu W, Zhou B, Wang P, Ma Y, Jiang Y, Mo D, Wu J, Ma J, Wang X, Miao Y, Nian Y, Zheng J, Li J, Yan F, Li G. N6-methyladenosine modification of 3'tRF-AlaAGC impairs PD-1 blockade efficacy by promoting lactic acid accumulation in the tumor microenvironment of gastric carcinoma. Drug Resist Updat 2025; 79:101197. [PMID: 39752904 DOI: 10.1016/j.drup.2024.101197] [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: 10/18/2024] [Revised: 12/05/2024] [Accepted: 12/21/2024] [Indexed: 02/24/2025]
Abstract
The balance between CD8+ T cells and regulatory T (Treg) cells in the tumor microenvironment (TME) plays a crucial role in the immune checkpoint inhibition (ICI) therapy in gastric carcinoma (GC). However, related factors leading to the disturbance of TME and resistance to ICI therapy remain unknown. In this study, we applied N6-methyladenosine (m6A) small RNA Epitranscriptomic Microarray and screened out 3'tRF-AlaAGC based on its highest differential expression level and lowest inter-group variance. N6-methyladenosine modification significantly enhanced the stability of 3'tRF-AlaAGC, which strengthened glycolysis and lactic acid (LA) production in GC cells by binding to PTBP1 (Polypyrimidine Tract Binding Protein 1). In the peritoneal GC implantation model established in huPBMC-NCG mice, 3'tRF-AlaAGC significantly increased the proportion of PD1+ Treg cells. Furthermore, in high-LA environments driven by glucose consumption of GC cells, Treg cells actively uptake LA through MCT1, facilitating NFAT1 translocation into the nucleus and enhancing PD1 expression, whereas PD1 expression by effector T cell is diminished. Meanwhile, T cell suppression assays were performed under low-LA or high-LA conditions, and the proliferation of CD8+ T cells was dampened by adding Sintilimab in a high-LA but not in a low-LA environment, suggesting the preferential activation of PD1+ Treg cell. These findings deciphered the complexities of the immune microenvironment in GC, providing prospects for identifying robust biomarkers that could improve the evaluation of therapeutic effectiveness and prognosis in immune therapy for GC.
Collapse
Affiliation(s)
- Weiguo Xu
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Bin Zhou
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Ping Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Shanghai, China
| | - Yuyan Ma
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yu Jiang
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Dongping Mo
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Jun Wu
- Department of Clinical Laboratory, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Ma
- Institute of Agri-products Processing, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Xiao Wang
- Department of Radiology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yinxing Miao
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Yong Nian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Junyu Zheng
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Jie Li
- Department of General Surgery, Huaian Hospital, Huaian, China
| | - Feng Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Gang Li
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China.
| |
Collapse
|
19
|
Lopez de Rodas M, Villalba-Esparza M, Sanmamed MF, Chen L, Rimm DL, Schalper KA. Biological and clinical significance of tumour-infiltrating lymphocytes in the era of immunotherapy: a multidimensional approach. Nat Rev Clin Oncol 2025; 22:163-181. [PMID: 39820025 DOI: 10.1038/s41571-024-00984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have improved clinical outcomes across several solid tumour types. Prominent efforts have focused on understanding the anticancer mechanisms of these agents, identifying biomarkers of response and uncovering resistance mechanisms to develop new immunotherapeutic approaches. This research has underscored the crucial roles of the tumour microenvironment and, particularly, tumour-infiltrating lymphocytes (TILs) in immune-mediated tumour elimination. Numerous studies have evaluated the prognostic and predictive value of TILs and the mechanisms that govern T cell dysfunction, fuelled by technical developments in single-cell transcriptomics, proteomics, high-dimensional spatial platforms and advanced computational models. However, questions remain regarding the definition of TILs, optimal strategies to study them, specific roles of different TIL subpopulations and their clinical implications in different treatment contexts. Additionally, most studies have focused on the abundance of major TIL subpopulations but have not developed standardized quantification strategies or analysed other crucial aspects such as their functional profile, spatial distribution and/or arrangement, tumour antigen-reactivity, clonal diversity and heterogeneity. In this Review, we discuss a conceptual framework for the systematic study of TILs and summarize the evidence regarding their biological properties and biomarker potential for ICI therapy. We also highlight opportunities, challenges and strategies to support future developments in this field.
Collapse
Affiliation(s)
- Miguel Lopez de Rodas
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba-Esparza
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada and Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
20
|
Cui X, Song Y, Han J, Yuan Z. The multifaceted role of SMAD4 in immune cell function. Biochem Biophys Rep 2025; 41:101902. [PMID: 39802394 PMCID: PMC11721226 DOI: 10.1016/j.bbrep.2024.101902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) signaling pathway, with SMAD4 as its central mediator, plays a pivotal role in regulating cellular functions, including growth, differentiation, apoptosis, and immune responses. While extensive research has elucidated SMAD4's role in tumorigenesis, its functions within immune cells remain underexplored. This review synthesizes current knowledge on SMAD4's diverse roles in various immune cells such as T cells, B cells, dendritic cells, and macrophages, highlighting its impact on immune homeostasis and pathogen response. Understanding SMAD4's role in immune cells is crucial, as its dysregulation can lead to autoimmune disorders, chronic inflammation, and immune deficiencies. The review emphasizes the significance of SMAD4 in immune regulation, proposing that deeper investigation could reveal novel therapeutic targets for immune-mediated conditions. Insights into SMAD4's involvement in processes like T cell differentiation, B cell class switch recombination, and macrophage polarization underscore its potential as a therapeutic target for a range of diseases, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Xinmu Cui
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Jianfeng Han
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
- Cellular Biomedicine Group Inc, Shanghai, 201203, China
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| |
Collapse
|
21
|
Dhar S, Sarkar T, Bose S, Pati S, Chakraborty D, Roy D, Panda AK, Guin A, Mukherjee S, Jana K, Sarkar DK, Sa G. FOXP3 Transcriptionally Activates Fatty Acid Scavenger Receptor CD36 in Tumour-Induced Treg Cells. Immunology 2025; 174:296-309. [PMID: 39736083 DOI: 10.1111/imm.13887] [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: 01/20/2024] [Revised: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
The host immune system is adapted in a variety of ways by tumour microenvironment and growing tumour interacts to promote immune escape. One of these adaptations is manipulating the metabolic processes of cells in the tumour microenvironment. The growing tumour aggressively utilise glucose, its primary energy source available in tumour site, and produce lactate by Warburg effect. In such a hostile environment, tumour-infiltrating immune cells are unable to survive metabolically. Tumour-infiltrating CD4+ Treg cells, on the other hand, adapted to an alternative energy-generating system, switching from the highly-competitive glucose to the fatty-acid metabolic pathway, by down-regulating glucose-metabolising genes and up-regulating fatty-acid metabolising genes. Tregs with high-levels of the fatty acid scavenger receptor CD36, a key component of the fatty-acid metabolic pathway, aided this metabolic shift. Treg cell formation was hampered when the fatty-acid metabolic pathway was disrupted, showing that it is necessary for Treg cell development. FOXP3, the Treg lineage-specific transcription factor, regulates fatty-acid metabolism by inducing CD36 transcription. A high-fat diet enhanced Treg development while suppressing anti-tumour immunity, whereas a low-fat diet suppressed Treg development. The altered metabolism of tumour-infiltrating Treg cells enables their rapid generation and survival in the hostile tumour microenvironment, aiding cancer progression. Fascinatingly, mice fed with a low-fat diet showed a positive prognosis with chemotherapy than mice fed with a high-fat diet. Thus, a maximum efficacy of chemotherapy might be achieved by altering diet composition during chemotherapy, providing a promising indication for future cancer treatment.
Collapse
Affiliation(s)
- Subhanki Dhar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Tania Sarkar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sayantan Bose
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Dia Roy
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Abir K Panda
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Aharna Guin
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sumon Mukherjee
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
22
|
Zhou X, Dunham D, Sindher SB, Long A, Fernandes A, Chang I, Assa'ad A, Pongracic J, Spergel JM, Tam J, Tilles S, Wang J, Boyd SD, Chinthrajah RS, Nadeau KC. HLA-DR + regulatory T cells and IL-10 are associated with success or failure of desensitization outcomes. Allergy 2025; 80:762-774. [PMID: 39291303 DOI: 10.1111/all.16311] [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: 02/14/2024] [Revised: 07/14/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Omalizumab (XOLAIR®)-assisted multi-food oral immunotherapy (mOIT) has been shown to safely, effectively, and rapidly desensitize patients with multiple food allergies. In our clinical trial (NCT02626611) on omalizumab-assisted mOIT, different desensitization outcomes (success or failure of desensitization) were observed following a period of either continued or discontinued mOIT. However, the association between the immunological changes induced by omalizumab-assisted mOIT and desensitization outcomes has not yet been fully elucidated. In this study, due to the key roles of regulatory T (Treg) cells and the type 2 helper T cell (Th2) pathway in immune tolerance to food allergens, we aimed to characterize their association with the desensitization outcomes of omalizumab-assisted mOIT. METHODS Mass cytometry and multiplex cytokine assays were performed on blood samples obtained from participants with allergies to peanut, cashew, or milk in our phase 2 clinical study (NCT02626611). Comprehensive statistical and bioinformatic analyses were conducted on high-dimensional cytometry-based single-cell data and high-throughput multiplex cytokine data. RESULTS Our results demonstrated that the frequency of HLA-DR+ Treg cells, and the production of Th2 cytokines (IL-4, IL-5, IL-13, and IL-9) as well as the immunoregulatory cytokine IL-10 by peripheral blood mononuclear cells (PBMCs) was significantly increased in cultures with allergen compared to cultures with media alone at baseline (Week 0). We also observed increased frequency of allergen responsive HLA-DR+ Treg cells and enhanced production of IL-10 by PBMCs in participants who achieved successful desensitization compared to those with failure of desensitization. However, the production of Th2 cytokines by PBMCs did not show significant differences between participants with different desensitization outcomes (success vs. failure of desensitization), despite omalizumab-assisted mOIT inducing a significant reduction in the production of Th2 cytokines. CONCLUSIONS We demonstrated that the frequency of HLA-DR+ Treg cells and IL-10 cytokine production by PBMCs are associated with desensitization outcomes of omalizumab-assisted mOIT. These findings suggest potential immunological parameters that could be targeted to enhance desensitization success rates.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Diane Dunham
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Sayantani B Sindher
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Andrew Long
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Andrea Fernandes
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Iris Chang
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Amal Assa'ad
- Division of Allergy and Immunology, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA
| | - Jacqueline Pongracic
- Division of Allergy and Immunology, The Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan Tam
- Division of Clinical Immunology and Allergy, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Stephen Tilles
- Seattle Allergy and Asthma Research Institute, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| | - Julie Wang
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Scott D Boyd
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - R Sharon Chinthrajah
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Huang W, Wang J, Liu C, Yang C, Chen Z, Ding J, Jiang W, Wang Y, Meng Y, Li L, Liu Y, Liu X, Li H, Sun B. Norepinephrine promotes activated B cells to identify and kill effector CD8 + T cells through FasL/Fas pathway in spleen mononuclear cells isolated from experimental autoimmune encephalomyelitis. Brain Behav Immun 2025; 125:294-307. [PMID: 39824471 DOI: 10.1016/j.bbi.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025] Open
Abstract
It has been reported that the nervous system can regulate immune reactions through various mechanisms. However, the role of splenic sympathetic nerve activity in the autoimmune reactions during the pathogenesis of experimental autoimmune encephalomyelitis (EAE) remained unclear. Here, we blocked the activity of the splenic sympathetic nerve and found that the number of adaptive immune cells, such as CD4+ T cells, CD8+ T cells and B cells, were upregulated. Additionally, there was an increase in the secretion of inflammatory cytokines in the spleen, and the neurological symptoms of EAE were exacerbated. In vitro experiments, we found that norepinephrine (NE), the neurotransmitter of the splenic sympathetic nerve, indirectly drove the death of effector CD8+ T cells. Furthermore, activated B cells, under the influence of NE, specifically recognized effector CD8+ T cells by upregulating MHC-I molecules and killed these cells via the FasL/Fas pathway. Our findings provide a new perspective on B cells killing effect in vitro, which was boosted by NE and demonstrate that the splenic sympathetic nerve controls the degree of autoimmune responses in EAE. This adds a new dimension to the diversity of NE's regulatory effects on adaptive immune cells and suggests a potential new therapeutic approach for autoimmune diseases.
Collapse
Affiliation(s)
- Wei Huang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Chao Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Changxin Yang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Zhengyi Chen
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Jianwen Ding
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Wenkang Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Yanping Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Yanting Meng
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Lei Li
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang, PR China
| | - Yumei Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Xijun Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Hulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China.
| | - Bo Sun
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China.
| |
Collapse
|
24
|
Fang X, Mo C, Zheng L, Gao F, Xue FS, Zheng X. Transfusion-Related Acute Lung Injury: from Mechanistic Insights to Therapeutic Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413364. [PMID: 39836498 PMCID: PMC11923913 DOI: 10.1002/advs.202413364] [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: 10/21/2024] [Revised: 12/08/2024] [Indexed: 01/23/2025]
Abstract
Transfusion-related acute lung injury (TRALI) is a potentially lethal complication of blood transfusions, characterized by the rapid onset of pulmonary edema and hypoxemia within six hours post-transfusion. As one of the primary causes of transfusion-related mortality, TRALI carries a significant mortality rate of 6-12%. However, effective treatment strategies for TRALI are currently lacking, underscoring the urgent need for a comprehensive and in-depth understanding of its pathogenesis. This comprehensive review provides an updated and detailed analysis of the current landscape of TRALI, including its clinical presentation, pathogenetic hypotheses, animal models, cellular mechanisms, signaling pathways, and potential therapeutic targets. By highlighting the critical roles of these pathways and therapies, this review offers valuable insights to inform the development of preventative and therapeutic strategies and to guide future research efforts aimed at addressing this life-threatening condition.
Collapse
Affiliation(s)
- Xiaobin Fang
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Zheng
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Fei Gao
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Fu-Shan Xue
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University & Fujian Emergency Medical Center, Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Key Laboratory of Critical Medicine, Fujian Provincial Co-constructed Laboratory of "Belt and Road,", Fuzhou, Fujian, China
| |
Collapse
|
25
|
Dadey RE, Cui J, Rajasundaram D, Yano H, Liu C, Cohen JA, Liu AW, Kaplan DH, Workman CJ, Vignali DAA. Regulatory T cells in the tumor microenvironment display a unique chromatin accessibility profile. Immunohorizons 2025; 9:vlae014. [PMID: 39965167 PMCID: PMC11841976 DOI: 10.1093/immhor/vlae014] [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: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 02/20/2025] Open
Abstract
Regulatory T cells (Tregs) are a suppressive CD4+ T cell population that limit the antitumor immune response. In this study, we analyzed the chromatin accessibility of Tregs in the murine tumor microenvironment (TME) to identify tumor-specific accessible peaks and if these are altered over time in the tumor microenvironment, with or without anti-PD-1 immunotherapy. We found that despite little change in chromatin accessibility of Tregs in the tumor over time, Tregs have a distinct chromatin accessibility signature in the TME compared with Tregs in the periphery. This distinct tumor Treg chromatin accessibility profile highlights reduced accessibility at loci important for an CD4+ conventional T cell (CD4+ Foxp3-) effector phenotype. Analysis of chromatin accessibility in Tregs from B16 and MC38 tumor models indicated that Tregs from skin-resident tumors are most similar to naïve skin resident Tregs but still bear key differences attributable to the TME. We also found that Tregs do not alter their transcriptome or chromatin accessibility following immunotherapy. We conclude that although chromatin accessibility in Tregs is somewhat similar to their tissue residency, the TME may drive a unique chromatin accessibility profile. Treg chromatin accessibility in the tumor appears remarkably stable and unaltered by tumor type, over time, or following immunotherapy.
Collapse
Affiliation(s)
- Rebekah E Dadey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dhivyaa Rajasundaram
- Division of Health Informatics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chang Liu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Jonathan A Cohen
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Andrew W Liu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daniel H Kaplan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
26
|
Zheng J, Wang S, Xia L, Sun Z, Chan KM, Bernards R, Qin W, Chen J, Xia Q, Jin H. Hepatocellular carcinoma: signaling pathways and therapeutic advances. Signal Transduct Target Ther 2025; 10:35. [PMID: 39915447 PMCID: PMC11802921 DOI: 10.1038/s41392-024-02075-w] [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: 05/21/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 02/09/2025] Open
Abstract
Liver cancer represents a major global health concern, with projections indicating that the number of new cases could surpass 1 million annually by 2025. Hepatocellular carcinoma (HCC) constitutes around 90% of liver cancer cases and is primarily linked to factors incluidng aflatoxin, hepatitis B (HBV) and C (HCV), and metabolic disorders. There are no obvious symptoms in the early stage of HCC, which often leads to delays in diagnosis. Therefore, HCC patients usually present with tumors in advanced and incurable stages. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of HCC. Beyond the frequently altered and therapeutically targeted receptor tyrosine kinase (RTK) pathways in HCC, pathways involved in cell differentiation, telomere regulation, epigenetic modification and stress response also provide therapeutic potential. Investigating the key signaling pathways and their inhibitors is pivotal for achieving therapeutic advancements in the management of HCC. At present, the primary therapeutic approaches for advanced HCC are tyrosine kinase inhibitors (TKI), immune checkpoint inhibitors (ICI), and combination regimens. New trials are investigating combination therapies involving ICIs and TKIs or anti-VEGF (endothelial growth factor) therapies, as well as combinations of two immunotherapy regimens. The outcomes of these trials are expected to revolutionize HCC management across all stages. Here, we provide here a comprehensive review of cellular signaling pathways, their therapeutic potential, evidence derived from late-stage clinical trials in HCC and discuss the concepts underlying earlier clinical trials, biomarker identification, and the development of more effective therapeutics for HCC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhen Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, PR China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
27
|
Wang Y, Sun Y, Zhang X, Wang S, Huang X, Xu K, Liu Y, Huang Y, Xu J, Wei X, Cheng H, Pan L, Wang J, Gu Z. A Granzyme B-Cleavable T Cell-Targeted Bispecific Cell Vesicle Connector for Reversing New-Onset Type 1 Diabetes. J Am Chem Soc 2025; 147:4167-4179. [PMID: 39869523 DOI: 10.1021/jacs.4c13644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder in which pancreatic β-cells are destroyed by CD8+ T cells. Anti-CD3 antibody effectively treats early-stage T1D when β-cell autoantibodies are detected but before symptoms appear. However, it impairs the immune system temporarily, exposing individuals to infection. A therapeutic that can reverse new-onset T1D without harming the immune system remains urgently needed. Herein, we have constructed cellular vesicles presenting granzyme B-responsive fusion proteins (designated aCD8-GrzBcs-IL2) composed of a single-chain variable fragment of anti-CD8 antibodies and a mutein interleukin-2 (IL2). aCD8-GrzBcs-IL2 is designed to simultaneously inhibit CD8+ T cells and promote Treg cells, especially when CD8+ T cells are attacking β-cells. In vitro, these cellular vesicles can inhibit the cell-killing effect of CD8+ T cells and enhance the expansion of Treg cells. Notably, intravenous administration of aCD8-GrzBcs-IL2-expressed cellular vesicles reversed newly onset diabetes in 77.8% of nonobese diabetic (NOD) mice without reducing blood CD3+ T cells and CD8+ T cells, indicating a favorable safety profile.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanping Sun
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiuwen Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xuehui Huang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Kairui Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yun Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingqi Huang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianchang Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinwei Wei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Liqiang Pan
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Liangzhu Laboratory, Hangzhou 311121, China
- Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
28
|
Garcia-Fabiani MB, Haase S, Banerjee K, Zhu Z, McClellan BL, Mujeeb AA, Li Y, Tronrud CE, Varela ML, West ME, Yu J, Kadiyala P, Taher AW, Núñez FJ, Alghamri MS, Comba A, Mendez FM, Nicola Candia AJ, Salazar B, Nunez FM, Edwards MB, Qin T, Cartaxo RT, Niculcea M, Koschmann C, Venneti S, Vallcorba MP, Nasajpour E, Pericoli G, Vinci M, Kleinman CL, Jabado N, Chandler JP, Sonabend AM, DeCuypere M, Hambardzumyan D, Prolo LM, Mahaney KB, Grant GA, Petritsch CK, Welch JD, Sartor MA, Lowenstein PR, Castro MG. H3.3-G34R Mutation-Mediated Epigenetic Reprogramming Leads to Enhanced Efficacy of Immune Stimulatory Gene Therapy in Diffuse Hemispheric Gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.06.13.544658. [PMID: 37398299 PMCID: PMC10312611 DOI: 10.1101/2023.06.13.544658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Diffuse hemispheric glioma (DHG), H3 G34-mutant, representing 9-15% of cases, are aggressive Central Nervous System (CNS) tumors with poor prognosis. This study examines the role of epigenetic reprogramming of the immune microenvironment and the response to immune-mediated therapies in G34-mutant DHG. To this end, we utilized human G34-mutant DHG biopsies, primary G34-mutant DHG cultures, and genetically engineered G34-mutant mouse models (GEMMs). Our findings show that the G34 mutation alters histone marks' deposition at promoter and enhancer regions, leading to the activation of the JAK/STAT pathway, which in turn results in an immune-permissive tumor microenvironment. The implementation of Ad-TK/Ad-Flt3L immunostimulatory gene therapy significantly improved median survival, and lead to over 50% long term survivors. Upon tumor rechallenge in the contralateral hemisphere without any additional treatment, the long-term survivors exhibited robust anti-tumor immunity and immunological memory. These results indicate that immune-mediated therapies hold significant potential for clinical translation in treating patients harboring H3.3-G34 mutant DHGs, offering a promising strategy for improving outcomes in this challenging cancer subtype affecting adolescents and young adults (AYA). STATEMENT OF SIGNIFICANCE This study uncovers the role of the H3.3-G34 mutation in reprogramming the tumor immune microenvironment in diffuse hemispheric gliomas. Our findings support the implementation of precision medicine informed immunotherapies, aiming at improving enhanced therapeutic outcomes in adolescents and young adults harboring H3.3-G34 mutant DHGs.
Collapse
Affiliation(s)
- Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Present address: Leloir Institute Foundation, Buenos Aires, Argentina
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ziwen Zhu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anzar A. Mujeeb
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yingxiang Li
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Claire E. Tronrud
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria L. Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Molly E.J. West
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jin Yu
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
- Present address: Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ayman W. Taher
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Felipe J. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Flor M. Mendez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alejandro J. Nicola Candia
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brittany Salazar
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando M. Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marta B. Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rodrigo T. Cartaxo
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Michael Niculcea
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Carl Koschmann
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Sriram Venneti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Emon Nasajpour
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Giulia Pericoli
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Maria Vinci
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Claudia L. Kleinman
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - James P. Chandler
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael DeCuypere
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Division of Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Laura M. Prolo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
- Present address: Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Claudia K Petritsch
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
29
|
Fisher MS, Sennikov SV. T-regulatory cells for the treatment of autoimmune diseases. Front Immunol 2025; 16:1511671. [PMID: 39967659 PMCID: PMC11832489 DOI: 10.3389/fimmu.2025.1511671] [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: 10/15/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Autoimmune diseases result from imbalances in the immune system and disturbances in the mechanisms of immune tolerance. T-regulatory cells (Treg) are key factors in the formation of immune tolerance. Tregs modulate immune responses and repair processes, controlling the innate and adaptive immune system. The use of Tregs in the treatment of autoimmune diseases began with the manipulation of endogenous Tregs using immunomodulatory drugs. Then, a method of adoptive transfer of Tregs grown in vitro was developed. Adoptive transfer of Tregs includes polyclonal Tregs with non-specific effects and antigen-specific Tregs in the form of CAR-Treg and TCR-Treg. This review discusses non-specific and antigen-specific approaches to the use of Tregs, their advantages, disadvantages, gaps in development, and future prospects.
Collapse
Affiliation(s)
- Marina S. Fisher
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University under the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Sergey V. Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University under the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
30
|
Buckner JH. Antigen-specific immunotherapies for autoimmune disease. Nat Rev Rheumatol 2025; 21:88-97. [PMID: 39681709 DOI: 10.1038/s41584-024-01201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Antigen-specific therapies have a long history in the treatment of allergy but have not been successful in autoimmunity. However, in the past 20 years, advances in the definition of the self-antigens that promote autoimmunity and the growing understanding of the mechanisms that maintain tolerance in health but fail in autoimmunity have led to antigen-specific approaches being considered for the treatment of autoimmune diseases. The core goal of each antigen-specific treatment approach is to remove the immune response that promotes autoimmunity whilst sparing protective responses. Approaches to antigen-specific therapy range from targeted deletion of autoreactive lymphocytes to tolerization of autoreactive T cells and active inhibition of autoimmune responses. Technologies such as vaccines, nanoparticles, cell-based therapies and gene editing are being harnessed to achieve these goals. Remaining challenges include the selection of the best antigen to target, modality and timing of administration of these therapies and the disease in which the therapies are used; overcoming these challenges will be vital to move antigen-specific therapies forward. Once established, antigen-specific therapy has the potential to be applied broadly in the area of autoimmunity.
Collapse
Affiliation(s)
- Jane H Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| |
Collapse
|
31
|
Mohseni SO, Au KM, Issa W, Ruan L, Stuve O, Wang AZ. Multiple sclerosis treatments a review of current biomedical engineering approaches. Biomaterials 2025; 313:122807. [PMID: 39241553 DOI: 10.1016/j.biomaterials.2024.122807] [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: 05/21/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
Multiple Sclerosis (MS) is an autoimmune condition targeting the central nervous system (CNS) characterized by focal demyelination with inflammation, causing neurodegeneration and gliosis. This is accompanied by a refractory period in relapsing MS or chronic progression in primary progressive MS. Current MS treatments target disease relapses and aim to reduce further demyelination and disability. These include the treatment of acute exacerbations through global immunomodulation upon corticosteroid administration, which are accompanied by adverse reactions. Disease modifying therapies (DMTs) which provide targeted immunosuppression of T and B cells, and sequestration of leukocytes out of CNS, have led to further improvements in demyelination prevention and disease burden reduction. Despite their efficacy, DMTs are ineffective in remyelination, pathology reversal and have minimal effects in progressive MS. The advent of modern biomedical engineering approaches in combination with a better understanding of MS pathology, has led to the development of novel, regenerative approaches to treatment. Such treatments utilize neural stem cells (NSCs) and can reduce disease relapses and reverse damage caused by the disease through localized tissue regeneration. While at initial stages, pre-clinical and clinical studies utilizing NSCs and immune modulation have shown promising outcomes in tissue regeneration, creating a potential new era in MS therapy.
Collapse
Affiliation(s)
- Sayyed Ourmazd Mohseni
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kin Man Au
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Wadih Issa
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Lifu Ruan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew Z Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
32
|
Dong C, Zhao Y, Han Y, Li M, Wang G. Targeting glutamine metabolism crosstalk with tumor immune response. Biochim Biophys Acta Rev Cancer 2025; 1880:189257. [PMID: 39746457 DOI: 10.1016/j.bbcan.2024.189257] [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: 04/05/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Glutamine, akin to glucose, is a fundamental nutrient for human physiology. Tumor progression is often accompanied by elevated glutamine consumption, resulting in a disrupted nutritional balance and metabolic reprogramming within the tumor microenvironment. Furthermore, immune cells, which depend on glutamine for metabolic support, may experience functional impairments and dysregulation. Although the role of glutamine in tumors has been extensively studied, the specific impact of glutamine competition on immune responses, as well as the precise cellular alterations within immune cells, remains incompletely understood. In this review, we summarize the consequences of glutamine deprivation induced by tumor-driven glutamine uptake on immune cells, assessing the underlying mechanisms from the perspective of various components of the immune microenvironment. Additionally, we discuss the potential synergistic effects of glutamine supplementation and immunotherapy, offering insights into future research directions. This review provides compelling evidence for the integration of glutamine metabolism and immunotherapy as a promising strategy in cancer therapy.
Collapse
Affiliation(s)
- Chenshuang Dong
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Yan Zhao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yecheng Han
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Guiling Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
33
|
Ahn J, Kim B, Bello AB, Moon JJ, Arai Y, Lee SH. Regenerative Functions of Regulatory T Cells and Current Strategies Utilizing Mesenchymal Stem Cells in Immunomodulatory Tissue Regeneration. Tissue Eng Regen Med 2025; 22:167-180. [PMID: 39804546 PMCID: PMC11794763 DOI: 10.1007/s13770-024-00690-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are essential for maintaining immune homeostasis and facilitating tissue regeneration by fostering an environment conducive to tissue repair. However, in damaged tissues, excessive inflammatory responses can overwhelm the immunomodulatory capacity of Tregs, compromising their functionality and potentially hindering effective regeneration. Mesenchymal stem cells (MSCs) play a key role in enhancing Treg function. MSCs enhance Treg activity through indirect interactions, such as cytokine secretion, and direct interactions via membrane proteins. METHODS This review examines the regenerative functions of Tregs across various tissues, including bone, cartilage, muscle, and skin, and explores strategies to enhance Treg functionality using MSCs. Advanced techniques, such as the overexpression of relevant genes in MSCs, are highlighted for their potential to further enhance Treg function. Additionally, emerging technologies utilizing extracellular vesicles (EVs) and cell membrane-derived vesicles derived from MSCs offer promising alternatives to circumvent the potential side effects associated with live cell therapies. This review proposes approaches to enhance Treg function and promote tissue regeneration and also outlines future research directions. RESULTS AND CONCLUSION This review elucidates recent technological advancements aimed at enhancing Treg function using MSCs and examines their potential to improve tissue regeneration efficiency.
Collapse
Grants
- 2022R1A2C3004850 Ministry of Science and ICT, South Korea
- RS-2024-00405381 Ministry of Science and ICT, South Korea
- RS-2023-00257290 Ministry of Science and ICT, South Korea
- RS-2023-00246418 Ministry of Education
- RS-2023-00275407 Ministry of Education
- 21C0703L1 Ministry of Science and ICT, Ministry of Health & Welfare
- HX23C1734 Ministry of Science and ICT, Ministry of Trade, Industry and Energy, Ministry of Health & Welfare, The Ministry of Food and Drug Safety
- Ministry of Science and ICT, Ministry of Health & Welfare
- Ministry of Science and ICT, Ministry of Trade, Industry and Energy, Ministry of Health & Welfare, The Ministry of Food and Drug Safety
Collapse
Affiliation(s)
- Jinsung Ahn
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea.
| |
Collapse
|
34
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Pfnür A, Mayer B, Dörfer L, Tumani H, Spitzer D, Huber-Lang M, Kapapa T. Regulatory T Cell- and Natural Killer Cell-Mediated Inflammation, Cerebral Vasospasm, and Delayed Cerebral Ischemia in Aneurysmal Subarachnoid Hemorrhage-A Systematic Review and Meta-Analysis Approach. Int J Mol Sci 2025; 26:1276. [PMID: 39941044 PMCID: PMC11818301 DOI: 10.3390/ijms26031276] [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: 12/10/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) involves a significant influx of blood into the cerebrospinal fluid, representing a severe form of stroke. Despite advancements in aneurysm closure and neuro-intensive care, outcomes remain impaired due to cerebral vasospasm and delayed cerebral ischemia (DCI). Previous pharmacological therapies have not successfully reduced DCI while improving overall outcomes. As a result, significant efforts are underway to better understand the cellular and molecular mechanisms involved. This review focuses on the activation and effects of immune cells after SAH and their interactions with neurotoxic and vasoactive substances as well as inflammatory mediators. Particular attention is given to clinical studies highlighting the roles of natural killer (NK) cells and regulatory T cells (Treg) cells. Alongside microglia, astrocytes, and oligodendrocytes, NK cells and Treg cells are key contributors to the inflammatory cascade following SAH. Their involvement in modulating the neuro-inflammatory response, vasospasm, and DCI underscores their potential as therapeutic targets and prognostic markers in the post-SAH recovery process. We conducted a systematic review on T cell- and natural killer cell-mediated inflammation and their roles in cerebral vasospasm and delayed cerebral ischemia. We conducted a meta-analysis to evaluate outcomes and mortality in studies focused on NK cell- and T cell-mediated mechanisms.
Collapse
Affiliation(s)
- Andreas Pfnür
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, University of Ulm, Helmholtzstr. 22, 89081 Ulm, Germany
| | - Lena Dörfer
- Institute for Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstr. 8/, 89081 Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, University Hospital Ulm, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Daniel Spitzer
- Department of Neurology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstr. 8/, 89081 Ulm, Germany
| | - Thomas Kapapa
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| |
Collapse
|
36
|
Li X, Hu D. Ligand-restricted synNotch switches enable precision cell therapy. Trends Immunol 2025; 46:91-93. [PMID: 39875238 PMCID: PMC11835521 DOI: 10.1016/j.it.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
Lim and colleagues demonstrate that synNotch transcriptional circuits engineered into T cells can be used to precisely control location-specific expression of payloads responding to antigen triggers, thus locally inhibiting unwanted immunity or neuroinflammation. With no off-tumor toxicity or systemic immunosuppression upon elimination of mouse brain tumors, this approach can achieve better efficacy than anticipated.
Collapse
Affiliation(s)
- Xuyang Li
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Ludwig Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Lustgarten Dedicated Laboratory for Pancreatic Cancer Research and the Bloomberg~Kimmel Institute Cancer Genetics and Genomics Research Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dan Hu
- Ann Romney Center for Neurologic Diseases, Harvard Medical School and Mass General Brigham, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Wang A, Wang Y, Liang R, Li B, Pan F. Improving regulatory T cell-based therapy: insights into post-translational modification regulation. J Genet Genomics 2025; 52:145-156. [PMID: 39357622 DOI: 10.1016/j.jgg.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Regulatory T (Treg) cells are pivotal for maintaining immune homeostasis and play essential roles in various diseases, such as autoimmune diseases, graft-versus-host disease (GVHD), tumors, and infectious diseases. Treg cells exert suppressive function via distinct mechanisms, including inhibitory cytokines, granzyme or perforin-mediated cytolysis, metabolic disruption, and suppression of dendritic cells. Forkhead Box P3 (FOXP3), the characteristic transcription factor, is essential for Treg cell function and plasticity. Cumulative evidence has demonstrated that FOXP3 activity and Treg cell function are modulated by a variety of post-translational modifications (PTMs), including ubiquitination, acetylation, phosphorylation, methylation, glycosylation, poly(ADP-ribosyl)ation, and uncharacterized modifications. This review describes Treg cell suppressive mechanisms and summarizes the current evidence on PTM regulation of FOXP3 and Treg cell function. Understanding the regulatory role of PTMs in Treg cell plasticity and function will be helpful in designing therapeutic strategies for autoimmune diseases, GVHD, tumors, and infectious diseases.
Collapse
Affiliation(s)
- Aiting Wang
- Center for Cancer Immunology Research, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| | - Yanwen Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Liang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Fan Pan
- Center for Cancer Immunology Research, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
38
|
Picone F, Giudice V, Iside C, Venturini E, Di Pietro P, Vecchione C, Selleri C, Carrizzo A. Lymphocyte Subset Imbalance in Cardiometabolic Diseases: Are T Cells the Missing Link? Int J Mol Sci 2025; 26:868. [PMID: 39940640 PMCID: PMC11816853 DOI: 10.3390/ijms26030868] [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: 12/18/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Cardiometabolic and cardiovascular diseases (CVDs) remain the leading cause of death worldwide, with well-established risk factors such as smoking, obesity, and diabetes contributing to plaque formation and chronic inflammation. However, emerging evidence suggests that the immune system plays a more significant role in the development and progression of CVD than previously thought. Specifically, the finely tuned regulation of lymphocyte subsets governs post-injury inflammation and tissue damage resolution and orchestrates the functions and activation of endothelial cells, cardiomyocytes, and fibroblasts in CVD-associated lesions (e.g., atherosclerotic plaques). A deeper understanding of the immune system's involvement in CVD development and progression will provide new insights into disease biology and uncover novel therapeutic targets aimed at re-establishing immune homeostasis. In this review, we summarize the current state of knowledge on the distribution and involvement of lymphocyte subsets in CVD, including atherosclerosis, diabetes, hypertension, myocardial infarction, and stroke.
Collapse
Affiliation(s)
- Francesca Picone
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Concetta Iside
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | | | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| |
Collapse
|
39
|
Filoni J, Ferrari A, Jofra T, Putignano AR, Da Dalt L, Cesarano S, Di Dedda C, Bonacina F, Marchesi F, Norata GD, Bonini C, Piemonti L, Monti P. Metabolic reprogramming of naïve regulatory T cells by IL-7 and IL-15 promotes their persistence and performance upon adoptive transfer. Commun Biol 2025; 8:99. [PMID: 39838096 PMCID: PMC11751088 DOI: 10.1038/s42003-024-07381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Tregs for adoptive therapy are traditionally expanded ex vivo using high doses of IL-2. However, the final Treg product has limited survival once infused in patients, potentially affecting therapeutic effectiveness. Here, we tested a novel expansion protocol in which highly purified naïve Tregs were expanded with a combination of IL-7 and IL-15, in the absence of IL-2. The final Treg product was enriched with cells displaying an immature CD45RA+CD62L+CD95+ phenotype, reminiscent of conventional memory stem T cells. The combination of IL-7 and IL-15 confers Tregs a glycolytic metabolism and improved metabolic fitness, characterized by an increased capacity to adapt metabolism according to glucose and oxygen availability. Tregs expanded with IL-7 and IL-15 showed longer persistence and an improved capacity to control xeno-GvHD in NSG mice. This work suggests that metabolic reprogramming induced by IL-7 and IL-15 provides better Treg performance for adoptive therapy.
Collapse
Affiliation(s)
- Jessica Filoni
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Arianna Ferrari
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Tatiana Jofra
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Anna Rita Putignano
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital Rozzano, Rozzano, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Susanna Cesarano
- Experimental Hematology Unit, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Carla Di Dedda
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Federica Marchesi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Monti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy.
| |
Collapse
|
40
|
Dhayanithy G, Radhakrishnan S, Ann Martin C, Caroline Martin J, Hakeem AR, Jothimani D, Kalkura SN, Rela M. Understanding immunological insights of liver transplantation: a practice for attaining operational tolerance. Clin Exp Immunol 2025; 219:uxae125. [PMID: 39973343 DOI: 10.1093/cei/uxae125] [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: 04/17/2024] [Revised: 11/15/2024] [Accepted: 02/19/2025] [Indexed: 02/21/2025] Open
Abstract
Liver transplantation has been at the forefront of medical research, with efforts concentrated on understanding the intricate cellular and molecular dynamics involved this complex procedure. This body of work has chronicled critical clinical advancements, identified challenges, and highlighted progressive improvements in surgical practices. These concerted efforts have significantly contributed to the evolution and enhancement of liver transplantation, elevating it to its current level of sophistication. A successful liver transplant now demands an integrated, multidisciplinary approach that includes not only expanding the donor pool from deceased to living donors but also embracing advances in surgical methods, efficiently managing post-transplant complications, and, importantly, achieving operational tolerance. The latter, operational tolerance, is a state wherein the recipient's immune system is coaxed into accepting the transplanted organ without the long-term use of immunosuppressive drugs, thereby minimizing potential side effects, and improving quality of life. Understanding the critical immune mechanisms that aim to prevent graft rejection is essential from an immunological perspective. This review aims to highlight the crucial areas of host versus graft immune responses, making a clear distinction between organs received from living and deceased donors. It examines how these immune responses, both innate and adaptive, are initiated and proposes the exploration of molecular docking sites as a strategy to curb unwanted immune reactions. Additionally, this review explores the promising potential of biomarkers in predicting graft rejection, and emphasizes the importance of achieving tolerance and the continuous quest for innovative strategies to enhance the success and longevity of liver transplants.
Collapse
Affiliation(s)
| | | | | | - Josette Caroline Martin
- Department of Pathology, Sri Venkateshwara Medical College Hospital and Research Institute, Pondicherry, India
| | | | - Dinesh Jothimani
- Dr. Rela Institute and Medical Centre, Chromepet, Chennai, India
| | - Subbaraya Narayana Kalkura
- Crystal Growth Centre, Anna University, Guindy, Chennai, India
- National Foundation for Liver Research, Chromepet, Chennai, India
| | - Mohamed Rela
- National Foundation for Liver Research, Chromepet, Chennai, India
- Dr. Rela Institute and Medical Centre, Chromepet, Chennai, India
| |
Collapse
|
41
|
Nuiyen A, Sanguansermsri D, Sayasathid J, Thatsakorn K, Thapmongkol S, Ngoenkam J, Pongcharoen S. Nck1 regulates the in vitro development of human regulatory T cells through AKT pathway. Clin Exp Immunol 2025; 219:uxaf011. [PMID: 39963999 PMCID: PMC11923542 DOI: 10.1093/cei/uxaf011] [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: 04/20/2024] [Revised: 12/13/2024] [Accepted: 02/14/2025] [Indexed: 03/21/2025] Open
Abstract
T cell receptor (TCR) signalling is crucial in determining the fate of thymocyte differentiation in the thymus. The high-avidity interaction between TCR and self-peptide-MHC complexes induces development of regulatory T cells (Tregs), lineage commitment for which is controlled by expression of transcription factor Forkhead box P3 (FoxP3). The non-catalytic region of the tyrosine kinase (Nck) comprises two members, Nck1 and Nck2, with Nck1 playing a dominant role in TCR-mediated T cell activation and function. Nck's role, while established in thymocyte development, remains unelucidated in development of Tregs. In this study, we aimed to determine the function of Nck1 in the in vitro development and differentiation of human thymocytes. Human thymocytes were transfected with shRNA plasmid to silence Nck1 expression. The number of FoxP3+ Tregs decreased noticeably in Nck1 knockdown thymocytes after co-cultivation with myeloid dendritic cells (mDCs) and thymic epithelial cells for 14 days. Furthermore, decreased phosphorylation of AKT and FoxO1 was observed in Nck1-silenced thymocytes, in association with reduced FoxO1 nuclear localization. Taken together, these findings identify the pivotal role of Nck1 in Treg development.
Collapse
MESH Headings
- Humans
- T-Lymphocytes, Regulatory/immunology
- Oncogene Proteins/metabolism
- Oncogene Proteins/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Cell Differentiation/immunology
- Forkhead Box Protein O1/metabolism
- Forkhead Box Protein O1/genetics
- Cells, Cultured
- Thymocytes/immunology
- Thymocytes/cytology
- Thymocytes/metabolism
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Phosphorylation
- Coculture Techniques
- Lymphocyte Activation/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Aussanee Nuiyen
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Donruedee Sanguansermsri
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Jarun Sayasathid
- Division of Cardiothoracic Surgery, Department of Surgery, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| | - Kanthachat Thatsakorn
- Division of Cardiothoracic Surgery, Department of Surgery, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| | - Siraphop Thapmongkol
- Division of Cardiothoracic Surgery, Department of Surgery, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
42
|
Glamočlija S, Sabljić L, Tomić S, Đokić J, Radulović N, Gruden-Movsesijan A, Kosanović M. Trichinella spiralis extracellular vesicles induce anti-inflammatory and regulatory immune responses in vitro. Int J Parasitol 2025:S0020-7519(25)00008-6. [PMID: 39842685 DOI: 10.1016/j.ijpara.2025.01.008] [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: 07/30/2024] [Revised: 10/21/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
The helminth Trichinella spiralis, through its excretory-secretory (ES L1) products, induces immune regulatory mechanisms that modulate the host's immune response not only to itself, but also to bystander antigens, foreign or self in origin, which can result in the alleviation of inflammatory diseases. Under the influence of ES L1, dendritic cells (DCs) acquire a tolerogenic phenotype and the capacity to induce Th2 and regulatory responses. Since ES L1 products represent a complex mixture of proteins and extracellular vesicles (TsEVs) the aim of this study was to investigate the impact of TsEVs, isolated from ES L1 products, on phenotypic and functional characteristics of DCs and to elucidate whether TsEVs could reproduce the immunomodulatory effects of the complete ES L1 product. Monocyte-derived DCs treated with TsEVs acquired semi-matured phenotypes, characterized by low expression of human leukocyte antigen - DR isotype (HLA-DR), cluster of differentiation (CD) 86 (CD86), and CD40, moderate expression of CD83 and C-C chemokine receptor type 7 (CCR7), and increased expression of tolerogenic markers indoleamine 2,3-dioxygenase 1 (IDO-1) and immunoglobulin-like transcript 3 (ILT3), together with the unchanged production of IL-12 and IL-23, and elevated production of IL-10 and transforming growth factor (TGF)-β, compared with controls. Gene expression analysis of TsEV-treated DCs revealed elevated levels of mTOR, Ahr, NF-κB2, RelB, SOCS1 and SOCS3, which participate in signaling pathways involved in DC maturation and the subsequent regulation of release of both anti-inflammatory and pro-inflammatory cytokines. TsEVs promoted the capacity of DCs to drive polarization of Th2 and anti-inflammatory responses, and impaired their capacity to induce Th1/Th17 polarization. Moreover, TsEV-treated DCs possessed a high capacity to induce conventional FoxP3 + regulatory T cells, as well as unconventional T regulatory (Tr1) cells. Tolerogenic properties of TsEV-treated DCs were retained even after challenge with a pro-inflammatory stimulus. These findings highlight the potential of TsEVs to induce immune tolerance, suggesting their potential use as therapeutics for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Sofija Glamočlija
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Ljiljana Sabljić
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering IMGGE University of Belgrade Republic of Serbia
| | - Nataša Radulović
- Institute for Biological Research "Siniša Stanković" University of Belgrade Republic of Serbia
| | - Alisa Gruden-Movsesijan
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Maja Kosanović
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia.
| |
Collapse
|
43
|
Manole S, Nguyen DH, Min JJ, Zhou S, Forbes N. Setting "cold" tumors on fire: Cancer therapy with live tumor-targeting bacteria. MED 2025; 6:100549. [PMID: 39689707 DOI: 10.1016/j.medj.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/18/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Immunotherapy with checkpoint blockade has shown remarkable efficacy in many patients with a variety of different types of cancer. However, the majority of patients with cancer have yet to benefit from this revolutionary therapy. Studies have shown that checkpoint blockade works best against immune-inflamed tumors characterized by the presence of tumor-infiltrating lymphocytes (TILs). In this review, we summarize studies using live tumor-targeting bacteria to treat cancer and describe various strategies to engineer the tumor-targeting bacteria for maximized immunoregulatory effects. We propose that tumor-localized infections by such engineered bacteria can create an immune microenvironment in favor of a more effective antitumor immunity with or without other therapies, such as immune checkpoint blockade (ICB). Finally, we will briefly outline some exemplary oncology clinical trials involving ICB plus live therapeutic bacteria, with a focus on their ability to modulate antitumor immune responses.
Collapse
Affiliation(s)
- Simin Manole
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeonnam 58128, South Korea.
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Neil Forbes
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA; Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA; Department of Microbiology, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
44
|
Kong Y, Li J, Zhao X, Wu Y, Chen L. CAR-T cell therapy: developments, challenges and expanded applications from cancer to autoimmunity. Front Immunol 2025; 15:1519671. [PMID: 39850899 PMCID: PMC11754230 DOI: 10.3389/fimmu.2024.1519671] [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: 10/30/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has rapidly emerged as a groundbreaking approach in cancer treatment, particularly for hematologic malignancies. However, the application of CAR-T cell therapy in solid tumors remains challenging. This review summarized the development of CAR-T technologies, emphasized the challenges and solutions in CAR-T cell therapy for solid tumors. Also, key innovations were discussed including specialized CAR-T, combination therapies and the novel use of CAR-Treg, CAR-NK and CAR-M cells. Besides, CAR-based cell therapy have extended its reach beyond oncology to autoimmune disorders. We reviewed preclinical experiments and clinical trials involving CAR-T, Car-Treg and CAAR-T cell therapies in various autoimmune diseases. By highlighting these cutting-edge developments, this review underscores the transformative potential of CAR technologies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yanwei Wu
- School of Medicine, Shanghai University, Shanghai, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
45
|
Lee KY, Mei Y, Liu H, Schwarz H. CD137-expressing regulatory T cells in cancer and autoimmune diseases. Mol Ther 2025; 33:51-70. [PMID: 39668561 PMCID: PMC11764688 DOI: 10.1016/j.ymthe.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining immune homeostasis, with critical roles in preventing aberrant immune responses that occur in autoimmune diseases and chronic inflammation. Conversely, the abundance of Tregs in cancer is associated with impaired anti-tumor immunity, and tumor immune evasion. Recent work demonstrates that CD137, a well-known costimulatory molecule for T cells, is highly expressed on Tregs in pathological conditions, while its expression is minimal or negligible on peripheral Tregs. The expression of CD137 marks Tregs with potent immunosuppressive phenotype that foster cancer progression and are protective against certain autoimmune diseases. Hence CD137 has emerged as a marker for Tregs. However, several important questions still remain regarding the expression and function of CD137 in Tregs. Here, we provide an overview of our current knowledge of Treg mechanisms of action, with a focus on the role of CD137 in modulating Treg activity. We also explore the implications of CD137+ Tregs in both cancer and autoimmune diseases, emphasizing the significance of targeting these cells for therapeutic intervention in these conditions.
Collapse
Affiliation(s)
- Kang Yi Lee
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Yu Mei
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Haiyan Liu
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore.
| | - Herbert Schwarz
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
| |
Collapse
|
46
|
Johnson AL, Khela HS, Korleski J, Sall S, Li Y, Zhou W, Smith-Connor K, Lopez-Bertoni H, Laterra J. TGFBR2 High mesenchymal glioma stem cells phenocopy regulatory T cells to suppress CD4+ and CD8+ T cell function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631757. [PMID: 39829747 PMCID: PMC11741370 DOI: 10.1101/2025.01.07.631757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Attempts to activate an anti-tumor immune response in glioblastoma (GBM) have been met with many challenges due to its inherently immunosuppressive tumor microenvironment. The degree and mechanisms by which molecularly and phenotypically diverse tumor-propagating glioma stem cells (GSCs) contribute to this state are poorly defined. In this study, our multifaceted approach combining bioinformatics analyses of clinical and experimental datasets, single-cell sequencing, and molecular and pharmacologic manipulation of patient-derived cells identified GSCs expressing immunosuppressive effectors mimicking regulatory T cells (Tregs). We show that this I mmunosuppressive T reg- L ike (ITL) GSC state is specific to the mesenchymal GSC subset and is associated with and driven specifically by TGF-β type II receptor (TGFBR2) in contrast to TGFBR1. Transgenic TGFBR2 expression in patient-derived GBM neurospheres promoted a mesenchymal transition and induced a 6-gene ITL signature consisting of CD274 (PD-L1), NT5E (CD73), ENTPD1 (CD39), LGALS1 (galectin-1), PDCD1LG2 (PD-L2), and TGFB1. This TGFBR2-driven ITL signature was identified in clinical GBM specimens, patient-derived GSCs and systemic mesenchymal malignancies. TGFBR2 High GSCs inhibited CD4+ and CD8+ T cell viability and their capacity to kill GBM cells, effects reversed by pharmacologic and shRNA-based TGFBR2 inhibition. Collectively, our data identify an immunosuppressive GSC state that is TGFBR2-dependent and susceptible to TGFBR2-targeted therapeutics.
Collapse
|
47
|
Zeng C, Xu C, Wei Y, Ma F, Wang Y. Training and experimental validation a novel anoikis- and epithelial‒mesenchymal transition-related signature for evaluating prognosis and predicting immunotherapy efficacy in gastric cancer. J Cancer 2025; 16:1078-1100. [PMID: 39895782 PMCID: PMC11786038 DOI: 10.7150/jca.106029] [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: 10/30/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025] Open
Abstract
Anoikis resistance and improper activation of epithelial‒mesenchymal transition (EMT) are critical factors in tumor metastasis and progression. Despite their interaction, the combined impact of anoikis and EMT on prognosis and immunotherapy in gastric cancer remains underexplored. In this study, we identified 354 anoikis- and EMT-related genes (AERGs) through Venn analysis and performed unsupervised clustering to classify gastric cancer patients into two molecular clusters: A and B. Molecular cluster A showed poor prognosis and an immunosuppressive tumor microenvironment, suggesting a "cold tumor" phenotype. Then, a novel AERG-related prognostic model comprising CD24, CRYAB, MMP11, MUC4, PRKAA2, SERPINE1, SKP2, and TP53 was constructed and validated, accurately predicting the 1-, 3-, and 5-year survival rates of gastric cancer patients. Multivariate analysis revealed that the AERG-related risk score was an independent prognostic factor (hazard ratio = 1.651, 95% confidence interval = 1.429-1.907, P<0.001). Further studies demonstrated that, compared to the high-risk group, the low-risk group exhibited higher CD8+ T cell infiltration, tumor mutational burden, immunophenoscores, and lower tumor immune dysfunction and exclusion scores, indicating potential sensitivity to immunotherapy. RT‒qPCR and immunohistochemical staining validated the expression levels of the model's molecular markers. Overall, our AERG-related model shows promise for predicting outcomes and guiding the selection of tailored and precise therapies for gastric cancer patients.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chang Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuhan Wei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yue Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu Province, 213000, China
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province, 213000, China
| |
Collapse
|
48
|
Bae AN, Lee H, Yang H, Mukherjee S, Im SS, Lee JH, Park JH. Enhancing Regulatory T cell function by mevalonate pathway inhibition prevents liver fibrosis. Biochem Biophys Res Commun 2025; 742:151094. [PMID: 39632293 DOI: 10.1016/j.bbrc.2024.151094] [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: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Liver fibrosis is a well-established risk factor for liver cancer development. Despite extensive mechanistic studies on liver fibrosis, the role of the immune cell network in fibrotic disease remains poorly understood. In this study, we demonstrate that regulatory T cells (Tregs) are involved in preventing liver fibrosis by regulating the mevalonate pathway. Blocking the mevalonate pathway increased the granzyme B secretion from Tregs, while restoring the pathway reduced it. Statin treatment, which inhibits the mevalonate pathway, alleviated liver fibrosis progression and enhanced the immunosuppressive function of Tregs in vivo. Mechanistically, mevalonate products, including geranylgeranyl pyrophosphate, inhibited the phosphorylation and activation of LKB1, that is a key regulator of Treg homeostasis. Furthermore, these products disrupted the interaction between LKB1 and cAMP-dependent protein kinase (PKA), leading to further reduction of LKB1 phosphorylation. These findings suggest that targeting LKB1 in Tregs through statin treatment prevents the progression of liver fibrosis, offering a promising and safe therapeutic strategy for liver disease and liver cancer.
Collapse
Affiliation(s)
- An-Na Bae
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Hajin Lee
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Huiseong Yang
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Sulagna Mukherjee
- Department of Physiology, School of Medicine, Keimyung University, Daegu, South Korea
| | - Seung-Soon Im
- Department of Physiology, School of Medicine, Keimyung University, Daegu, South Korea
| | - Jae-Ho Lee
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Jong Ho Park
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea.
| |
Collapse
|
49
|
Vijayakumar A, Vasudevan S, John S, Ozbun MA, Bartee E, Palanisamy V. Navigating a complex dance: the interplay between RNA-binding proteins and T cells in oral epithelial plasticity. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00054. [PMID: 39816132 PMCID: PMC11731067 DOI: 10.1097/in9.0000000000000054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
The oral epithelium, a dynamic interface constantly facing environmental challenges, relies on intricate molecular pathways to maintain its homeostasis. This comprehensive review delves into the nuanced interplay between T-lymphocytic cells (T cells) and RNA-binding proteins (RBPs) within the oral epithelium, elucidating their roles in orchestrating immune responses and influencing tissue plasticity. By synthesizing current knowledge, we aim to unravel the molecular intricacies that govern this interplay, with a focus on potential therapeutic implications for oral health and diseases. Understanding the regulatory networks shaped by T cells and RBPs in the oral epithelial microenvironment holds promise for innovative strategies in managing conditions associated with epithelial dysfunction.
Collapse
Affiliation(s)
- Anitha Vijayakumar
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Sekar Vasudevan
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Samu John
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Michelle A. Ozbun
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Eric Bartee
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Viswanathan Palanisamy
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
50
|
Elliott J, Koldej R, Khot A, Ritchie D. Graft-Versus-Host Disease Mouse Models: A Clinical-Translational Perspective. Methods Mol Biol 2025; 2907:1-56. [PMID: 40100591 DOI: 10.1007/978-1-0716-4430-0_1] [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: 03/20/2025]
Abstract
A variety of graft-versus-host disease (GVHD) models have been developed in mice for the purpose of allowing laboratory investigation of the pathobiology, prevention, and treatment of GVHD in humans. While such models are crucial in advancing our knowledge in this field, there are some key limitations that need to be considered when translating laboratory discoveries into the clinical context. This chapter will discuss current clinical practices in transplantation and GVHD and the relative strengths and weaknesses of mouse models that attempt to replicate these states.
Collapse
Affiliation(s)
- Jessica Elliott
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia.
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia.
| | - Rachel Koldej
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Amit Khot
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - David Ritchie
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|