1
|
Meden A, Claes S, Van Loy T, Zorman M, Proj M, Schols D, Gobec S, De Jonghe S. Structure-activity relationship study of navarixin analogues as dual CXCR2 and CCR7 antagonists. Bioorg Chem 2025; 159:108423. [PMID: 40179581 DOI: 10.1016/j.bioorg.2025.108423] [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: 11/14/2024] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Despite the promise of the human chemokine receptor 7 (CCR7) as drug target for the treatment of cancer metastasis and autoimmune diseases, there are no potent and selective CCR7 antagonists known in literature. In this work, a 1,2,5-thiadiazole 1,1-dioxide with low μM activity as a CXCR2 and CCR7 antagonist was selected as starting point for a structure-activity relationship study. The replacement of the central thiadiazole dioxide motif with squaramide led to low nanomolar CCR7 antagonism. Additional systematic structural variations afforded various squaramide analogues that displayed potent CCR7 antagonism in a calcium mobilization assay with IC50 values in the low nM range. Unfortunately, the same compounds also displayed potent CXCR2 antagonistic activity and should therefore be considered as dual CCR7/CXCR2 antagonists.
Collapse
Affiliation(s)
- Anže Meden
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | - Sandra Claes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium
| | - Maša Zorman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | - Matic Proj
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia.
| | - Steven De Jonghe
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium.
| |
Collapse
|
2
|
Lyu X, Tang Q, Zou Y, Liu X. Identification of a Gene Expression Signature to Predict the Risk of Abdominal Aortic Aneurysm in Psoriasis Patients. Clin Cosmet Investig Dermatol 2025; 18:961-978. [PMID: 40260094 PMCID: PMC12011042 DOI: 10.2147/ccid.s495890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/29/2025] [Indexed: 04/23/2025]
Abstract
Background Psoriasis is an immune-mediated, hereditary condition that presents itself in the skin or joints, or even both. Increasing evidence indicates that psoriasis is connected to an elevated risk of abdominal aortic aneurysm (AAA), owing to their shared inflammatory pathogenesis. Nevertheless, the interplay between psoriasis and AAA lacks sufficient documentation. Methods Through WGCNA and DEGs, psoriasis and AAA phenotype-related genes were identified. Identifying risk genes involved in both psoriasis and AAA involved generating candidate genes by finding the common intersection of hub genes, followed by using LASSO regression. Following this, a nomogram was created to forecast the development of psoriasis alongside AAA, and was then assessed through a ROC curve, DCA, calibration curve, and PR curve. Five algorithms, namely CIBERSORT, ssGSEA, ESTIMATE, MCPcounter, and QuanTIseq, were utilized to assess immune infiltration differences between high and low-risk groups. Simultaneously, we verified the differential gene expression in different tissues. Results A total of 1073 psoriasis hub genes and 128 AAA hub genes were generated. A Venn diagram revealed 20 candidate genes that were common to both hub genes of psoriasis and AAA. Of these, six genes (CCR7, CD3D, GBP5, HCLS1, IL7R, and ITGAL) were identified as risk genes. The gene signature generated by these genes demonstrated high accuracy in predicting psoriasis and AAA. Using five algorithms for immune infiltration analysis, an abundance of inflammatory cells was observed in high-risk subgroups. The above six genes were found to be highly expressed in both psoriasis tissue and abdominal aortic aneurysm tissue. Conclusions The study resulted in the identification of a novel gene signature, including six high-risk genes, that has enhanced our knowledge of the common causes and control mechanisms of psoriasis and AAA. These findings are anticipated to pave the way for promising therapeutic targets in mitigating the comorbidities of cardiovascular disease.
Collapse
Affiliation(s)
- Xintong Lyu
- Department of Dermatology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, People’s Republic of China
| | - Qingti Tang
- Department of Dermatology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, People’s Republic of China
| | - Yu Zou
- Department of Dermatology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, People’s Republic of China
| | - Xiaorong Liu
- Department of Dermatology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
3
|
Satofuka H, Suzuki H, Tanaka T, Ubukata R, Hirose M, Yamamoto H, Kaneko Y, Fujisawa S, Li G, Kaneko MK, Kato Y. A novel anti-mouse CCR7 monoclonal antibody, C 7Mab-7, demonstrates high sensitivity in flow cytometry, western blot, and immunohistochemistry. Biochem Biophys Rep 2025; 41:101948. [PMID: 40028039 PMCID: PMC11870228 DOI: 10.1016/j.bbrep.2025.101948] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
C-C chemokine receptor type 7 (CCR7) is a member of the G protein-coupled receptor family and functions as a lymph node-homing receptor for immune cells. Upon ligand binding, CCR7 promotes the migration of immune cells to secondary lymphoid organs. In cancers, CCR7 has been revealed as a critical molecule in lymph node metastasis. Consequently, anti-CCR7 monoclonal antibodies (mAbs) have been developed as cancer therapeutic agents. In this study, we established an anti-mouse CCR7 (mCCR7) mAb, C7Mab-7 (rat IgG1, kappa) using the Cell-Based Immunization and Screening (CBIS) method. C7Mab-7 demonstrated high sensitivity in flow cytometry. The dissociation constant (K D) value of C7Mab-7 was determined to be 2.5 × 10⁻⁹ M for mCCR7-overexpressed Chinese hamster ovary-K1 (CHO/mCCR7) cells. Furthermore, C7Mab-7 detected mCCR7 with high sensitivity in western blot and immunohistochemistry. C7Mab-7, developed by the CBIS method, accelerates the development of CCR7-targeted antibody therapies and cancer diagnostics.
Collapse
Affiliation(s)
- Hiroyuki Satofuka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Rena Ubukata
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Miu Hirose
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Haruto Yamamoto
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yu Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Shiori Fujisawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
4
|
Klauer LK, Rejeski HA, Ugur S, Rackl E, Abdulmajid J, Fischer Z, Pepeldjiyska E, Frischhut A, Schmieder N, Völker A, Rank A, Schmid C, Schmohl J, Amberger DC, Schmetzer HM. Leukemia-Derived Dendritic Cells Induce Anti-Leukemic Effects Ex Vivo in AML Independently of Patients' Clinical and Biological Features. Int J Mol Sci 2025; 26:1700. [PMID: 40004163 PMCID: PMC11855365 DOI: 10.3390/ijms26041700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
New therapies are highly needed to stabilize remission in patients with acute myeloid leukemia (AML). This study investigates the value of dendritic cells derived from leukemic blasts (DCleu) to enhance anti-leukemic immunity after T-cell-enriched mixed lymphocyte cultures (MLCs). We correlated induced anti-leukemic activity with patient data, including biological, clinical and prognostic factors. Additionally, we correlated the frequencies of DC/DCleu and leukemic-specific T cells with the achieved anti-leukemic activity after MLC. We show that mature DC/DCleu can be generated using the immunomodulating Kit-M, which contains granulocyte-macrophage colony-stimulating-factor (GM-CSF) and prostaglandin E1 (PGE1), without inducing blast proliferation from leukemic whole blood (WB) samples. Activated leukemia-specific immune and memory cells increased after MLC with Kit-M-pretreated WB, leading to improved blast lysis. Enhanced anti-leukemic activity positively correlated with the frequencies of generated DC/DCleu, proliferating leukemic-specific T cells and memory T cells, but not with leukemic blast counts, hemoglobin levels or platelet counts at diagnosis. No correlation was found between improved blast lysis and patients' prognostic data, including age, gender, ELN risk groups, disease stage and response to induction chemotherapy. These findings underscore the potential of DC/DCleu to evoke robust immune responses and potential immunological memory against AML. Overall, this innovative approach could pave the way for the development of improved immunotherapeutic strategies that function in vivo.
Collapse
Affiliation(s)
- Lara Kristina Klauer
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Hazal Aslan Rejeski
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Selda Ugur
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Elias Rackl
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Joudi Abdulmajid
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
- Faculty of Biology, University Bielefeld, 33615 Bielefeld, Germany
| | - Zuzanna Fischer
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Elena Pepeldjiyska
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Annalena Frischhut
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Nicolas Schmieder
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Antje Völker
- Department of Statistics, Ludwig-Maximilian-University Munich, 80539 Munich, Germany
| | - Andreas Rank
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
- Department of Haematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Christoph Schmid
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
- Department of Haematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Jörg Schmohl
- Department of Haematology and Oncology, Diakonie-Klinikum, 70176 Stuttgart, Germany
| | - Daniel Christoph Amberger
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- First Department of Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Helga Maria Schmetzer
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| |
Collapse
|
5
|
Han R, Luo Y, Gao J, Zhou H, Wang Y, Chen J, Zheng G, Ling C. HDAC3: A Multifaceted Modulator in Immunotherapy Sensitization. Vaccines (Basel) 2025; 13:182. [PMID: 40006729 PMCID: PMC11860249 DOI: 10.3390/vaccines13020182] [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: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Histone deacetylase 3 (HDAC3) has emerged as a critical epigenetic regulator in tumor progression and immune modulation, positioning it as a promising target for enhancing cancer immunotherapy. This work comprehensively explores HDAC3's multifaceted roles, focusing on its regulation of key immune-modulatory pathways such as cGAS-STING, ferroptosis, and the Nrf2/HO-1 axis. These pathways are central to tumor immune evasion, antigen presentation, and immune cell activation. Additionally, the distinct effects of HDAC3 on various immune cell types-including its role in enhancing T cell activation, restoring NK cell cytotoxicity, promoting dendritic cell maturation, and modulating macrophage polarization-are thoroughly examined. These findings underscore HDAC3's capacity to reshape the tumor immune microenvironment, converting immunologically "cold tumors" into "hot tumors" and thereby increasing their responsiveness to immunotherapy. The therapeutic potential of HDAC3 inhibitors is highlighted, both as standalone agents and in combination with immune checkpoint inhibitors, to overcome resistance and improve treatment efficacy. Innovative strategies, such as the development of selective HDAC3 inhibitors, advanced nano-delivery systems, and integration with photodynamic or photothermal therapies, are proposed to enhance treatment precision and minimize toxicity. By addressing challenges such as toxicity, patient heterogeneity, and resistance mechanisms, this study provides a forward-looking perspective on the clinical application of HDAC3 inhibitors. It highlights its significant potential in personalized cancer immunotherapy, paving the way for more effective treatments and improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Rui Han
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Yujun Luo
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jingdong Gao
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine Suzhou, Suzhou 215009, China
| | - Huiling Zhou
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Yuqian Wang
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jiaojiao Chen
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Guoyin Zheng
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Changquan Ling
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
6
|
Sabljić L, Radulović N, Đokić J, Stojanovic DB, Radojević D, Glamočlija S, Dinić M, Golić N, Vasilev S, Uskoković P, Sofronić-Milosavljević L, Gruden-Movsesijan A, Tomić S. Biodegradable Electrospun PLGA Nanofibers-Encapsulated Trichinella Spiralis Antigens Protect from Relapsing Experimental Autoimmune Encephalomyelitis and Related Gut Microbiota Dysbiosis. Int J Nanomedicine 2025; 20:1921-1948. [PMID: 39963417 PMCID: PMC11830953 DOI: 10.2147/ijn.s499161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
Purpose Trichinella spiralis has evolved complex immunomodulatory mechanisms mediated by excretory-secretory products (ESL1) that enable its survival in the host. Consequently, ESL1 antigens display excellent potential for treating autoimmune diseases such as multiple sclerosis (MS). However, whether timely controlled delivery of ESL1 antigens in vivo, as in natural infections, could enhance its therapeutic potential for MS is still unknown. Methods To test this, we encapsulated ESL1 antigens into biodegradable poly (lactide-co-glycolic) acid (PLGA) nanofibers by emulsion electrospinning as a delivery system and assessed their release dynamics in vitro, and in an animal MS model, experimental autoimmune encephalomyelitis (EAE), induced 7 days after PLGA/ESL1 subcutaneous implantation. PLGA/ESL1 effects on EAE symptoms were monitored along with multiple immune cell subsets in target organs at the peak and recovery of EAE. Gut barrier function and microbiota composition were analyzed using qPCR, 16S rRNA sequencing, and metabolomic analyses. Results ESL1 antigens, released from PLGA and drained via myeloid antigen-presenting cells through lymph nodes, protected the animals from developing EAE symptoms. These effects correlated with reduced activation of myeloid cells, increased IL-10 expression, and reduced accumulation of proinflammatory natural killer (NK) cells, T helper (Th)1 and Th17 cells in the spleen and central nervous system (CNS). Additionally, CD4+CD25hiFoxP3+ regulatory T cells and IL-10-producing B cells were expanded in PLGA/ESL1-treated animals, compared to control animals. The migration of ESL1 to the guts correlated with locally reduced inflammation and gut barrier damage. Additionally, PLGA/ESL1-treated animals displayed an unaltered microbiota characterized only by a more pronounced protective mevalonate pathway and expanded short-chain fatty acid-producing bacteria, which are known to suppress inflammation. Conclusion The delivery of T. spiralis ESL1 antigens via biodegradable electrospun PLGA nanofiber implants efficiently protected the animals from developing EAE by inducing a beneficial immune response in the spleen, gut, and CNS. This platform provides excellent grounds for further development of novel MS therapies.
Collapse
Affiliation(s)
- Ljiljana Sabljić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Nataša Radulović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Dusica B Stojanovic
- Faculty of Metallurgy and Technology, University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Sofija Glamočlija
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Miroslav Dinić
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Nataša Golić
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Saša Vasilev
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Petar Uskoković
- Faculty of Metallurgy and Technology, University of Belgrade, Belgrade, Serbia
| | | | | | - Sergej Tomić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
7
|
Lunavat TR, Nieland L, van de Looij SM, de Reus AJEM, Couturier CP, Farran CAE, Miller TE, Lill JK, Schübel M, Xiao T, Ianni ED, Woods EC, Sun Y, Rufino-Ramos D, van Solinge TS, Mahjoum S, Grandell E, Li M, Mangena V, Dunn GP, Jenkins RW, Mempel TR, Breakefield XO, Breyne K. Intratumoral gene delivery of 4-1BBL boosts IL-12-triggered anti-glioblastoma immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636330. [PMID: 39975249 PMCID: PMC11838556 DOI: 10.1101/2025.02.03.636330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The standard of care in high-grade gliomas has remained unchanged in the past 20 years. Efforts to replicate effective immunotherapies in non-cranial tumors have led to only modest therapeutical improvements in glioblastoma (GB). Here, we demonstrate that intratumoral administration of recombinant interleukin-12 (rIL-12) promotes local cytotoxic CD8 POS T cell accumulation and conversion into an effector-like state, resulting in a dose-dependent survival benefit in preclinical GB mouse models. This tumor-reactive CD8 T cell response is further supported by intratumoral rIL-12-sensing dendritic cells (DCs) and is accompanied by the co-stimulatory receptor 4-1BB expression on both cell types. Given that DCs and CD8 POS T cells are functionally suppressed in the tumor microenvironments of de novo and recurrent glioma patients, we tested whether anti-tumor response at the rIL-12-inflamed tumor site could be enhanced with 4-1BBL, the ligand of 4-1BB. 4-1BBL was delivered using an adeno-associated virus (AAV) vector targeting GFAP-expressing cells and resulted in prolonged survival of rIL-12 treated GB-bearing mice. This study establishes that tumor antigen-specific CD8 T cell activity can be directed using an AAV-vector-mediated gene therapy approach, effectively enhancing anti-GB immunity.
Collapse
|
8
|
Waghmare PS, Kaushik D, Oz E, Proestos C, Oz F, Kumar M. Unraveling the hormonal approaches for the treatment of rheumatoid arthritis and its complementary interventions. Inflammopharmacology 2025; 33:443-460. [PMID: 39754003 DOI: 10.1007/s10787-024-01633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 02/22/2025]
Abstract
Rheumatoid Arthritis (RA) is an autoimmune, chronic, systemic inflammatory disease that causes redness, swelling, stiffness, and joint pain. It is a long-lasting disease that can have a widespread impact on the body, often affecting the hands, feet, and wrists. The immune cells, such as dendritic cells, T cells, B cells, macrophages, and neutrophils, play a significant role in bone degradation and inflammation. Several cytokines, including TNF-α and IL-17A, play a significant role in causing bone erosion, cartilage deterioration, and joint inflammation. Progesterone and estrogen have a crucial impact on the pathophysiology of RA, influencing the immune system. Research has demonstrated that hormone replacement therapy (HRT) can effectively reduce inflammation, improve disease activity, enhance joint health, alleviate pain, and promote bone strength. Treatments such as tamoxifen and raloxifene, known as selective estrogen receptor modulators (SERMs), are effective against chronic inflammatory illnesses like RA. The treatment with Gonadotropin-releasing hormone (GnRH) has an impact on the hypothalamic-pituitary-gonadal axis, which in turn affects the activity of RA illness. These alternative treatments hold promise in enhancing well-being and alleviating joint pain for individuals with RA.
Collapse
Affiliation(s)
- Priya Sharad Waghmare
- Department of Food Technology and Nutrition, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Deepika Kaushik
- Department of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, HP, 173229, India.
| | - Emel Oz
- Department of Food Engineering, Faculty of Agriculture, Ataturk University, Erzurum, 25240, Türkiye
| | - Charalampos Proestos
- Laboratory of Food Chemistry, Department of Chemistry, School of Sciences, National and Kapodistrian University of Athens Zografou, 157 84, Athens, Greece
| | - Fatih Oz
- Department of Food Engineering, Faculty of Agriculture, Ataturk University, Erzurum, 25240, Türkiye
| | - Mukul Kumar
- Department of Food Technology and Nutrition, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
9
|
Montagne JM, Mitchell JT, Tandurella JA, Christenson ES, Danilova LV, Deshpande A, Loth M, Sidiropoulos DN, Davis-Marcisak E, Bergman DR, Zhu Q, Wang H, Kagohara LT, Engle LL, Green BF, Favorov AV, Ho WJ, Lim SJ, Zhang R, Li P, Gai J, Mo G, Mitchell S, Wang R, Vaghasia A, Hou W, Xu Y, Zimmerman JW, Elisseeff JH, Yegnasubramanian S, Anders RA, Jaffee EM, Zheng L, Fertig EJ. CD137 agonism enhances anti-PD1 induced activation of expanded CD8 + T cell clones in a neoadjuvant pancreatic cancer clinical trial. iScience 2025; 28:111569. [PMID: 39811671 PMCID: PMC11730579 DOI: 10.1016/j.isci.2024.111569] [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: 07/26/2024] [Revised: 09/05/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Successful pancreatic ductal adenocarcinoma (PDAC) immunotherapy requires therapeutic combinations that induce quality T cells. Tumor microenvironment (TME) analysis following therapeutic interventions can identify response mechanisms, informing design of effective combinations. We provide a reference single-cell dataset from tumor-infiltrating leukocytes (TILs) from a human neoadjuvant clinical trial comparing the granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting allogeneic PDAC vaccine GVAX alone, in combination with anti-PD1 or with both anti-PD1 and CD137 agonist. Treatment with GVAX and anti-PD-1 led to increased CD8+ T cell activation and expression of cytoskeletal and extracellular matrix (ECM)-interacting components. Addition of CD137 agonist increased abundance of clonally expanded CD8+ T cells and increased immunosuppressive TREM2 signaling in tumor associated macrophages (TAMs), identified by comparison of ligand-receptor networks, corresponding to changes in metabolism and ECM interactions. These findings associate therapy with GVAX, anti-PD1, and CD137 agonist with enhanced CD8+ T cell function while inducing alternative immunosuppressive pathways in patients with PDAC.
Collapse
Affiliation(s)
- Janelle M. Montagne
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob T. Mitchell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore MD, USA
| | - Joseph A. Tandurella
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric S. Christenson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ludmila V. Danilova
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atul Deshpande
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Melanie Loth
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitrios N. Sidiropoulos
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily Davis-Marcisak
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore MD, USA
| | - Daniel R. Bergman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qingfeng Zhu
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luciane T. Kagohara
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Logan L. Engle
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin F. Green
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander V. Favorov
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Systems Biology and Computational Genetics, Vavilov Institute of General Genetics, Moscow, RF, Russia
| | - Won Jin Ho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Su Jin Lim
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rui Zhang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pan Li
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica Gai
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guanglan Mo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Mitchell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rulin Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ajay Vaghasia
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenpin Hou
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yao Xu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- InHealth Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert A. Anders
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elana J. Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Center for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA
| |
Collapse
|
10
|
Mundry CS, Triplett AA, Shah OS, Chaitankar V, McAndrews KL, Ly QP, Cox JL, Eberle KC, Mehla K, Swanson BJ, Lazenby A, Klute KA, Grandgenett PM, Hollingsworth MA. Single-cell RNA-sequencing of human spleens reveals an IDO-1 + tolerogenic dendritic cell subset in pancreatic cancer patients that is absent in normal individuals. Cancer Lett 2024; 607:217321. [PMID: 39547331 DOI: 10.1016/j.canlet.2024.217321] [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: 09/20/2024] [Revised: 10/18/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Local and systemic immunosuppression are prominent features of pancreatic cancer, rendering anti-tumor effector cells inactive and immunotherapeutic approaches ineffective. The spleen, an understudied point of antigen-presentation and T cell priming in humans, holds particular importance in pancreatic cancer due to its proximity to the developing tumor. As main effectors of antigen presentation, dendritic cells display antigens to lymphocytes, thereby bridging the innate and adaptive immune response. While tumor-infiltrating anti-inflammatory dendritic cells have been described, splenic dendritic cells have historically just been considered to stimulate the anti-tumor immune response. Here, we describe, for the first time, the presence of an immunosuppressive, tolerogenic IDO1+ dendritic cell subset in the spleens of pancreatic cancer patients that likely contributes to systemic immunosuppression that is associated with pancreatic ductal adenocarcinoma. Network analysis of scRNA seq data reveals extensive communication networks between the identified tolerogenic DC cluster and numerous immune cell populations in the spleen. Interactions with innate and adaptive immune cells suggest a broad influence on leukocyte trafficking and immune regulation within the spleen microenvironment. The identification of signaling pathways involving AHR and IDO-1, CCL19, NECTIN2, CLEC2D, and others elucidates potential mechanisms underlying the immunosuppressive functions of this cell type.
Collapse
Affiliation(s)
- Clara S Mundry
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aleata A Triplett
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Osama Shiraz Shah
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vijender Chaitankar
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kyle L McAndrews
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Quan P Ly
- Department of Surgery, Division of Surgical Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kirsten C Eberle
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kamiya Mehla
- Department of Oncology Science, OU Health Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Benjamin J Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Audrey Lazenby
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kelsey A Klute
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul M Grandgenett
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
11
|
Vida H, Sahar M, Nikdouz A, Arezoo H. Chemokines in neurodegenerative diseases. Immunol Cell Biol 2024. [PMID: 39723647 DOI: 10.1111/imcb.12843] [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: 12/04/2023] [Revised: 06/09/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Neurodegeneration and neuroinflammation disorders are mainly the result of the deposition of various proteins, such as α-synuclein, amyloid-β and prions, which lead to the initiation and activation of inflammatory responses. Different chemokines are involved in the infiltration and movement of inflammatory leukocytes into the central nervous system (CNS) that express chemokine receptors. Dysregulation of several members of chemokines has been shown in the CNS, cerebrospinal fluid and peripheral blood of patients who have neurodegenerative disorders. Upon infiltration of various cells, they produce many inflammatory mediators such as cytokines. Besides them, some CNS-resident cells, such as neurons and astrocytes, are also involved in the pathogenesis of neurodegeneration by producing chemokines. In this review, we summarize the role of chemokines and their related receptors in the pathogenesis of neurodegeneration and neuroinflammation disorders, including multiple sclerosis, Parkinson's disease and Alzheimer's disease. Therapeutic strategies targeting chemokines or their related receptors are also discussed in this article.
Collapse
Affiliation(s)
- Hashemi Vida
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mehranfar Sahar
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Urmia University of Medical Sciences, Urmia, Iran
| | - Amin Nikdouz
- Department of Translational Medicine, Universita degli Studi del Piemonte Orientale Amedeo Avogadro, Vercelli, Italy
| | - Hosseini Arezoo
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
12
|
Liu M, Jiang Z, Liu M, Ni H, Li Y, Fang J, Du Q, Dong Y. SLAMF1 as a novel molecule mediating the causal association between rheumatoid arthritis and interstitial lung disease: A Mendelian randomization study combined with transcriptomics and in vivo validation. Int Immunopharmacol 2024; 142:113082. [PMID: 39260308 DOI: 10.1016/j.intimp.2024.113082] [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: 08/13/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is a common complication of rheumatoid arthritis (RA) that result in significant morbidity and mortality. Understanding the molecular mechanisms underlying RA-ILD is crucial for effective prevention. This study aims to identify the specific molecule that mediate the causal association between RA and ILD, as well as to explore its potential mechanisms in the pathogenesis of RA-ILD. METHODS Using two-sample Mendelian randomization (MR) analyses, we investigated the causal relationship among 16,987 blood genes, RA and ILD. Subsequently, a two-step MR technique was employed to identify significant genes that mediate the association between RA and ILD, and to quantify their proportion of mediation effect. To validate the genes as mediators, the replication MR analysis was conducted and the in vivo experiment was performed using an established animal model of RA-ILD. Furthermore, integrated bioinformatic analyses were conducted to elucidate the specific biological functions of the determined mediator in pathogenesis of RA-ILD. RESULTS Nine genes, namely MAPK8IP2, TAF11, SLAMF1, DAB2IP, GLUL, SLC4A10, PRSS35, NFX1, and PLK3, were identified as mediators. Among them, SLAMF1 was validated as the most significant mediator, accounting for 4.693% of the mediating effect on the causal relationship between RA and ILD. Upregulated mRNA expression of SLAMF1 was observed in the animal model of RA-ILD compared to controls. Bioinformatic analyses revealed that SLAMF1 was overexpressed in patients with lung fibrosis and correlated with a poor prognosis. Specifically, SLAMF1 was found to be predominantly overexpressed in T cells in lung tissues of patients with lung fibrosis. Additionally, the functional role of SLAMF1 was associated with multiple immune cell infiltrations and the biological process of extracellular matrix synthesis in pulmonary tissues from patients with lung fibrosis. CONCLUSION SLAMF1 may play a crucial role as a molecular mediator in the causal association between RA and ILD, and participate in multiple mechanisms underlying the pathogenesis of RA-ILD. This research provides insights into how the development of RA influences the risk of ILD and offers potential interventional targets against RA-ILD.
Collapse
Affiliation(s)
- Muqiu Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Zhihao Jiang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Min Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Haojie Ni
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Yanwu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China.
| | - Qun Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China.
| | - Yan Dong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China.
| |
Collapse
|
13
|
Di S, Li Y, Fu X, Xie C, Jiang Y, Liang W, Wang Z, Wang C, Wang L, Zhu Y, Zhang J. Transcriptomic Analysis Reveals the Potential Mechanism of Cardamine circaeoides Hook.f. & Thomson in Lowering Serum Uric Acid by Reducing Inflammatory State Through CCR7 Target. Int J Mol Sci 2024; 25:12967. [PMID: 39684678 DOI: 10.3390/ijms252312967] [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: 10/13/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Cardamine circaeoides Hook.f. & Thomson (CC) is a traditional medicinal herb with multiple biological activities. In previous studies, we have identified its serum uric acid (SUA) lowering effects and speculated that Cardamine circaeoides water extract (CCE) may exert anti-hyperuricemia effects related to its anti-inflammatory activity. This study aims to further investigate the molecular mechanism underlying these effects at the mRNA level through transcriptomic analysis, quantitative reverse transcription polymerase chain reaction (RT-qPCR), molecular docking, and Western blotting. CCE effectively reduced SUA and improved renal function in a dose-dependent manner in hyperuricemia rats. Cytokine-cytokine receptor interaction pathway was significantly altered by CCE. An additional study identified a number of genes (IL27, Inhbe, CCR7, CXCR3, IL12RB1, CXCR5, Mstn, and GDF5) as regulators of the inflammatory response. Meanwhile, three key targets (IL27, Inhbe, and CCR7) were found to be significantly expressed at the mRNA level and have strong binding affinity with 22 components, among which Kaempferol 3-sophoroside 7-glucoside, Kaempferol-3-O-sophoroside, and Quercetin 3-sophoroside 7-glucoside have strong binding activities. Following this, Western blotting showed a significant increase in CCR7 expression. Our findings indicated that CCE regulated the cytokine-cytokine receptor interaction pathway through CCR7 to reduce the inflammatory state and exert an SUA-lowering effect.
Collapse
Affiliation(s)
- Songrui Di
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yipeng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiangchen Fu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chenyu Xie
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanxin Jiang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Weican Liang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zixu Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yingli Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
14
|
da Silva CP, Silva MDS, Santana HM, Paloschi MV, Ferreira E Ferreira AA, Brilhante LMV, Cruz LF, Serrath SN, Eulálio MDMC, Setúbal SDS, Vallochi AL, Nery NM, Zuliani JP. Bothrops atrox snake venom decreased MHC-II and CD86 expression in bone marrow-derived dendritic cells. Acta Trop 2024; 260:107426. [PMID: 39393479 DOI: 10.1016/j.actatropica.2024.107426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/13/2024]
Abstract
The effect of Bothrops atrox venom (BaV) on the maturation of bone marrow-derived dendritic cells (BMDCs) from mice was investigated, with a focus on selected cell markers, TAP1 expression, and the release of pro-inflammatory cytokines during this process. The objective was to evaluate BaV's impact on dendritic cell (DC) function, as DCs are pivotal in antigen presentation and responsible for initiating the immune response mediated by naïve T cells, as well as regulating the immune system. Bone marrow cells were obtained from Swiss mice, and hematopoietic precursors were differentiated into BMDCs using GM-CSF and IL-4. On the 7th day, BaV and LPS were introduced into the culture, and the cells were analyzed 24 h later. BaV's ability to stimulate BMDC maturation was assessed through the analysis of surface marker expression. The findings demonstrated that BMDCs are highly influenced by culture environment factors, such as GM-CSF and IL-4, and are sensitive to additional stimuli like LPS and BaV. Mature DCs exhibited elevated levels of critical markers for T cell activation, such as MHC-II, CD80, and CD86, displaying specific phenotypic characteristics. However, the observed reduction in MHC-II and CD86 expression following BaV exposure suggests a substantial impact on the immunological activation capacity of these cells, potentially interfering with the adaptive immune response. Furthermore, the selective release of cytokines, such as IL-6, but not TNF-α or IL-1β, indicates differentiated modulation of inflammatory responses by DCs under various stimulation conditions.
Collapse
Affiliation(s)
- Carolina P da Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Milena D S Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Hallison M Santana
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Mauro V Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Alex A Ferreira E Ferreira
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Lívia M V Brilhante
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Larissa F Cruz
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Suzanne N Serrath
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Micaela de M C Eulálio
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Sulamita da S Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Adriana L Vallochi
- Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Neriane M Nery
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil.
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
15
|
Maeda S, Kai K, Kawasaki K, Tanaka T, Ide T, Noshiro H. Analysis of CD1a-Positive Monocyte-Derived Cells in the Regional Lymph Nodes of Patients with Gallbladder Cancer. Int J Mol Sci 2024; 25:12763. [PMID: 39684473 DOI: 10.3390/ijms252312763] [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: 11/07/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Dendritic cells (DCs) are known to be major antigen-presenting cells, and lymph nodes (LNs) play an important role in DC-mediated immune response. CD1a is known as a marker of monocyte-derived DCs. The present study focused on the infiltration of CD1a-positive DCs (CD1a-DCs) into regional LNs in 70 cases of gallbladder cancer (GBC). After univariate analyses, the results showed that LN infiltration by CD1a-DCs was associated with unfavorable clinical outcomes in patients with GBC, with all cases categorized in the CD1a-DCs high group had nodal metastasis. LN infiltration by CD1a-DCs was not an independent prognostic factor identified by multivariate analyses. After subgroup analyses of cases with LN metastasis (n = 32), no significant impacts of CD1a-DCs infiltration into metastatic LNs were observed. In contrast, CD1a-DCs infiltration into primary tumors had a significant impact on surgical outcomes. The results of strong confounding between CD1a-DCs and LN metastasis support the theory that CD1a-DCs are developed from monocytes at tumor sites. As the results of previous research focused on CD1a-DCs infiltration into regional LNs of other organs varied, the role and significance of CD1a-DCs infiltration in regional LNs may be different according to the tumor histology or its primary site. Thus, further studies are needed to clarify the role and significance of CD1a-DCs infiltration into regional LNs of solid cancers.
Collapse
Affiliation(s)
- Sachiko Maeda
- Department of Pathology & Microbiology, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Department of Surgery, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, Saga 849-8501, Japan
| | - Kanako Kawasaki
- Department of Pathology & Microbiology, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Tomokazu Tanaka
- Department of Surgery, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Takao Ide
- Department of Surgery, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Hirokazu Noshiro
- Department of Surgery, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| |
Collapse
|
16
|
Fan G, Gao R, Xie T, Li L, Tang L, Han X, Shi Y. DKK1+ tumor cells inhibited the infiltration of CCL19+ fibroblasts and plasma cells contributing to worse immunotherapy response in hepatocellular carcinoma. Cell Death Dis 2024; 15:797. [PMID: 39505867 PMCID: PMC11541906 DOI: 10.1038/s41419-024-07195-3] [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: 04/06/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
Intra-tumor immune infiltration plays a pivotal role in the interaction with tumor cells in hepatocellular carcinoma (HCC). However, its phenotype and related spatial structure remained elusive. To address these limitations, we conducted a comprehensive study combining spatial data (38,191 spots from eight samples) and single-cell data (56,022 cells from 20 samples). Our analysis revealed two distinct infiltration patterns: immune exclusion and immune activation. Plasma cells emerged as the primary cell type within intra-tumor immune clusters. Notably, we observed the co-location of CCL19+ fibroblasts with plasma cells, which secrete chemokines and promote T-cell activation and leukocyte migration. Conversely, in immune-exclusion samples, this co-location was primarily observed in the adjacent normal area. This co-localization correlated with T cell infiltration and the formation of tertiary lymphoid structures, validated by multiplex immunofluorescence conducted on twenty HCC samples. Both CCL19+ fibroblasts and plasma cells were associated with favorable survival outcomes. In an immunotherapy cohort, HCC patients who responded favorably exhibited higher infiltration of CCL19+ fibroblasts and plasma cells. Additionally, we observed the accumulation of DKK1+ tumor cells within the tumor area in immune-exclusion samples, particularly at the tumor boundary, which inhibited the infiltration of CCL19+ fibroblasts and plasma cells into the tumor area. Furthermore, in immune-exclusion samples, the SPP1 signaling pathway demonstrated the highest activity in communication between tumor and immune clusters, and CCL19-CCR7 played a pivotal role in the self-communication of immune clusters. This study elucidates immune exclusion and immune activation patterns in HCC and identifies relevant factors contributing to immune resistance.
Collapse
Affiliation(s)
- Guangyu Fan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Ruyun Gao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| |
Collapse
|
17
|
Wang C, Macintyre AN, Oguin TH, McCarthy KR, Moody MA, Yuan F. Spatiotemporal control of immune responses with nucleic acid cocktail vaccine. ADVANCED THERAPEUTICS 2024; 7:2400263. [PMID: 40248361 PMCID: PMC12002596 DOI: 10.1002/adtp.202400263] [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: 06/18/2024] [Indexed: 04/19/2025]
Abstract
Nucleic acid vaccines play important roles in prevention and treatment of diseases. However, limited immunogenicity remains a major obstacle for DNA vaccine applications in the clinic. To address the issue, the present study investigates a cocktail approach to DNA vaccination. In this proof-of-the-concept study, the cocktail consists of two DNAs encoding viral hemagglutinin (HA) and granulocyte-macrophage colony stimulatory factor (GM-CSF), respectively. Data from the study demonstrate that recruitment and activation of antigen-presenting cells (APCs) can be substantially improved by spatiotemporal regulation of GM-CSF and HA expressions at the site of vaccination. The types of recruited APCs and their phenotypes are also controllable by adjusting the cocktail compositions. Compared to mono-ingredient vaccine, the optimized cocktail vaccine is able to enhance the anti-viral humoral and T cell immune responses. No significant systemic inflammation has been detected after either prime or boost immunization using the cocktail vaccine. Data in the study suggest that the DNA cocktail is a safe, effective, and controllable platform for improving vaccine efficacy.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Andrew N. Macintyre
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27708, USA
| | - Thomas H. Oguin
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
| | - Kevin R. McCarthy
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA 15261, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
18
|
Liu H, Lu Y, Zong J, Zhang B, Li X, Qi H, Yu T, Li Y. Engineering dendritic cell biomimetic membrane as a delivery system for tumor targeted therapy. J Nanobiotechnology 2024; 22:663. [PMID: 39465376 PMCID: PMC11520105 DOI: 10.1186/s12951-024-02913-7] [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: 05/17/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024] Open
Abstract
Targeted immunotherapies make substantial strides in clinical cancer care due to their ability to counteract the tumor's capacity to suppress immune responses. Advances in biomimetic technology with minimally immunogenic and highly targeted, are addressing issues of targeted drug delivery and disrupting the tumor's immunosuppressive environment to trigger immune activation. Specifically, the use of dendritic cell (DC) membranes to coat nanoparticles ensures targeted delivery due to DC's unique ability to activate naive T cells, spotlighting their role in immunotherapy aimed at disrupting the tumor microenvironment. The potential of DC's biomimetic membrane to mediate immune activation and target tumors is gaining momentum, enhancing the effectiveness of cancer treatments in conjunction with other immune responses. This review delves into the methodologies behind crafting DC membranes and the fusion of dendritic and tumor cell membranes for encapsulating therapeutic nanoparticles. It explores their applications and recent advancements in combating cancer, offering an all-encompassing perspective on DC biomimetic nanosystems, immunotherapy driven by antigen presentation, and the collaborative efforts of drug delivery in chemotherapy and photodynamic therapies. Current evidence shows promise in augmenting combined therapeutic approaches for cancer treatment and holds translational potential for various cancer treatments in a clinical setting.
Collapse
Affiliation(s)
- Huiyang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Yiming Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China.
| |
Collapse
|
19
|
Pan L, Huang H, Zhang P, Li H, Lu L, Wei M, Zheng P, Wang Q, Guo J, Qin Y. Immunofluorescence-Verified Sphingolipid Signatures Indicate Improved Prognosis in Liver Cancer Patients. J Cancer 2024; 15:6239-6255. [PMID: 39513103 PMCID: PMC11540515 DOI: 10.7150/jca.101330] [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: 07/23/2024] [Accepted: 09/28/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy, with its pathogenesis involving a complex interplay of molecular mechanisms, including cell cycle dysregulation, evasion of apoptosis, enhanced angiogenesis, and aberrant immune responses. Precision medicine approaches that target specific molecular subtypes through multi-omics integration hold promise for improving patient survival. Among the various molecular players, sphingolipids have emerged as pivotal regulators of tumor growth and apoptosis, positioning them as key targets in the search for novel anticancer therapies. Methods: To identify critical genes involved in sphingolipid metabolism (SM), we employed the AUCell algorithm and correlation analysis in conjunction with scRNA-seq data. A robust prognostic risk model was developed using Cox proportional hazards and Lasso regression, and its predictive performance was validated using an independent cohort from the International Cancer Genome Consortium (ICGC). The model's evaluation also incorporated analyses of the tumor microenvironment (TME), immunotherapy responses, mutational landscape, and pathway enrichment across different risk strata. Finally, we conducted multiplex immunofluorescence assays to investigate the functional role of ZC3HAV1 in HCC. Results: Our analysis yielded a 9-gene signature risk model with strong prognostic capabilities, effectively stratifying HCC patients into high- and low-risk groups, with significant differences in survival outcomes. Notably, the model revealed distinct variations in the immune microenvironment and responsiveness to immunotherapy between the risk groups. Further experimental validation identified ZC3HAV1 as a key gene, with multiplex immunofluorescence suggesting its involvement in promoting malignant progression in HCC through modulation of the epithelial-mesenchymal transition (EMT). Conclusion: This sphingolipid metabolism-based prognostic model is not only predictive of survival in HCC but also indicative of immunotherapy efficacy in certain patient subsets. Our findings underscore the crucial role of sphingolipid metabolism in shaping the immune microenvironment, offering new avenues for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Lujuan Pan
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, Guangxi, China
| | - Huijuan Huang
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Hua Li
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Libai Lu
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, Guangxi, China
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Mingwei Wei
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Pin Zheng
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Junyu Guo
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Yueqiu Qin
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, Guangxi, China
| |
Collapse
|
20
|
Zhao L, Shireman J, Probelsky S, Rigg B, Wang X, Huff WX, Kwon JH, Dey M. CCL21 Induces Plasmacytoid Dendritic Cell Migration and Activation in a Mouse Model of Glioblastoma. Cancers (Basel) 2024; 16:3459. [PMID: 39456552 PMCID: PMC11506458 DOI: 10.3390/cancers16203459] [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: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that are traditionally divided into two distinct subsets: myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). pDCs are known for their ability to secrete large amounts of cytokine type I interferons (IFN- α). In our previous work, we have demonstrated that pDC infiltration promotes glioblastoma (GBM) tumor immunosuppression through decreased IFN-α secretion via TLR-9 signaling and increased suppressive function of regulatory T cells (Tregs) via increased IL-10 secretion, resulting in poor overall outcomes in mouse models of GBM. Further dissecting the overall mechanism of pDC-mediated GBM immunosuppression, in this study, we identified CCL21 as highly upregulated by multiple GBM cell lines, which recruit pDCs to tumor sites via CCL21-CCR7 signaling. Furthermore, pDCs are activated by CCL21 in the GBM microenvironment through intracellular signaling of β-arrestin and CIITA. Finally, we found that CCL21-treated pDCs directly suppress CD8+ T cell proliferation without affecting regulatory T cells (Tregs) differentiation, which is considered the canonical pathway of immunotolerant regulation. Taken together, our results show that pDCs play a multifaced role in GBM immunosuppression, and CCL21 could be a novel therapeutic target in GBM to overcome pDC-mediated immunosuppression.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Jack Shireman
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Samantha Probelsky
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Bailey Rigg
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Xiaohu Wang
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Wei X. Huff
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Jae H. Kwon
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Mahua Dey
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| |
Collapse
|
21
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [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: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
22
|
Zhang Q, Wang W, Shang S, Li X, Zhao T, Zhang P, Wu D, Zhou K, Lu X. Unveiling the immune-modulating power of THz-FEL irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 259:113017. [PMID: 39226855 DOI: 10.1016/j.jphotobiol.2024.113017] [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: 05/29/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
As terahertz (THz) technology advances, the interaction between THz radiation and the living body, particularly its effects on the immune system, has attracted extensive attention but remains poorly understood. This study firstly elucidated that exposure to 3 THz-FEL radiation markedly suppressed contact hypersensitivity reactions in mice induced by DNFB, as evidenced by a reduction in ear thickness and a discernible recovery in the Th1/Th2 cell balance. 3 THz irradiation led to cellular stress in the irradiated skin locale, increasing the levels of IL-4 and IL-10 and modulating the activity and migration of dendritic cells and mast cells. Furthermore, THz irradiation precipitated a rapid alteration in the skin lipidome, altering several categories of bioactive lipids. These findings offer new insights into the immunomodulatory effects of THz radiation on living organisms and the potential underlying mechanisms, with implications for the development of therapeutic approaches in managing skin allergic diseases.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Weijun Wang
- China Academy of Engineering Physics, Institute of Applied Electronics, Mianyang 621900, Sichuan, China; National Key Laboratory of Science and Technology on Advanced Laser and High Power Microwave, Mianyang 621900, Sichuan, China
| | - Sen Shang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xu Li
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Tingting Zhao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Peng Zhang
- China Academy of Engineering Physics, Institute of Applied Electronics, Mianyang 621900, Sichuan, China; National Key Laboratory of Science and Technology on Advanced Laser and High Power Microwave, Mianyang 621900, Sichuan, China
| | - Dai Wu
- China Academy of Engineering Physics, Institute of Applied Electronics, Mianyang 621900, Sichuan, China; National Key Laboratory of Science and Technology on Advanced Laser and High Power Microwave, Mianyang 621900, Sichuan, China
| | - Kui Zhou
- China Academy of Engineering Physics, Institute of Applied Electronics, Mianyang 621900, Sichuan, China; National Key Laboratory of Science and Technology on Advanced Laser and High Power Microwave, Mianyang 621900, Sichuan, China.
| | - Xiaoyun Lu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
23
|
Mao X, Shi M, Zhang B, Fu R, Cai M, Yu S, Lin K, Zhang C, Li D, Chen G, Luo W. Integration of single-cell and bulk RNA sequencing revealed immune heterogeneity and its association with disease activity in rheumatoid arthritis patients. Immunol Res 2024; 72:1120-1135. [PMID: 39009881 DOI: 10.1007/s12026-024-09513-5] [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: 05/22/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory, systemic autoimmune disease characterized by cartilage, bone damage, synovial inflammation, hyperplasia, autoantibody production, and systemic features. To obtain an overall profile of the immune environment in RA patients and its association with clinical features, we performed single-cell transcriptome and T-cell receptor sequencing of mononuclear cells from peripheral blood (PBMC) and synovial fluid (SF) from RA patients, integrated with two large cohorts with bulk RNA sequencing for further validation and investigation. Dendritic cells (DCs) exhibited relatively high functional heterogeneity and tissue specificity in relation to both antigen presentation and proinflammatory functions. Peripheral helper T cells (TPHs) are likely to originate from synovial tissue, undergo activation and exhaustion, and are subsequently released into the peripheral blood. Notably, among all immune cell types, TPHs were found to have the most intense associations with disease activity. In addition, CD8 effector T cells could be clustered into two groups with different cytokine expressions and play distinct roles in RA development. By integrating single-cell data with bulk sequencing from two large cohorts, we identified interactions among TPHs, CD8 cells, CD16 monocytes, and DCs that strongly contribute to the proinflammatory local environment in RA joints. Of note, the swollen 28-joint counts exhibited a more pronounced association with this immune environment compared to other disease activity indexes. The immune environment alternated significantly from PBMCs to SF, which indicated that a series of immune cells was involved in proinflammatory responses in the local joints of RA patients. By integrating single-cell data with two large cohorts, we have uncovered associations between specific immune cell populations and clinical features. This integration provides a rapid and precise methodology for assessing local immune activation, offering valuable insights into the pathophysiological mechanisms at play in RA.
Collapse
Affiliation(s)
- Xiaofan Mao
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Maohua Shi
- Department of Rheumatology, The First People's Hospital of Foshan, Foshan, China
| | - Beiying Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Rongdang Fu
- Department of Hepatology, The First People's Hospital of Foshan, Foshan, China
| | - Mengyun Cai
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Sifei Yu
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Kairong Lin
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Chuling Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Dingru Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Guoqiang Chen
- Department of Rheumatology, The First People's Hospital of Foshan, Foshan, China.
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China.
| |
Collapse
|
24
|
Wang J, Shao Y, Deng X, Du J. Causal Relationship Between Post-Traumatic Stress Disorder and Immune Cell Traits: A Mendelian Randomization Study. Brain Behav 2024; 14:e70073. [PMID: 39350630 PMCID: PMC11443039 DOI: 10.1002/brb3.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/29/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
INTRODUCTION Post-traumatic stress disorder (PTSD) is a debilitating psychological disorder that occurs after exposure to catastrophic-level experiences. Although alterations in immune function have been identified in individuals with PTSD, the causal relationship between the two remains unclear. METHODS To investigate the causal relationship between PTSD and immune function, we conducted the forward and backward two-sample Mendelian randomization (MR) analyses, based on summary-level genome-wide association studies (GWAS) data on PTSD and immune cell traits. RESULTS For the forward MR analysis, PTSD was found to reduce the levels of CD62L- dendritic cell (DC) (beta = -0.254, FDR = 0.01), CD86+ myeloid DC (beta = -0.238, FDR = 0.014), CD62L- myeloid DC (beta = -0.26, FDR = 0.01), CD62L- CD86+ myeloid DC absolute count (beta = -0.264, FDR = 0.024), and CD62L- CD86+ myeloid DC (beta = -0.328, FDR = 0.002). In contrast, PTSD was observed to increase the level of CD28- CD8dim T-cell absolute count (beta = 0.27, FDR = 0.029). For the backward MR analysis, the odds ratio (OR) for CD33 on CD33dim HLA DR+ CD11b- in relation to PTSD risk was found to be 1.045 (95% CI = 1.021-1.069, FDR = 0.008). The OR for FSC-A on HLA DR+ CD8br was 1.048 (95% CI = 1.018-1.079, FDR = 0.039) and for CCR2 on CD14- CD16+ monocyte was 1.059 (95% CI = 1.027-1.092, FDR = 0.008). No significant pleiotropy was detected in both forward and backward MR analyses. CONCLUSION The bidirectional MR study shed light on the intricate interplay between immune function and PTSD. The identification of a bidirectional causal relationship between T cells and PTSD opens new avenues for considering innovative approaches to the prevention and early intervention of PTSD.
Collapse
Affiliation(s)
- Jian Wang
- Shenzhen Mental Health CenterShenzhen Kangning HospitalShenzhenGuangdongChina
| | - Yuan Shao
- Shenzhen Mental Health CenterShenzhen Kangning HospitalShenzhenGuangdongChina
| | - Xianhua Deng
- Shenzhen Mental Health CenterShenzhen Kangning HospitalShenzhenGuangdongChina
| | - Jianbin Du
- Department of Geriatric PsychiatryThe Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation HospitalWuxiJiangsuChina
| |
Collapse
|
25
|
Orrapin S, Moonmuang S, Udomruk S, Yongpitakwattana P, Pruksakorn D, Chaiyawat P. Unlocking the tumor-immune microenvironment in osteosarcoma: insights into the immune landscape and mechanisms. Front Immunol 2024; 15:1394284. [PMID: 39359731 PMCID: PMC11444963 DOI: 10.3389/fimmu.2024.1394284] [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: 03/01/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Osteosarcoma has a unique tumor microenvironment (TME), which is characterized as a complex microenvironment comprising of bone cells, immune cells, stromal cells, and heterogeneous vascular structures. These elements are intricately embedded in a mineralized extracellular matrix, setting it apart from other primary TMEs. In a state of normal physiological function, these cell types collaborate in a coordinated manner to maintain the homeostasis of the bone and hematopoietic systems. However, in the pathological condition, i.e., neoplastic malignancies, the tumor-immune microenvironment (TIME) has been shown to promote cancer cells proliferation, migration, apoptosis and drug resistance, as well as immune escape. The intricate and dynamic system of the TIME in osteosarcoma involves crucial roles played by various infiltrating cells, the complement system, and exosomes. This complexity is closely associated with tumor cells evading immune surveillance, experiencing uncontrolled proliferation, and facilitating metastasis. In this review, we elucidate the intricate interplay between diverse cell populations in the osteosarcoma TIME, each contributing uniquely to tumor progression. From chondroblastic and osteoblastic osteosarcoma cells to osteoclasts, stromal cells, and various myeloid and lymphoid cell subsets, the comprehensive single-cell analysis provides a detailed roadmap of the complex osteosarcoma ecosystem. Furthermore, we summarize the mutations, epigenetic mechanisms, and extracellular vesicles that dictate the immunologic landscape and modulate the TIME of osteosarcoma. The perspectives of the clinical implementation of immunotherapy and therapeutic approaches for targeting immune cells are also intensively discussed.
Collapse
Affiliation(s)
- Santhasiri Orrapin
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sutpirat Moonmuang
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
| | - Sasimol Udomruk
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Petlada Yongpitakwattana
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Dumnoensun Pruksakorn
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Parunya Chaiyawat
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
26
|
Salem MB, Elzallat M, Mostafa Mohammed D, Hammam OA, Tamim A. Abdel-Wareth M, Hassan M. Helix pomatia mucin alleviates DSS-induced colitis in mice: Unraveling the cross talk between microbiota and intestinal chemokine. Heliyon 2024; 10:e37362. [PMID: 39296159 PMCID: PMC11407997 DOI: 10.1016/j.heliyon.2024.e37362] [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: 04/08/2024] [Revised: 08/17/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Gut microbiota imbalance and alterations in the chemokine-chemokine receptor interactions are pivotal in the initiation and advancement of ulcerative colitis (UC). The current UC treatments are prolonged, exhibit high recurrence rates, and may lead to colorectal cancer. So, this study explores the efficacy of Helix pomatia (H. pomatia) mucin in preventing DSS-induced UC. This research focuses on investigating the underlying mechanisms, such as oxidative stress, inflammation, and alterations in gut microbiota and chemokine-chemokine receptor interactions, to understand the anti-inflammatory and antioxidant characteristics of the mucin. Using 4 % DSS in drinking water, UC was induced in C57BL/6 mice. For seven days, mice were given oral doses of either H. pomatia mucin or sulfasalazine. The study assessed changes in oxidative stress, gut microbiota, and histopathology, along with expression of IL-6, CXCR4, CCR7, CXCL9, and CXCL10. The H. pomatia mucin exhibited unique contents, including high glycolic acid (200 ± 2.08 mg/L), collagen (88 ± 2.52 mg/L), allantoin (20 ± 2 mg/L), and concentrated vitamins and minerals. Treatment with H. pomatia mucin in high dose demonstrated reduction in DAI, an increase in fecal Firmicutes, and elevated expression of colonic CCR7, CXCL9, and CXCL10, accompanied by enhanced CXCR4 (75 %) and diminished IL-6 (1.33 %) immunostaining. It also alleviated oxidative stress, reduced fecal Bacteroidetes, and mitigated inflammation, indicating its potential efficacy against DSS-induced UC. In conclusion, H. pomatia mucin is a promising candidate that could be an effective adjuvant in the management and prophylaxis of UC.
Collapse
Affiliation(s)
- Maha B. Salem
- Pharmacology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohamed Elzallat
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Olfat A. Hammam
- Pathology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | | | - Marwa Hassan
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
27
|
Chen J, Ma Y, Liu Y, Zhao H, Qi X, Sun Y, Zhou X, Zhou J, Ma X, Wang L. CCL17 and CCL19 are markers of disease progression in alveolar echinococcosis. Cytokine 2024; 181:156669. [PMID: 38875750 DOI: 10.1016/j.cyto.2024.156669] [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: 05/10/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVES Alveolar echinococcosis (AE) represents one of the deadliest helminthic infections, characterized by an insidious onset and high lethality. METHODS This study utilized the Gene Expression Omnibus (GEO) database, applied Weighted Correlation Network Analysis (WGCNA) and Differential Expression Analysis (DEA), and employed the Matthews Correlation Coefficient (MCC) to identify CCL17 and CCL19 as key genes in AE. Immunohistochemistry and immunofluorescence co-localization techniques were used to examine the expression of CCL17 and CCL19 in liver tissue lesions of AE patients. Additionally, a mouse model of multilocular echinococcus larvae infection was developed to study the temporal expression patterns of these genes, along with liver fibrosis and inflammatory responses. RESULTS The in vitro model simulating echinococcal larva infection mirrored the hepatic microenvironment post-infection with multilocular echinococcal tapeworms. Quantitative RT-PCR analysis showed that liver fibrosis occurred in AE patients, with proximal activation and increased expression of CCL17 and CCL19 over time post-infection. Notably, expression peaked during the late stages of infection. Similarly, F4/80, a macrophage marker, exhibited corresponding trends in expression. Upon stimulation of normal hepatocytes by vesicular larvae in cellular experiments, there was a significant increase in CCL17 and CCL19 expression at 12 h post-infection, mirroring the upregulation observed with F4/80. CONCLUSION CCL17 and CCL19 facilitate macrophage aggregation via the chemokine pathway and their increased expression correlates with the progression of infection, suggesting their potential as biomarkers for AE progression.
Collapse
Affiliation(s)
- Jiahui Chen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Yuyu Ma
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Yumei Liu
- Xinjiang Medical University Affiliated Traditional Chinese Medicine Hospital, Medical Testing Center, Xinjiang, China
| | - Hui Zhao
- The First Affiliated Hospital of Xinjiang Medical University, Medical Testing Center, Xinjiang, China
| | - Xinwei Qi
- The First Affiliated Hospital of Xinjiang Medical University, Medical Testing Center, Xinjiang, China
| | - Yuqin Sun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Xuan Zhou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Jinping Zhou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Xiumin Ma
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China.
| | - Liang Wang
- The Fifth Affiliated Hospital of Xinjiang Medical University, Medical Testing Center, Xinjiang, China.
| |
Collapse
|
28
|
Kim J, Kang S, Kim J, Yong SB, Lahiji SF, Kim YH. Dual Adjuvant-Loaded Peptide Antigen Self-Assembly Potentiates Dendritic Cell-Mediated Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403663. [PMID: 39073756 PMCID: PMC11423174 DOI: 10.1002/advs.202403663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Clinical translation of current cancer vaccine research has been hampered by limited antitumor immune responses due to inefficient antigen delivery and presentation, suboptimal DC and T cell activation. Biomaterial-based nanovaccine offers targeted antigen delivery, protection from degradation in vivo, and prolonged tumor therapeutic efficacy. This study introduces a lipid-coated deoxycholic acid-survivin nanoassembly (DA-L-DSA). Survivin, overexpressed in several cancer cells and involved in cancer cell growth and immune evasion, is selected as a tumor-associated antigen. An major histocompatibility complex class I binding epitope of survivin is engineered into the nanoassembly. R848, TLR 7/8 agonist, and SD-208, TGF-beta receptor1 kinase inhibitor, are coencapsulated into the nanoassembly as potent adjuvants to boost DC maturation and enhance antigen presentation. The DA-L-DSA effectively stimulates the maturation of dendritic cells, migrates into lymph nodes, and enhances T-cell activation and Th1 response. A substantial influx of cytotoxic T lymphocytes into primary tumors is observed in a murine melanoma model and demonstrates anti-metastatic effects in a spontaneous breast cancer metastasis model. Furthermore, DA-L-DSA exhibits a remarkable synergistic effect in the combination therapy with immune checkpoint inhibitors alleviating immunosuppressive tumor microenvironment. Taken together, these findings suggest DA-L-DSA as a promising immuno-therapeutic platform that could be applicable to diverse intractable cancers.
Collapse
Affiliation(s)
- Jaehyun Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seyoung Kang
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jisu Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seok-Beom Yong
- Nucleic Acid Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do, 28116, Republic of Korea
| | - Shayan Fakhraei Lahiji
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 04763, Republic of Korea
- Cursus Bio Inc. Icure Tower, Seoul, 06170, Republic of Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 04763, Republic of Korea
- Cursus Bio Inc. Icure Tower, Seoul, 06170, Republic of Korea
- Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, 04763, Republic of Korea
| |
Collapse
|
29
|
Zhang W, Zhang J, Jiao D, Tang Q, Gao X, Li Z, Yang F, Zhao Z, Yang L. Single-Cell RNA Sequencing Reveals a Unique Fibroblastic Subset and Immune Disorder in Lichen Sclerosus Urethral Stricture. J Inflamm Res 2024; 17:5327-5346. [PMID: 39157587 PMCID: PMC11330248 DOI: 10.2147/jir.s466317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024] Open
Abstract
Purpose Lichen sclerosus urethral stricture disease (LS USD) is a refractory and progressive disease primarily affecting the anterior urethra in males. Various potential etiological factors, such as genetics, autoimmunity, infection, and exposure to infectious urine, have been suggested. However, the accurate etiology of LS in the male urethra remains unclear. Patients and Methods In this study, we conducted single-cell RNA sequencing to identify the transcriptional profiles of three patients with LS USD and three patients with non-LS USD. Immunofluorescence was used to confirm the single-cell sequence results. Results Our study revealed distinct subsets of vein endothelial cells (ECs), smooth muscle cells (SMCs), and fibroblasts (FBs) with high proportions in LS USD, contributing to the tissue microenvironment primarily involved in proinflammatory and immune responses. In particular, FBs displayed a unique subset, Fib7, which is exclusively present in LS USD, and exhibited high expression levels of SAA1 and SAA2. The accumulation of macrophages, along with the dysregulated ratios of M1/M2-like phenotype macrophages, may be engaged in the pathogenesis of LS USD. Through cell-cell communication analysis, we identified significant interactions involving CXCL8/ACKR1 and CCR7/CCL19 in LS USD. Remarkably, Fib7 exhibited exclusive communication with IL-1B macrophages through the SAA1/FPR2 receptor-ligand pair. Conclusion Our study provides a profound understanding of the tissue microenvironment in LS USD, which may be valuable for understanding the pathogenesis of LS USD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Jiayu Zhang
- Department of Urology, Air Force Hospital of Southern Theater Command, Guangzhou, Guangdong, 510062, People’s Republic of China
| | - Dian Jiao
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Qisheng Tang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Xiaoping Gao
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Zhenyu Li
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Zhiguang Zhao
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Longfei Yang
- Department of Transfusion Medicine, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| |
Collapse
|
30
|
Xu Y, Gu L, Zhu L, Miao Y, Cui X. A novel anti-CD47 protein antibody and toll-like receptor agonist complex detects tumor surface CD-47 changes in early stage lung cancer by in vivo imaging. Int J Biol Macromol 2024; 274:133322. [PMID: 38908646 DOI: 10.1016/j.ijbiomac.2024.133322] [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: 05/07/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
CD47, a cell surface protein known for inhibiting phagocytosis, plays a critical role in the tumor microenvironment (TME) and is a potential biomarker for cancer. However, directly applying αCD47, a hydrophilic macromolecular antibody that targets CD47, in vivo for cancer detection can have adverse effects on normal cells, cause systemic toxicities, and lead to resistance against anti-cancer therapies. In this study, we developed a novel complex incorporating aluminum-based metal-organic frameworks (Al-MOF) loaded with indocyanine green (ICG), αCD47, and resiquimod (R848), a hydrophobic small molecule Toll-like receptor 7/8 (TLR7/8) agonist. Upon activation with an infrared 808 nm laser, the nanocomposites exhibited photothermal effects that triggered the release of the loaded reagents, induced ROS production, and induced changes in the TME. This led to the polarization of immune-suppressive M2 macrophages towards an immune-stimulatory M1 phenotype, promoted dendritic cell (DC) maturation, and enabled mature DCs to facilitate antigen presentation, T-cell activation, and critical roles in tumor immunity. Furthermore, in vivo imaging successfully detected the specific binding of αCD47 with CD47 on tumor cells. Overall, the complex composed of αCD47 antibody and toll-like receptor agonist showed promising efficacy in both tumor diagnosis and therapy, providing a potential strategy for detecting early lung cancer and modulating the tumor microenvironment for improved treatment outcomes.
Collapse
Affiliation(s)
- Yunhua Xu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Linping Gu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Li Zhu
- Department of Radiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yayou Miao
- Department of Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xueying Cui
- Department of Nutrition, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
31
|
Fan Y, Meng Y, Hu X, Liu J, Qin X. Uncovering novel mechanisms of chitinase-3-like protein 1 in driving inflammation-associated cancers. Cancer Cell Int 2024; 24:268. [PMID: 39068486 PMCID: PMC11282867 DOI: 10.1186/s12935-024-03425-y] [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: 02/25/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that is induced and regulated by multiple factors during inflammation in enteritis, pneumonia, asthma, arthritis, and other diseases. It is associated with the deterioration of the inflammatory environment in tissues with chronic inflammation caused by microbial infection or autoimmune diseases. The expression of CHI3L1 expression is upregulated in several malignant tumors, underscoring the crucial role of chronic inflammation in the initiation and progression of cancer. While the precise mechanism connecting inflammation and cancer is unclear, the involvement of CHI3L1 is involved in chronic inflammation, suggesting its role as a contributing factor to in the link between inflammation and cancer. CHI3L1 can aggravate DNA oxidative damage, induce the cancerous phenotype, promote the development of a tumor inflammatory environment and angiogenesis, inhibit immune cells, and promote cancer cell growth, invasion, and migration. Furthermore, it participates in the initiation of cancer progression and metastasis by binding with transmembrane receptors to mediate intracellular signal transduction. Based on the current research on CHI3L1, we explore introduce the receptors that interact with CHI3L1 along with the signaling pathways that may be triggered during chronic inflammation to enhance tumorigenesis and progression. In the last section of the article, we provide a brief overview of anti-inflammatory therapies that target CHI3L1.
Collapse
Affiliation(s)
- Yan Fan
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Yuan Meng
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xingwei Hu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xiaosong Qin
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China.
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China.
| |
Collapse
|
32
|
Gao Y, Huang D, Huang S, Li H, Xia B. Rational design of ROS generation nanosystems to regulate innate immunity of macrophages, dendrtical and natural killing cells for immunotherapy. Int Immunopharmacol 2024; 139:112695. [PMID: 39024751 DOI: 10.1016/j.intimp.2024.112695] [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: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Innate immunity serves as the first line of host defense in the body against pathogenic infections or malignant diseases. Reactive oxygen species (ROS), as vital signaling mediators, can efficiently elicit innate immune responses to oxidative-related stress or damage. In the era of nanomedicine, various immunostimulatory nanosystems have been extensively designed and synthesized to elicit immune responses for the immunotherapy of cancer or infectious diseases. In this review, we emphasize that ROS derived from nanosystems regulates innate immune cells to potentiate immunotherapeutic efficacy, such as primarily dendritic cells, macrophages, or natural killer cells. Meanwhile, we also summarize the pathway of ROS generation triggered by exogenous nanosystems in innate immune cells of DCs, macrophages, and NK cells.
Collapse
Affiliation(s)
- Yan Gao
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China
| | - Di Huang
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China
| | - Shuodan Huang
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China
| | - Huiying Li
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, PR China.
| | - Bing Xia
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China; Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, PR China.
| |
Collapse
|
33
|
Trivedi AH, Wang VZ, McClain EJ, Vyas PS, Swink IR, Snell ED, Cheng BC, DeMeo PJ. The Categorization of Perinatal Derivatives for Orthopedic Applications. Biomedicines 2024; 12:1544. [PMID: 39062117 PMCID: PMC11274709 DOI: 10.3390/biomedicines12071544] [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: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Musculoskeletal (MSK) pathology encompasses an array of conditions that can cause anything from mild discomfort to permanent injury. Their prevalence and impact on disability have sparked interest in more effective treatments, particularly within orthopedics. As a result, the human placenta has come into focus within regenerative medicine as a perinatal derivative (PnD). These biologics are sourced from components of the placenta, each possessing a unique composition of collagens, proteins, and factors believed to aid in healing and regeneration. This review aims to explore the current literature on PnD biologics and their potential benefits for treating various MSK pathologies. We delve into different types of PnDs and their healing effects on muscles, tendons, bones, cartilage, ligaments, and nerves. Our discussions highlight the crucial role of immune modulation in the healing process for each condition. PnDs have been observed to influence the balance between anti- and pro-inflammatory factors and, in some cases, act as biologic scaffolds for tissue growth. Additionally, we assess the range of PnDs available, while also addressing gaps in our understanding, particularly regarding biologic processing methods. Although certain PnD biologics have varying levels of support in orthopedic literature, further clinical investigations are necessary to fully evaluate their impact on human patients.
Collapse
Affiliation(s)
- Amol H. Trivedi
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
- Drexel University College of Medicine, Drexel University, University City Campus, Philadelphia, PA 19104, USA
| | - Vicki Z. Wang
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Edward J. McClain
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Praveer S. Vyas
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Isaac R. Swink
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Edward D. Snell
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Boyle C. Cheng
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Patrick J. DeMeo
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| |
Collapse
|
34
|
Xu Z, Xiao ZX, Wang J, Qiu HW, Cao F, Zhang SQ, Xu YD, Lei HQ, Xia H, He YR, Zha GF, Pang J. Novel mRNA adjuvant ImmunER enhances prostate cancer tumor-associated antigen mRNA therapy via augmenting T cell activity. Oncoimmunology 2024; 13:2373526. [PMID: 38948931 PMCID: PMC11212567 DOI: 10.1080/2162402x.2024.2373526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024] Open
Abstract
Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ze-Xiu Xiao
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen, Guangdong, China
| | - Jing Wang
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen, Guangdong, China
| | - Hao-Wei Qiu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Fei Cao
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shi-Qiang Zhang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuan-Dong Xu
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Han-Qi Lei
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Heng Xia
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yun-Ru He
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Gao-Feng Zha
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jun Pang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
35
|
Tian W, Blomberg AL, Steinberg KE, Henriksen BL, Jørgensen JS, Skovgaard K, Skovbakke SL, Goletz S. Novel genetically glycoengineered human dendritic cell model reveals regulatory roles of α2,6-linked sialic acids in DC activation of CD4+ T cells and response to TNFα. Glycobiology 2024; 34:cwae042. [PMID: 38873803 DOI: 10.1093/glycob/cwae042] [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: 01/19/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
Dendritic cells (DCs) are central for the initiation and regulation of appropriate immune responses. While several studies suggest important regulatory roles of sialoglycans in DC biology, our understanding is still inadequate primarily due to a lack of appropriate models. Previous approaches based on enzymatic- or metabolic-glycoengineering and primary cell isolation from genetically modified mice have limitations related to specificity, stability, and species differences. This study addresses these challenges by introducing a workflow to genetically glycoengineer the human DC precursor cell line MUTZ-3, described to differentiate and maturate into fully functional dendritic cells, using CRISPR-Cas9, thereby providing and validating the first isogenic cell model for investigating glycan alteration on human DC differentiation, maturation, and activity. By knocking out (KO) the ST6GAL1 gene, we generated isogenic cells devoid of ST6GAL1-mediated α(2,6)-linked sialylation, allowing for a comprehensive investigation into its impact on DC function. Glycan profiling using lectin binding assay and functional studies revealed that ST6GAL1 KO increased the expression of important antigen presenting and co-stimulatory surface receptors and a specifically increased activation of allogenic human CD4 + T cells. Additionally, ST6GAL1 KO induces significant changes in surface marker expression and cytokine response to TNFα-induced maturation, and it affects migration and the endocytic capacity. These results indicate that genetic glycoengineering of the isogenic MUTZ-3 cellular model offers a valuable tool to study how specific glycan structures influence human DC biology, contributing to our understanding of glycoimmunology.
Collapse
Affiliation(s)
- Weihua Tian
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Anne Louise Blomberg
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Kaylin Elisabeth Steinberg
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Betina Lyngfeldt Henriksen
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Josefine Søborg Jørgensen
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Kerstin Skovgaard
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Sarah Line Skovbakke
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Steffen Goletz
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| |
Collapse
|
36
|
Dai P, Ding M, Yu J, Gao Y, Wang M, Ling J, Dong S, Zhang X, Zeng X, Sun X. The Male Reproductive Toxicity Caused by 2-Naphthylamine Was Related to Testicular Immunity Disorders. TOXICS 2024; 12:342. [PMID: 38787121 PMCID: PMC11126000 DOI: 10.3390/toxics12050342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
2-naphthylamine (NAP) was classified as a group I carcinogen associated with bladder cancer. The daily exposure is mostly from cigarette and E-cigarette smoke. NAP can lead to testicular atrophy and interstitial tissue hyperplasia; however, the outcomes of NAP treatment on spermatogenesis and the associated mechanisms have not been reported. The study aimed to investigate the effect of NAP on spermatogenesis and sperm physiologic functions after being persistently exposed to NAP at 5, 20, and 40 mg/kg for 35 days. We found that sperm motility, progressive motility, sperm average path velocity, and straight-line velocity declined remarkably in the NAP (40 mg/kg) treated group, and the sperm deformation rate rose upon NAP administration. The testis immunity- and lipid metabolism-associated processes were enriched from RNA-sequence profiling. Plvap, Ccr7, Foxn1, Trim29, Sirpb1c, Cfd, and Lpar4 involved in testis immunity and Pnliprp1 that inhibit triglyceride and cholesterol absorption were confirmed to rise dramatically in the NAP-exposed group. The increased total cholesterol and CD68 levels were observed in the testis from the NAP-exposed group. Gpx5, serving as an antioxidant in sperm plasma, and Semg1, which contributes to sperm progressive motility, were both down-regulated. We concluded that the short-term exposure to NAP caused reproductive toxicity, primarily due to the inflammatory abnormality in the testis.
Collapse
Affiliation(s)
- Pengyuan Dai
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Mengqian Ding
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Jingyan Yu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Yuan Gao
- Experimental Animal Center, Nantong University, Nantong 226001, China;
| | - Miaomiao Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Jie Ling
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Shijue Dong
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Xiaoli Sun
- Center for Reproductive Medicine, The Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| |
Collapse
|
37
|
Zhong D, Shi Y, Ma W, Liang Y, Liu H, Qin Y, Zhang L, Yang Q, Huang X, Lu Y, Shang J. Single-cell profiling reveals the metastasis-associated immune signature of hepatocellular carcinoma. Immun Inflamm Dis 2024; 12:e1264. [PMID: 38780041 PMCID: PMC11112628 DOI: 10.1002/iid3.1264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
AIM Metastasis is the leading cause of mortality in hepatocellular carcinoma (HCC). The metastasis-associated immune signature in HCC is worth exploring. METHODS Bioinformatic analysis was conducted based on the single-cell transcriptome data derived from HCC patients in different stages. Cellular composition, pseudotime state transition, and cell-cell interaction were further analyzed and verified. RESULTS Generally, HCC with metastasis exhibited suppressive immune microenvironment, while HCC without metastasis exhibited active immune microenvironment. Concretely, effector regulatory T cells (eTregs) were found to be enriched in HCC with metastasis. PHLDA1 was identified as one of exhaustion-specific genes and verified to be associated with worse prognosis in HCC patients. Moreover, A novel cluster of CCR7+ dendritic cells (DCs) was identified with high expression of maturation and migration marker genes. Pseudotime analysis showed that inhibition of differentiation occurred in CCR7+ DCs rather than cDC1 in HCC with metastasis. Furthermore, interaction analysis showed that the reduction of CCR7+ DCs lead to impaired CCR7/CCL19 interaction in HCC with metastasis. CONCLUSIONS HCC with metastasis exhibited upregulation of exhaustion-specific genes of eTregs and inhibition of CCL signal of a novel DC cluster, which added new dimensions to the immune landscape and provided new immune therapeutic targets in advanced HCC.
Collapse
Affiliation(s)
- Deyuan Zhong
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Ying Shi
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacau SARChina
| | - Yuxin Liang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Hanjie Liu
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Yingying Qin
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Lu Zhang
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Qinyan Yang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Xiaolun Huang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Yuanjun Lu
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Jin Shang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
38
|
Liu C, Wu H, Li K, Chi Y, Wu Z, Xing C. Identification of biomarkers for abdominal aortic aneurysm in Behçet's disease via mendelian randomization and integrated bioinformatics analyses. J Cell Mol Med 2024; 28:e18398. [PMID: 38785203 PMCID: PMC11117452 DOI: 10.1111/jcmm.18398] [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: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Behçet's disease (BD) is a complex autoimmune disorder impacting several organ systems. Although the involvement of abdominal aortic aneurysm (AAA) in BD is rare, it can be associated with severe consequences. In the present study, we identified diagnostic biomarkers in patients with BD having AAA. Mendelian randomization (MR) analysis was initially used to explore the potential causal association between BD and AAA. The Limma package, WGCNA, PPI and machine learning algorithms were employed to identify potential diagnostic genes. A receiver operating characteristic curve (ROC) for the nomogram was constructed to ascertain the diagnostic value of AAA in patients with BD. Finally, immune cell infiltration analyses and single-sample gene set enrichment analysis (ssGSEA) were conducted. The MR analysis indicated a suggestive association between BD and the risk of AAA (odds ratio [OR]: 1.0384, 95% confidence interval [CI]: 1.0081-1.0696, p = 0.0126). Three hub genes (CD247, CD2 and CCR7) were identified using the integrated bioinformatics analyses, which were subsequently utilised to construct a nomogram (area under the curve [AUC]: 0.982, 95% CI: 0.944-1.000). Finally, the immune cell infiltration assay revealed that dysregulation immune cells were positively correlated with the three hub genes. Our MR analyses revealed a higher susceptibility of patients with BD to AAA. We used a systematic approach to identify three potential hub genes (CD247, CD2 and CCR7) and developed a nomogram to assist in the diagnosis of AAA among patients with BD. In addition, immune cell infiltration analysis indicated the dysregulation in immune cell proportions.
Collapse
Affiliation(s)
- Chunjiang Liu
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Huadong Wu
- Department of vascular surgeryFirst affiliated Hospital of Huzhou UniversityHuzhouChina
| | - Kuan Li
- Department of General SurgeryKunshan Hospital of Traditional Chinese MedicineKunshanChina
| | - Yongxing Chi
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhaoying Wu
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chungen Xing
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
39
|
Song MS, Nam JH, Noh KE, Lim DS. Dendritic Cell-Based Immunotherapy: The Importance of Dendritic Cell Migration. J Immunol Res 2024; 2024:7827246. [PMID: 38628676 PMCID: PMC11019573 DOI: 10.1155/2024/7827246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are crucial for maintaining self-tolerance, initiating immune responses against pathogens, and patrolling body compartments. Despite promising aspects, DC-based immunotherapy faces challenges that include limited availability, immune escape in tumors, immunosuppression in the tumor microenvironment, and the need for effective combination therapies. A further limitation in DC-based immunotherapy is the low population of migratory DC (around 5%-10%) that migrate to lymph nodes (LNs) through afferent lymphatics depending on the LN draining site. By increasing the population of migratory DCs, DC-based immunotherapy could enhance immunotherapeutic effects on target diseases. This paper reviews the importance of DC migration and current research progress in the context of DC-based immunotherapy.
Collapse
Affiliation(s)
- Min-Seon Song
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Ji-Hee Nam
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Kyung-Eun Noh
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Dae-Seog Lim
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
40
|
Wang Y, Zhao S, Zhang X, Xia D, Xiao Y, Zhou X, Zhan T, Xia X, Shu Y, Xu H, Li W. Single-Cell Transcriptome Analysis Reveals Interaction between CCL19+ Inflammatory Keratinocytes and CCR7+ Dendritic Cells and B Cells in Pemphigus. J Invest Dermatol 2024:S0022-202X(24)00213-6. [PMID: 38537931 DOI: 10.1016/j.jid.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 04/20/2024]
Affiliation(s)
- Yiyi Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiwen Zhang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dengmei Xia
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Xiao
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xingli Zhou
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tongying Zhan
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuyang Xia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China; Research Center of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei Li
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China; Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
41
|
Vanalken N, Boon K, Szpakowska M, Chevigné A, Schols D, Van Loy T. Systematic Assessment of Human CCR7 Signalling Using NanoBRET Biosensors Points towards the Importance of the Cellular Context. BIOSENSORS 2024; 14:142. [PMID: 38534251 DOI: 10.3390/bios14030142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
The human CC chemokine receptor 7 (CCR7) is activated by two natural ligands, CC chemokine ligand 19 (CCL19) and 21 (CCL21). The CCL19-CCL21-CCR7 axis has been extensively studied in vitro, but there is still debate over whether CCL21 is an overall weaker agonist or if the axis displays biased signalling. In this study, we performed a systematic analysis at the transducer level using NanoBRET-based methodologies in three commonly used cellular backgrounds to evaluate pathway and ligand preferences, as well as ligand bias and the influence of the cellular system thereon. We found that both CCL19 and CCL21 activated all cognate G proteins and some non-cognate couplings in a cell-type-dependent manner. Both ligands recruited β-arrestin1 and 2, but the potency was strongly dependent on the cellular system. Overall, CCL19 and CCL21 showed largely conserved pathway preferences, but small differences were detected. However, these differences only consolidated in a weak ligand bias. Together, these data suggest that CCL19 and CCL21 share mostly overlapping, weakly biased, transducer profiles, which can be influenced by the cellular context.
Collapse
Affiliation(s)
- Nathan Vanalken
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, 3000 Leuven, Belgium
| | - Katrijn Boon
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, 3000 Leuven, Belgium
| | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, 3000 Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, 3000 Leuven, Belgium
| |
Collapse
|
42
|
Reitsema RD, Kumawat AK, Hesselink BC, van Baarle D, van Sleen Y. Effects of ageing and frailty on circulating monocyte and dendritic cell subsets. NPJ AGING 2024; 10:17. [PMID: 38438383 PMCID: PMC10912203 DOI: 10.1038/s41514-024-00144-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/14/2024] [Indexed: 03/06/2024]
Abstract
Ageing is associated with dysregulated immune responses, resulting in impaired resilience against infections and low-grade inflammation known as inflammageing. Frailty is a measurable condition in older adults characterized by decreased health and physical impairment. Dendritic cells (DCs) and monocytes play a crucial role in initiating and steering immune responses. To assess whether their frequencies and phenotypes in the blood are affected by ageing or frailty, we performed a flow cytometry study on monocyte and DC subsets in an immune ageing cohort. We included (n = 15 in each group) healthy young controls (HYC, median age 29 years), healthy older controls (HOC, 73 years) and Frail older controls (76 years). Monocyte subsets (classical, intermediate, non-classical) were identified by CD14 and CD16 expression, and DC subsets (conventional (c)DC1, cDC2, plasmacytoid (p)DC) by CD11c, CD1c, CD141 and CD303 expression. All subsets were checked for TLR2, TLR4, HLA-DR, CD86, PDL1, CCR7 and CD40 expression. We observed a lower proportion of pDCs in HOC compared to HYC. Additionally, we found higher expression of activation markers on classical and intermediate monocytes and on cDC2 in HOC compared to HYC. Frail participants had a higher expression of CD40 on classical and non-classical monocytes compared to the HOC group. We document a substantial effect of ageing on monocytes and DCs. Reduced pDCs in older people may underlie their impaired ability to counter viral infections, whereas enhanced expression of activation markers could indicate a state of inflammageing. Future studies could elucidate the functional consequences of CD40 upregulation with frailty.
Collapse
Affiliation(s)
- Rosanne D Reitsema
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ashok K Kumawat
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Bernd-Cornèl Hesselink
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
43
|
Shao M, Wang M, Wang X, Feng X, Zhang L, Lv H. SQLE is a promising prognostic and immunological biomarker and correlated with immune Infiltration in Sarcoma. Medicine (Baltimore) 2024; 103:e37030. [PMID: 38335381 PMCID: PMC10861000 DOI: 10.1097/md.0000000000037030] [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: 09/23/2023] [Revised: 11/24/2023] [Accepted: 01/02/2024] [Indexed: 02/12/2024] Open
Abstract
Squalene epoxidase (SQLE) is an essential enzyme involved in cholesterol biosynthesis. However, its role in sarcoma and its correlation with immune infiltration remains unclear. All original data were downloaded from The Cancer Genome Atlas (TCGA). SQLE expression was explored using the TCGA database, and correlations between SQLE and cancer immune characteristics were analyzed via the TISIDB databases. Generally, SQLE is predominantly overexpressed and has diagnostic and prognostic value in sarcoma. Upregulated SQLE was associated with poorer overall survival, poorer disease-specific survival, and tumor multifocality in sarcoma. Mechanistically, we identified a hub gene that included a total of 82 SQLE-related genes, which were tightly associated with histone modification pathways in sarcoma patients. SQLE expression was negatively correlated with infiltrating levels of dendritic cells and plasmacytoid dendritic cells and positively correlated with Th2 cells. SQLE expression was negatively correlated with the expression of chemokines (CCL19 and CX3CL1) and chemokine receptors (CCR2 and CCR7) in sarcoma. In conclusion, SQLE may be used as a prognostic biomarker for determining prognosis and immune infiltration in sarcoma.
Collapse
Affiliation(s)
- Mengwei Shao
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Mingbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xiliang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xiaodong Feng
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Lifeng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Huicheng Lv
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
44
|
Narvaez D, Nadal J, Nervo A, Costanzo MV, Paletta C, Petracci FE, Rivero S, Ostinelli A, Freile B, Enrico D, Pombo MT, Amat M, Aguirre ED, Chacon M, Waisberg F. The Emerging Role of Tertiary Lymphoid Structures in Breast Cancer: A Narrative Review. Cancers (Basel) 2024; 16:396. [PMID: 38254885 PMCID: PMC10814091 DOI: 10.3390/cancers16020396] [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: 10/26/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 01/24/2024] Open
Abstract
This narrative review aims to clarify the role of tertiary lymphoid structures in breast cancer. We examine their development, composition, and prognostic value, and current ways of recognizing them. A comprehensive literature review was performed using the PubMed/Medline, Scopus, and EMBASE databases. A significant area of interest in breast cancer research involves targeting immune checkpoint molecules, particularly in the triple-negative subtype, where treatment options remain limited. However, existing biomarkers have limitations in accurately predicting treatment response. In this context, tertiary lymphoid structures (TLSs) emerge as a prognostic biomarker and also as a promising predictive marker for response. TLSs are ectopic lymphoid formations or neo-organogenesis that can develop after prolonged exposure to inflammatory signals mediated by chemokines and cytokines. Their presence is inversely correlated with estrogen receptor (ER) and/or progesterone receptor (PR) expression, but positively associated with a higher pathologic complete response rate and improved overall survival. In certain scenarios, TLS-positive tumors were associated with improved outcomes regardless of the presence of PDL-1 (programmed cell death ligand 1) expression or TILs (tumor-infiltrating lymphocytes).
Collapse
Affiliation(s)
- Dana Narvaez
- Breast Cancer Division, Alexander Fleming Institute, Buenos Aires 1425, Argentina; (J.N.); (A.N.); (M.V.C.); (C.P.); (F.E.P.); (S.R.); (A.O.); (B.F.); (D.E.); (M.T.P.); (M.A.); (E.D.A.); (M.C.); (F.W.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
O’Brien G, Kamuda M, Cruz-Garcia L, Polozova M, Tichy A, Markova M, Sirak I, Zahradnicek O, Widłak P, Ponge L, Polanska J, Badie C. Transcriptional Inflammatory Signature in Healthy Donors and Different Radiotherapy Cancer Patients. Int J Mol Sci 2024; 25:1080. [PMID: 38256152 PMCID: PMC10816540 DOI: 10.3390/ijms25021080] [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: 12/10/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer and ionizing radiation exposure are associated with inflammation. To identify a set of radiation-specific signatures of inflammation-associated genes in the blood of partially exposed radiotherapy patients, differential expression of 249 inflammatory genes was analyzed in blood samples from cancer patients and healthy individuals. The gene expression analysis on a cohort of 63 cancer patients (endometrial, head and neck, and prostate cancer) before and during radiotherapy (24 h, 48 h, ~1 week, ~4-8 weeks, and 1 month after the last fraction) identified 31 genes and 15 up- and 16 down-regulated genes. Transcription variability under normal conditions was determined using blood drawn on three separate occasions from four healthy donors. No difference in inflammatory expression between healthy donors and cancer patients could be detected prior to radiotherapy. Remarkably, repeated sampling of healthy donors revealed an individual endogenous inflammatory signature. Next, the potential confounding effect of concomitant inflammation was studied in the blood of seven healthy donors taken before and 24 h after a flu vaccine or ex vivo LPS (lipopolysaccharide) treatment; flu vaccination was not detected at the transcriptional level and LPS did not have any effect on the radiation-induced signature identified. Finally, we identified a radiation-specific signature of 31 genes in the blood of radiotherapy patients that were common for all cancers, regardless of the immune status of patients. Confirmation via MQRT-PCR was obtained for BCL6, MYD88, MYC, IL7, CCR4 and CCR7. This study offers the foundation for future research on biomarkers of radiation exposure, radiation sensitivity, and radiation toxicity for personalized radiotherapy treatment.
Collapse
Affiliation(s)
- Gráinne O’Brien
- Cancer Mechanisms and Biomarkers Group, Centre for Radiation, Chemical and Environmental Hazards, UK Health Security Agency, Oxfordshire OX11 0RQ, UK; (G.O.); (L.C.-G.); (M.P.)
| | - Malgorzata Kamuda
- Department of Data Mining, Silesian University of Technology, 44-100 Gliwice, Poland (J.P.)
| | - Lourdes Cruz-Garcia
- Cancer Mechanisms and Biomarkers Group, Centre for Radiation, Chemical and Environmental Hazards, UK Health Security Agency, Oxfordshire OX11 0RQ, UK; (G.O.); (L.C.-G.); (M.P.)
| | - Mariia Polozova
- Cancer Mechanisms and Biomarkers Group, Centre for Radiation, Chemical and Environmental Hazards, UK Health Security Agency, Oxfordshire OX11 0RQ, UK; (G.O.); (L.C.-G.); (M.P.)
| | - Ales Tichy
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Králové, University of Defence, 662 10 Brno, Czech Republic
- Biomedical Research Centre, University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic
| | - Marketa Markova
- Institute of Hematology and Blood Transfusion, 128 00 Praha, Czech Republic;
| | - Igor Sirak
- Department of Oncology and Radiotherapy and 4th Department of Internal Medicine—Hematology, University Hospital, 500 05 Hradec Králové, Czech Republic;
| | - Oldrich Zahradnicek
- Department of Radiation Dosimetry, Nuclear Physics Institute, Czech Academy of Sciences, 180 00 Prague, Czech Republic;
| | - Piotr Widłak
- Clinical Research Support Centre, Medical University of Gdańsk, Gdańsk, M. Skłodowskiej-Curie 3a Street, 80-210 Gdańsk, Poland;
| | - Lucyna Ponge
- Maria Skłodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland;
| | - Joanna Polanska
- Department of Data Mining, Silesian University of Technology, 44-100 Gliwice, Poland (J.P.)
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers Group, Centre for Radiation, Chemical and Environmental Hazards, UK Health Security Agency, Oxfordshire OX11 0RQ, UK; (G.O.); (L.C.-G.); (M.P.)
| |
Collapse
|
46
|
Liu B, Wang Y, Han G, Zhu M. Tolerogenic dendritic cells in radiation-induced lung injury. Front Immunol 2024; 14:1323676. [PMID: 38259434 PMCID: PMC10800505 DOI: 10.3389/fimmu.2023.1323676] [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: 10/18/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Radiation-induced lung injury is a common complication associated with radiotherapy. It is characterized by early-stage radiation pneumonia and subsequent radiation pulmonary fibrosis. However, there is currently a lack of effective therapeutic strategies for radiation-induced lung injury. Recent studies have shown that tolerogenic dendritic cells interact with regulatory T cells and/or regulatory B cells to stimulate the production of immunosuppressive molecules, control inflammation, and prevent overimmunity. This highlights a potential new therapeutic activity of tolerogenic dendritic cells in managing radiation-induced lung injury. In this review, we aim to provide a comprehensive overview of tolerogenic dendritic cells in the context of radiation-induced lung injury, which will be valuable for researchers in this field.
Collapse
Affiliation(s)
| | - Yilong Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | | | - Maoxiang Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
47
|
Chen J, Pan Y, Liu Q, Li G, Chen G, Li W, Zhao W, Wang Q. The Interplay between Meningeal Lymphatic Vessels and Neuroinflammation in Neurodegenerative Diseases. Curr Neuropharmacol 2024; 22:1016-1032. [PMID: 36380442 PMCID: PMC10964105 DOI: 10.2174/1570159x21666221115150253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
Meningeal lymphatic vessels (MLVs) are essential for the drainage of cerebrospinal fluid, macromolecules, and immune cells in the central nervous system. They play critical roles in modulating neuroinflammation in neurodegenerative diseases. Dysfunctional MLVs have been demonstrated to increase neuroinflammation by horizontally blocking the drainage of neurotoxic proteins to the peripheral lymph nodes. Conversely, MLVs protect against neuroinflammation by preventing immune cells from becoming fully encephalitogenic. Furthermore, evidence suggests that neuroinflammation affects the structure and function of MLVs, causing vascular anomalies and angiogenesis. Although this field is still in its infancy, the strong link between MLVs and neuroinflammation has emerged as a potential target for slowing the progression of neurodegenerative diseases. This review provides a brief history of the discovery of MLVs, introduces in vivo and in vitro MLV models, highlights the molecular mechanisms through which MLVs contribute to and protect against neuroinflammation, and discusses the potential impact of neuroinflammation on MLVs, focusing on recent progress in neurodegenerative diseases.
Collapse
Affiliation(s)
- Junmei Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yaru Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Qihua Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Guangyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Gongcan Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| |
Collapse
|
48
|
Lin Y, Peng G, Bruner DW, Miller AH, Saba NF, Higgins KA, Shin DM, Claussen H, Johnston HR, Houser MC, Wommack EC, Xiao C. Associations of differentially expressed genes with psychoneurological symptoms in patients with head and neck cancer: A longitudinal study. J Psychosom Res 2023; 175:111518. [PMID: 37832274 PMCID: PMC11789059 DOI: 10.1016/j.jpsychores.2023.111518] [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: 07/27/2023] [Revised: 10/02/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023]
Abstract
OBJECTIVE Patients with head and neck cancer (HNC) experience psychoneurological symptoms (PNS, i.e., depression, fatigue, sleep disturbance, pain, and cognitive dysfunction) during intensity-modulated radiotherapy (IMRT) that negatively impact their functional status, quality of life, and overall survival. The underlying mechanisms for PNS are still not fully understood. This study aimed to examine differentially expressed genes and pathways related to PNS for patients undergoing IMRT (i.e., before, end of, 6 months, and 12 months after IMRT). METHODS Participants included 142 patients with HNC (mean age 58.9 ± 10.3 years, 72.5% male, 83.1% White). Total RNA extracted from blood leukocytes were used for genome-wide gene expression assays. Linear mixed effects model was used to examine the association between PNS and gene expression across time. Gene Ontology (GO) enrichment analysis was employed to identify pathways related to PNS. RESULTS A total of 1352 genes (162 upregulated, 1190 downregulated) were significantly associated with PNS across time (false discovery rate (FDR) < 0.05). Among these genes, 112 GO terms were identified (FDR < 0.05). The top 20 GO terms among the significant upregulated genes were related to immune and inflammatory responses, while the top 20 GO terms among the significant downregulated genes were associated with telomere maintenance. CONCLUSION This study is the first to identify genes and pathways linked to immune and inflammatory responses and telomere maintenance that are associated with PNS in patients with HNC receiving IMRT. Inflammation and aging markers may be candidate biomarkers for PNS. Understanding biological markers may produce targets for novel interventions.
Collapse
Affiliation(s)
- Yufen Lin
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, USA; Winship Cancer Institute, Emory University, Atlanta, USA
| | - Gang Peng
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, USA
| | - Deborah W Bruner
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, USA; Winship Cancer Institute, Emory University, Atlanta, USA; School of Medicine, Emory University, Atlanta, USA
| | - Andrew H Miller
- Winship Cancer Institute, Emory University, Atlanta, USA; School of Medicine, Emory University, Atlanta, USA
| | - Nabil F Saba
- Winship Cancer Institute, Emory University, Atlanta, USA; School of Medicine, Emory University, Atlanta, USA
| | - Kristin A Higgins
- Winship Cancer Institute, Emory University, Atlanta, USA; School of Medicine, Emory University, Atlanta, USA
| | - Dong M Shin
- Winship Cancer Institute, Emory University, Atlanta, USA; School of Medicine, Emory University, Atlanta, USA
| | - Henry Claussen
- Emory Integrated Computational Core, Emory University, Atlanta, USA
| | | | - Madelyn C Houser
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, USA
| | | | - Canhua Xiao
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, USA; Winship Cancer Institute, Emory University, Atlanta, USA.
| |
Collapse
|
49
|
Wang M, Gao Y, Chen H, Shen Y, Cheng J, Wang G. Bioinformatics strategies to identify differences in molecular biomarkers for ischemic stroke and myocardial infarction. Medicine (Baltimore) 2023; 102:e35919. [PMID: 37986378 PMCID: PMC10659606 DOI: 10.1097/md.0000000000035919] [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: 08/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 11/22/2023] Open
Abstract
Ischemic strokes (ISs) are commonly treated by intravenous thrombolysis using a recombinant tissue plasminogen activator; however, successful treatment can only occur within 3 hours after the stroke. Therefore, it is crucial to determine the causes and underlying molecular mechanisms, identify molecular biomarkers for early diagnosis, and develop precise preventive treatments for strokes. We aimed to clarify the differences in gene expression, molecular mechanisms, and drug prediction approaches between IS and myocardial infarction (MI) using comprehensive bioinformatics analysis. The pathogenesis of these diseases was explored to provide directions for future clinical research. The IS (GSE58294 and GSE16561) and MI (GSE60993 and GSE141512) datasets were downloaded from the Gene Expression Omnibus database. IS and MI transcriptome data were analyzed using bioinformatics methods, and the differentially expressed genes (DEGs) were screened. A protein-protein interaction network was constructed using the STRING database and visualized using Cytoscape, and the candidate genes with high confidence scores were identified using Degree, MCC, EPC, and DMNC in the cytoHubba plug-in. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were performed using the database annotation, visualization, and integrated discovery database. Network Analyst 3.0 was used to construct transcription factor (TF) - gene and microRNA (miRNA) - gene regulatory networks of the identified candidate genes. The DrugBank 5.0 database was used to identify gene-drug interactions. After bioinformatics analysis of IS and MI microarray data, 115 and 44 DEGS were obtained in IS and MI, respectively. Moreover, 8 hub genes, 2 miRNAs, and 3 TFs for IS and 8 hub genes, 13 miRNAs, and 2 TFs for MI were screened. The molecular pathology between IS and MI presented differences in terms of GO and KEGG enrichment pathways, TFs, miRNAs, and drugs. These findings provide possible directions for the diagnosis of IS and MI in the future.
Collapse
Affiliation(s)
- Min Wang
- School of Clinical Medicine, Dali University, Dali, Yunnan, P.R. China
| | - Yuan Gao
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Huaqiu Chen
- Xichang People’s Hospital, Xichang, Sichuan, P.R. China
| | - Ying Shen
- The First Hospital of Liangshan, Xichang, Sichuan, P.R. China
| | - Jianjie Cheng
- The First Affiliated Hospital of Dali University, Yunnan, P.R. China
| | - Guangming Wang
- School of Clinical Medicine, Dali University, Dali, Yunnan, P.R. China
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan, P.R. China
| |
Collapse
|
50
|
Gu Q, Zhou S, Chen C, Wang Z, Xu W, Zhang J, Wei S, Yang J, Chen H. CCL19: a novel prognostic chemokine modulates the tumor immune microenvironment and outcomes of cancers. Aging (Albany NY) 2023; 15:12369-12387. [PMID: 37944262 PMCID: PMC10683612 DOI: 10.18632/aging.205184] [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: 06/19/2023] [Accepted: 10/06/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND CCL19 is a chemokine involved in cancer research due to its important role in the tumor microenvironment (TME) and clinical relevance in cancers. This study aimed to analyze transcription expression, genomic alteration, association with tumor immune microenvironment of CCL19 expression and its prediction value for prognosis and responses to immunotherapy for patients with cancers. METHODS RNA sequencing data and corresponding clinicopathological information of a total of large-scale cancer patients were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases. Multiplex immunofluorescence (mIF) was implemented to identify differential infiltration of Treg, CD8+ T cells, and tumor-associated macrophages, while CCL19 immunohistochemistry was conducted on 182 breast cancer samples from a real-world cohort. RESULTS Based on large-scale multi-center survival analysis of cancer patients, we found the prognosis of patients with high CCL19 expression was prominently better than those with low CCL19 expression. For patients from multiple independent cohorts, suppressed CCL19 expression exerts significant progressive phenotype and apoptosis activity of cancers, especially in breast and ovarian cancer. Interestingly, anti-tumor immune cells, specifically the CD8+ T cells and macrophages, were clustered from TME by elevated CCL19 expression. Additionally, higher CCL19 levels reflected heightened immune activity and substantial heterogeneity. CONCLUSIONS In conclusion, our findings support the notion that elevated CCL19 expression is linked to favorable outcomes and enhanced anti-tumor immunity, characterized by increased CD8+ T cells within the TME. This suggests the potential of CCL19 as a prognostic marker, predictive biomarker for immunotherapy, therapeutic target of cancers.
Collapse
Affiliation(s)
- Qiang Gu
- Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong 226000, China
| | - Shifang Zhou
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cong Chen
- Department of Nursing, Fudan University Shanghai Cancer Center, Shanghai 201321, China
| | - Zhi Wang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Wenhao Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiarong Zhang
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shiyin Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Jianfeng Yang
- Department of Surgery, Shangnan Branch of Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200126, China
| | - Hongjing Chen
- Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong 226000, China
| |
Collapse
|