1
|
Franke A, Dahl S, Funck M, Bakker H, Garbers C, Lokau J. Interleukin-2 receptor α (IL-2Rα/CD25) shedding is differentially regulated by N- and O-glycosylation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119863. [PMID: 39427744 DOI: 10.1016/j.bbamcr.2024.119863] [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: 05/13/2024] [Revised: 08/30/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
The cytokine interleukin-2 (IL-2) is a critical regulator of immune responses, with an especially well-characterized role in regulating T-cell homeostasis. IL-2 signaling involves three distinct receptor subunits: the IL-2Rα (CD25), IL-2Rβ, and IL-2Rγ. The intracellular transduction of IL-2-induced signals is strictly dependent on IL-2Rβ and IL-2Rγ, while the IL-2Rα is not directly involved in signaling. Instead, it has the highest affinity towards IL-2 and is thus responsible for regulating the affinity of a cell for IL-2. In addition to the membrane-bound IL-2Rα, a soluble form of the receptor (sIL-2Rα) has been described, which is present in the blood of healthy individuals, increased under various pathological conditions, and able to bind IL-2 and thus modulate its function. The sIL-2Rα is generated by proteolytic cleavage of the membrane-bound receptor. Here, we analyze whether glycosylation of the IL-2Rα regulates its proteolysis. We find that constitutive IL-2Rα shedding is affected by glycosylation and discover distinct roles for N- and O-glycosylation. Furthermore, we show that induced shedding by the metalloproteases ADAM10 and ADAM17 is also differentially regulated by distinct types of glycans. Finally, we identify a specific role for an N-glycan at an exosite in ADAM17-mediated proteolysis that does not affect ADAM10, indicating distinct substrate recognition mechanisms. These results further the understanding of the mechanisms leading to sIL-2Rα generation, and thus offer the opportunity to specifically modulate the generation of the soluble receptor.
Collapse
Affiliation(s)
- Amelie Franke
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sophia Dahl
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Monika Funck
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Hans Bakker
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany; Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
2
|
Biwott K, Singh P, Baráth S, Nyariki JN, Hevessy Z, Bacso Z. Dynamic P-glycoprotein expression in early and late memory states of human CD8 + T cells and the protective role of ruxolitinib. Biomed Pharmacother 2025; 182:117780. [PMID: 39740391 DOI: 10.1016/j.biopha.2024.117780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
ABCB1/MDR-1/P-glycoprotein (Pgp) is an ABC transporter responsible for cancer cell multi-drug resistance. It is expressed in cytotoxic T lymphocytes (CTL). Eliminating sensitive cancer cells during high-dose chemotherapy can also damage immune cells. Our study aimed to assess which maturing human CD8 + CTL memory subsets may be affected based on their Pgp protein expression. In an in vitro CTL differentiation model system, we tracked the maturation of naive, effector, and memory cells and the expression of Pgp. This system involves co-culturing blood lymphocytes with proliferation-inhibited JY antigen-presenting B-lymphoblastoid cells expressing HLA-I A2. These JY-primed maturing CTLs were TCR-activated using beads, and the effect of the maturation-modifying JAK1/2 inhibitor ruxolitinib was examined. Multidimensional analysis identified six major CTL subsets: naive, young memory (Tym), stem cell memory (Tscm), central memory (Tcm), effector memory (Tem), and effectors (Te). These subsets were further divided into thirteen specific subsets: TymCD127 + , TymCD127-, Tscm, TcmCD95 + , TcmCD73 +CD95 + , TcmCD95+CD127 + , TcmPD1 + , TemCD95 + , TemraCD127 + , TemraCD127-, TeCD95 + , and TeCD73 +CD95 + . Pgp expression was detectable in naïve cells and dynamically changed across the thirteen identified subsets. Increased Pgp was detected in young memory T cells and in Tscm, TcmCD95 + , and TcmPD1 + human CTL subsets. Unlike other transiently appearing memory cells, the number of cells in these core Pgp-expressing memory subsets stabilized by the end of the contraction phase. Ruxolitinib treatment downregulated effector T-cell polarization while upregulating small memory subsets expressing Pgp. In conclusion, activation increased Pgp expression, whereas ruxolitinib treatment preserved small early and late memory subset core that primarily expressed Pgp.
Collapse
Affiliation(s)
- Kipchumba Biwott
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen 4032, Hungary; Department of Biochemistry and Biotechnology, Technical University of Kenya, Kenya.
| | - Parvind Singh
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | - Sándor Baráth
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | | | - Zsuzsanna Hevessy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen 4032, Hungary; Dean's office, Faculty of Pharmacy, University of Debrecen, Debrecen 4032, Hungary.
| |
Collapse
|
3
|
Chen R, Zou J, Chen J, Wang L, Kang R, Tang D. Immune aging and infectious diseases. Chin Med J (Engl) 2024; 137:3010-3049. [PMID: 39679477 PMCID: PMC11706578 DOI: 10.1097/cm9.0000000000003410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT The rise in global life expectancy has led to an increase in the older population, presenting significant challenges in managing infectious diseases. Aging affects the innate and adaptive immune systems, resulting in chronic low-grade inflammation (inflammaging) and immune function decline (immunosenescence). These changes would impair defense mechanisms, increase susceptibility to infections and reduce vaccine efficacy in older adults. Cellular senescence exacerbates these issues by releasing pro-inflammatory factors, further perpetuating chronic inflammation. Moreover, comorbidities, such as cardiovascular disease and diabetes, which are common in older adults, amplify immune dysfunction, while immunosuppressive medications further complicate responses to infections. This review explores the molecular and cellular mechanisms driving inflammaging and immunosenescence, focusing on genomic instability, telomere attrition, and mitochondrial dysfunction. Additionally, we discussed how aging-associated immune alterations influence responses to bacterial, viral, and parasitic infections and evaluated emerging antiaging strategies, aimed at mitigating these effects to improve health outcomes in the aging population.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Jiawang Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ling Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
4
|
Gupta N, Yadav AK, Verma PK, Srivastava M, Sahasrabuddhe AA, Dube A. Differential Immune Responses of Th1 Stimulatory Chimeric Antigens of Leishmania donovani in BALB/c Mice. ACS Infect Dis 2024; 10:4246-4257. [PMID: 39575598 DOI: 10.1021/acsinfecdis.4c00608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Visceral leishmaniasis (VL) is the third most severe infectious parasitic disease and is caused by the protozoan parasite Leishmania. To control the spread of the disease in endemic areas where the asymptomatic patients act as reservoirs as well as in nonendemic areas, an effective vaccine is indispensable. In this direction, we have developed three chimeric proteins by the combination of three already known Th1 stimulatory leishmanial antigens, i.e., enolase, aldolase, and triose phosphate isomerase (TPI). The newly developed chimeric proteins, i.e., enolase-aldolase, TPI-enolase, and aldolase-TPI along with BCG as an adjuvant were assessed and compared, examining humoral and cellular adaptive immune responses elicited in BALB/c mice. The three chimeric antigens exhibited differential immune responses shown by differences in Th1 and Th2 cytokine production in ex vivo stimulated splenocytes of immunized mice. It was observed that all three chimeric proteins are more immunogenic than their component proteins. However, while comparing the immune response of the three chimeric proteins, aldolase-TPI exhibited a better immunogenic (Th1-type) response, as evidenced by the highest IFN-γ production, a high IgG2a antibody isotype switching, a high % population of CD8+ and CD4+ T-cells, and a significantly high expression of iNOS2. Thus, the results suggest the potential of these chimeric antigens as strong immunogens that can be harnessed in vaccine development against VL.
Collapse
Affiliation(s)
- Niharika Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alok Kumar Yadav
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, India
| | - Pramod Kumar Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, India
| | - Mrigank Srivastava
- Molecular Immunology and Parasitology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, India
| | - Amogh Anant Sahasrabuddhe
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, India
| | - Anuradha Dube
- Molecular Immunology and Parasitology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, India
| |
Collapse
|
5
|
Larson EC, Ellis AL, Rodgers MA, Gubernat AK, Gleim JL, Moriarty RV, Balgeman AJ, de Menezes YT, Ameel CL, Fillmore DJ, Pergalske SM, Juno JA, Maiello P, Chishti HB, Lin PL, Godfrey DI, Kent SJ, Pellicci DG, Ndhlovu LC, O'Connor SL, Scanga CA. Transiently boosting Vγ9+Vδ2+ γδ T cells early in Mtb coinfection of SIV-infected juvenile macaques does not improve Mtb host resistance. Infect Immun 2024; 92:e0031324. [PMID: 39475292 PMCID: PMC11629613 DOI: 10.1128/iai.00313-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/01/2024] [Indexed: 11/06/2024] Open
Abstract
Children living with HIV have a higher risk of developing tuberculosis (TB), a disease caused by the bacterium Mycobacterium tuberculosis (Mtb). Gamma delta (γδ) T cells in the context of HIV/Mtb coinfection have been understudied in children despite in vitro evidence suggesting γδ T cells assist with Mtb control. We investigated whether boosting a specific subset of γδ T cells, phosphoantigen-reactive Vγ9+Vδ2+ cells, could improve TB outcome using a nonhuman primate model of pediatric HIV/Mtb coinfection. Juvenile Mauritian cynomolgus macaques (MCM), equivalent to 4- to 8-year-old children, were infected intravenously (i.v.) with SIV. After 6 months, MCM were coinfected with a low dose of Mtb and then randomized to receive zoledronate (ZOL), a drug that increases phosphoantigen levels, (n = 5; i.v.) at 3 and 17 days after Mtb accompanied by recombinant human IL-2 (s.c.) for 5 days following each ZOL injection. A similarly coinfected MCM group (n = 5) was injected with saline as a control. Vγ9+Vδ2+ γδ T cell frequencies spiked in the blood, but not airways, of ZOL+IL-2-treated MCM following the first dose, however, were refractory to the second dose. At necropsy 8 weeks after Mtb, ZOL+IL-2 treatment did not reduce pathology or bacterial burden. γδ T cell subset frequencies in granulomas did not differ between treatment groups. These data show that transiently boosting peripheral γδ T cells with ZOL+IL-2 soon after Mtb coinfection of SIV-infected MCM did not improve Mtb host defense.
Collapse
Affiliation(s)
- Erica C. Larson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy L. Ellis
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Abigail K. Gubernat
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Janelle L. Gleim
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ryan V. Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yonne T. de Menezes
- Department of Immunobiology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cassaundra L. Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Daniel J. Fillmore
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Skyler M. Pergalske
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Harris B. Chishti
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Philana Ling Lin
- Department of Pediatrics, UPMC’s Children’s Hospital of the University of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Centre Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Daniel G. Pellicci
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Lishomwa C. Ndhlovu
- Department of Medicine, Division of Infectious Disease, Weill Cornell Medicine, New York, New York, USA
| | - Shelby L. O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Yu F, Zhu Y, Li S, Hao L, Li N, Ye F, Jiang Z, Hu X. Dysfunction and regulatory interplay of T and B cells in chronic hepatitis B: immunotherapy and emerging antiviral strategies. Front Cell Infect Microbiol 2024; 14:1488527. [PMID: 39717542 PMCID: PMC11663751 DOI: 10.3389/fcimb.2024.1488527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
In the context of chronic hepatitis B virus (HBV) infection, the continuous replication of HBV within host hepatocytes is a characteristic feature. Rather than directly causing hepatocyte destruction, this replication leads to immune dysfunction and establishes a state of T-B immune tolerance. Successful clearance of the HBV virus is dependent on the close collaboration between humoral and cellular immunity. Humoral immunity, mediated by B-cell subpopulations, and cellular immunity, dominated by T-cell subpopulations show varying degrees of dysfunction during chronic hepatitis B (CHB). Notably, not all T- and B-cells produce positive immune responses. This review examine the most recent developments in the mutual regulation of T-B cells during chronic HBV infection. Our focus is on the prevailing immunotherapeutic strategies, such as T cell engineering, HBV-related vaccines, PD-1 inhibitors, and Toll-like receptor agonists. While nucleos(t)ide analogues (NUCs) and interferons have notable limitations, including inadequate viral suppression, drug resistance, and adverse reactions, several HBV entry inhibitors have shown promising clinical efficacy. To overcome the challenges posed by NUCs or monotherapy, the combination of immunotherapy and novel antiviral agents presents a promising avenue for future CHB treatment and potential cure.
Collapse
Affiliation(s)
- Fei Yu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yue Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shenghao Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Liyuan Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Na Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fanghang Ye
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhi Jiang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Sun Y, Liu J, Zhan D, Wei J, XianShi L, Zhang R, Duan C, Zhang D, Tang X, Lin T, Li L, Lai X. Depletion of Tregs from CD4 + CAR-T cells enhances the tumoricidal effect of CD8 + CAR-T cells in anti-CD19 CAR-T therapy. FEBS J 2024. [PMID: 39632397 DOI: 10.1111/febs.17326] [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: 11/18/2023] [Revised: 07/18/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, which targets CD19 for hematological malignancies, represents a breakthrough in cancer immunotherapy. However, some patients may develop resistance to CAR-T treatment, underscoring the importance of optimizing CAR-T design to enhance responsiveness. Here, we investigated the impact of different subpopulations in anti-CD19 CAR-T cells on the tumoricidal effect. Different populations of anti-CD19 CAR-T cells were isolated by magnetic-activated cell sorting (MACS). Their lytic activities on the acute lymphocytic leukemia cell line SUP-B15 and diffuse large B-cell lymphoma EB-3 cell line were examined in a co-culture system. The anti-tumorigenic outcome of different CAR-T cell compositions was evaluated in a xenograft mouse model of EB-3 cells. CD8+CAR-T cells exhibited the most potent tumoricidal activity against SUP-B15 and EB-3 cells. Additionally, CD4+ T helper cells enhanced the lytic effects of CD8+ CAR-T cells by increasing the availability of interleukin-2 (IL-2). Depleting CD25+Treg (T regulatory) cells from CD4+CAR-T population further augmented the tumoricidal activity of CD8+CAR-T cells by preventing IL-2 deprivation. Consistently, in vivo experiments demonstrated that the CD4+CD25+ Treg population dampened the antitumor activity of CD8+CAR-T cells, while depletion of Tregs from CD4+CAR-T cells enhanced the tumoricidal effect. These findings emphasize the potential role of CAR Treg cells in therapeutic resistance, suggesting that the depletion of Tregs in the anti-CD19 CAR-T population may serve as a strategy to augment the anticancer effect of CD8+CAR-T cells.
Collapse
Affiliation(s)
- Yunyan Sun
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Jinyan Liu
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Dong Zhan
- Department of Human Anatomy and Histology & Embrology, School of Basic Medical Sciences, Kunming Medical University, China
| | - Jia Wei
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Li XianShi
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Rui Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Ci Duan
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Disi Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Xiaorong Tang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Tuo Lin
- Yunnan College of Business Management, Kunming, China
| | - Limei Li
- Yunnan College of Business Management, Kunming, China
| | - Xun Lai
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| |
Collapse
|
8
|
Jamison BL, Lawrance M, Wang CJ, DeBerg HA, Ziegler LJ, Sansom DM, Gavin MA, Walker LSK, Campbell DJ. An IL-2 mutein increases regulatory T cell suppression of dendritic cells via IL-10 and CTLA-4 to promote T cell anergy. Cell Rep 2024; 43:114938. [PMID: 39488830 PMCID: PMC11602548 DOI: 10.1016/j.celrep.2024.114938] [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: 12/09/2023] [Revised: 08/15/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Interleukin-2 (IL-2) variants with increased CD25 dependence that selectively expand Foxp3+ regulatory T (TR) cells are in clinical trials for treating inflammatory diseases. Using an Fc-fused IL-2 mutein (Fc.IL-2 mutein) we developed that prevents diabetes in non-obese diabetic (NOD) mice, we show that Fc.IL-2 mutein induced an activated TR population with elevated proliferation, a transcriptional program associated with Stat5- and T cell receptor-dependent gene modules, and high IL-10 and CTLA-4 expression. Increased IL-10 signaling limited surface major histocompatibility complex class II upregulation during conventional dendritic cell (cDC) maturation, while increased CTLA-4-dependent transendocytosis led to the transfer of CD80 and CD86 co-stimulatory ligands from maturing cDCs to TR cells. In NOD mice, Fc.IL-2 mutein treatment promoted the suppression of cDCs in the inflamed pancreas and pancreatic lymph nodes, resulting in T cell anergy. Thus, IL-2 mutein-expanded TR cells have enhanced functional properties and restrict cDC function, offering promise for targeted immunotherapy use in autoimmune disease.
Collapse
Affiliation(s)
- Braxton L Jamison
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Matthew Lawrance
- Center for Systems Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Chun Jing Wang
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London NW3 2PP, UK
| | - Hannah A DeBerg
- Center for Systems Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Lauren J Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - David M Sansom
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London NW3 2PP, UK
| | - Marc A Gavin
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London NW3 2PP, UK
| | - Daniel J Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98126, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 981098, USA.
| |
Collapse
|
9
|
Kunimitsu M, Minematsu T, Koudounas S, Sanada H, Nakagami G. Relationship Between Dysbiotic Wound Microbiota and Critical Colonization: Involvement of FOXP3-Positive Cells in Rats. Ann Plast Surg 2024; 93:617-623. [PMID: 39356291 DOI: 10.1097/sap.0000000000004092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
INTRODUCTION Detection of critical colonization is gaining importance in wound management, but its pathophysiology remains unclear. We previously clarified that a dysbiotic wound microbiota differing from skin commensal microbiota may be involved in critical colonization and that such wounds contain fewer Forkhead box protein P3 (FOXP3)-positive cells in the tissue. However, it is not clear whether FOXP3-positive cells contribute to the development of critical colonization. Here, we examined whether inhibition of FOXP3-positive cell could induce critical colonization when the commensal microbiota was present in the wounds. METHODS Sprague-Dawley rats were administered FK506 or vehicle to inhibit differentiation into FOXP3-positive cells. Full-thickness wounds were made on the dorsal skin and inoculated with bacterial solution (dysbiosis group) or Luria-Bertani medium (commensal group). A bacterial solution was prepared by anaerobically culturing bacteria from the skin of donor rats on an artificial dermis in Luria-Bertani medium for 72 hours. Tissues were collected on day 4 postwounding for histological evaluation. RESULTS After microbiota transplantation, excessive inflammation occurred in the FK506 + commensal group. In contrast, wounds with transplanted dysbiotic microbiota showed the same level of neutrophil infiltration, regardless of FK506 administration. Furthermore, the wound area was larger in the FK506 + commensal group than in the vehicle + commensal group on day 4 postwounding ( P = 0.01). This area was also significantly larger in both the vehicle + dysbiosis ( P = 0.01) and FK506 + dysbiosis groups ( P = 0.03) than in the vehicle + commensal group. CONCLUSIONS This study has shown that dysbiosis may be at least related to developing critical colonization, and the results suggest that FOXP3-positive cells are involved in this process. Our study may contribute to establishing new interventions that prevent critical colonization by correcting wound microbiota.
Collapse
Affiliation(s)
| | | | - Sofoklis Koudounas
- Department of Skincare Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
10
|
Cao H, Xiao J, Baylink DJ, Nguyen V, Shim N, Lee J, Mallari DJR, Wasnik S, Mirshahidi S, Chen CS, Abdel-Azim H, Reeves ME, Xu Y. Development of a Competitive Nutrient-Based T-Cell Immunotherapy Designed to Block the Adaptive Warburg Effect in Acute Myeloid Leukemia. Biomedicines 2024; 12:2250. [PMID: 39457563 PMCID: PMC11504511 DOI: 10.3390/biomedicines12102250] [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: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: T-cell-based adoptive cell therapies have emerged at the forefront of cancer immunotherapies; however, failed long-term survival and inevitable exhaustion of transplanted T lymphocytes in vivo limits clinical efficacy. Leukemia blasts possess enhanced glycolysis (Warburg effect), exploiting their microenvironment to deprive nutrients (e.g., glucose) from T cells, leading to T-cell dysfunction and leukemia progression. Methods: Thus, we explored whether genetic reprogramming of T-cell metabolism could improve their survival and empower T cells with a competitive glucose-uptake advantage against blasts and inhibit their uncontrolled proliferation. Results: Here, we discovered that high-glucose concentration reduced the T-cell expression of glucose transporter GLUT1 (SLC2A1) and TFAM (mitochondrion transcription factor A), an essential transcriptional regulator of mitochondrial biogenesis, leading to their impaired expansion ex vivo. To overcome the glucose-induced genetic deficiency in metabolism, we engineered T cells with lentiviral overexpression of SLC2A1 and/or TFAM transgene. Multi-omics analyses revealed that metabolic reprogramming promoted T-cell proliferation by increasing IL-2 release and reducing exhaustion. Moreover, the engineered T cells competitively deprived glucose from allogenic blasts and lessened leukemia burden in vitro. Conclusions: Our findings propose a novel T-cell immunotherapy that utilizes a dual strategy of starving blasts and cytotoxicity for preventing uncontrolled leukemia proliferation.
Collapse
Affiliation(s)
- Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Nathan Shim
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jae Lee
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Dave J. R. Mallari
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Saied Mirshahidi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Biospecimen Laboratory, Department of Medicine and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hisham Abdel-Azim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Transplant and Cell Therapy, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Hematology and Oncology, Department of Pediatrics, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
11
|
Speeckaert R, Belpaire A, Lambert J, Speeckaert M, van Geel N. Th Pathways in Immune-Mediated Skin Disorders: A Guide for Strategic Treatment Decisions. Immune Netw 2024; 24:e33. [PMID: 39513029 PMCID: PMC11538609 DOI: 10.4110/in.2024.24.e33] [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: 04/11/2024] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 11/15/2024] Open
Abstract
In recent years, there have been significant breakthroughs in the identification of immunological components of skin diseases and in the development of immunomodulatory drugs. Novel therapies create exciting prospects for personalized care. This article provides an overview of the role played by Th1, Th2, Th17, and follicular Th pathways in the most common skin diseases. Additionally, it elucidates the impact of current and upcoming treatments on each of these signaling cascades. Skin diseases predominantly influenced by a single dominant Th pathway such as psoriasis and atopic dermatitis are well-suited for biologics. However, in many other disorders a complex interplay between different immune pathways exists. This can lead to inconsistent efficacy of biologics based on individual patient profiles. In case of activation of several Th pathways, it may be more suitable to consider conventional therapies or JAK inhibitors. Increasing immunological insights have transitioned from laboratory research to practical applications, a trend that is expected to continue growing in the future.
Collapse
Affiliation(s)
| | - Arno Belpaire
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jo Lambert
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Marijn Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Nanja van Geel
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
12
|
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Japp AS, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Prak ETL, Kaestner KH, Naji A, Betts MR. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590798. [PMID: 39345402 PMCID: PMC11429609 DOI: 10.1101/2024.04.23.590798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. Studies of human T1D immunobiology have predominantly focused on circulating immune cells in the blood, while mouse models suggest diabetogenic lymphocytes primarily reside in pancreas-draining lymph nodes (pLN). A comprehensive study of immune cells in human T1D was conducted using pancreas draining lymphatic tissues, including pLN and mesenteric lymph nodes, and the spleen from non-diabetic control, β cell autoantibody positive non-diabetic (AAb+), and T1D organ donors using complementary approaches of high parameter flow cytometry and CITEseq. Immune perturbations suggestive of a proinflammatory environment were specific for T1D pLN and AAb+ pLN. In addition, certain immune populations correlated with high T1D genetic risk independent of disease state. These datasets form an extensive resource for profiling human lymphatic tissue immune cells in the context of autoimmunity and T1D.
Collapse
Affiliation(s)
- Gregory J Golden
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vincent H Wu
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jacob T Hamilton
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
| | - Alberto Sada Japp
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Maria Betina Pampena
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - James J Knox
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jay S Gardner
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael R Betts
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Maurice D, Costello P, Diring J, Gualdrini F, Frederico B, Treisman R. IL-2 delivery to CD8 + T cells during infection requires MRTF/SRF-dependent gene expression and cytoskeletal dynamics. Nat Commun 2024; 15:7956. [PMID: 39261466 PMCID: PMC11391060 DOI: 10.1038/s41467-024-52230-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Paracrine IL-2 signalling drives the CD8 + T cell expansion and differentiation that allow protection against viral infections, but the underlying molecular events are incompletely understood. Here we show that the transcription factor SRF, a master regulator of cytoskeletal gene expression, is required for effective IL-2 signalling during L. monocytogenes infection. Acting cell-autonomously with its actin-regulated cofactors MRTF-A and MRTF-B, SRF is dispensible for initial TCR-mediated CD8+ T cell proliferation, but is required for sustained IL-2 dependent CD8+ effector T cell expansion, and persistence of memory cells. Following TCR activation, Mrtfab-null CD8+ T cells produce IL-2 normally, but homotypic clustering is impaired both in vitro and in vivo. Expression of cytoskeletal structural and regulatory genes, most notably actins, is defective in Mrtfab-null CD8+ T cells. Activation-induced cell clustering in vitro requires F-actin assembly, and Mrtfab-null cell clusters are small, contain less F-actin, and defective in IL-2 retention. Clustering of Mrtfab-null cells can be partially restored by exogenous actin expression. IL-2 mediated CD8+ T cell proliferation during infection thus depends on the control of cytoskeletal dynamics and actin gene expression by MRTF-SRF signalling.
Collapse
Affiliation(s)
- Diane Maurice
- Signalling and transcription Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Autoimmunity Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Patrick Costello
- Signalling and transcription Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jessica Diring
- Signalling and transcription Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Francesco Gualdrini
- Signalling and transcription Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- European Institute of Oncology (IEO), Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, 20139, Italy
| | - Bruno Frederico
- Immunobiology Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Early Oncology, R&D, AstraZeneca, Cambridge, UK
| | - Richard Treisman
- Signalling and transcription Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
14
|
van der Mescht MA, de Beer Z, Steel HC, Anderson R, Masenge A, Moore PL, Bastard P, Casanova JL, Abdullah F, Ueckermann V, Rossouw TM. Aberrant innate immune profile associated with COVID-19 mortality in Pretoria, South Africa. Clin Immunol 2024; 266:110323. [PMID: 39029640 DOI: 10.1016/j.clim.2024.110323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
The African continent reported the least number of COVID-19 cases and deaths of all the continents, although the exact reasons for this are still unclear. In addition, little is known about the immunological profiles associated with COVID-19 mortality in Africa. The present study compared clinical and immunological parameters, as well as treatment outcomes in patients admitted with COVID-19 in Pretoria, South Africa, to determine if these parameters correlated with mortality in this population. The in-hospital mortality rate for the cohort was 15.79%. The mortality rate in people living with HIV (PLWH) was 10.81% and 17.16% in people without HIV (p = 0.395). No differences in age (p = 0.099), gender (p = 0.127) or comorbidities were found between deceased patients and those who survived. All four of the PLWH who died had a CD4+ T-cell count <200 cells/mm3, a significantly higher HIV viral load than those who survived (p = 0.009), and none were receiving antiretroviral therapy. Seven of 174 (4%) patients had evidence of auto-antibodies neutralizing Type 1 interferons (IFNs). Two of the them died, and their presence was significantly associated with mortality (p = 0.042). In the adjusted model, the only clinical parameters associated with mortality were: higher fraction of inspired oxygen (FiO2) (OR: 3.308, p = 0.011) indicating a greater need for oxygen, high creatinine (OR: 4.424, p = 0.001) and lower platelet counts (OR: 0.203, p = 0.009), possibly secondary to immunothrombosis. Overall, expression of the co-receptor CD86 (p = 0.021) on monocytes and percentages of CD8+ effector memory 2 T-cells (OR: 0.45, p = 0.027) was lower in deceased patients. Decreased CD86 expression impairs the development and survival of effector memory T-cells. Deceased patients had higher concentrations of RANTES (p = 0.003), eotaxin (p = 0.003) and interleukin (IL)-8 (p < 0.001), all involved in the activation and recruitment of innate immune cells. They also had lower concentrations of transforming growth factor (TGF)-β1 (p = 0.40), indicating an impaired anti-inflammatory response. The immunological profile associated with COVID-19 mortality in South Africa points to the role of aberrate innate immune responses.
Collapse
Affiliation(s)
- Mieke A van der Mescht
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Zelda de Beer
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Tshwane District Hospital, Pretoria, South Africa
| | - Helen C Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Andries Masenge
- Department of Statistics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Penny L Moore
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa; National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, New York, NY, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, New York, NY, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique-Hôpitaux de Paris (AP-HP), Paris, France; Howard Hughes Medical Institute, New York, NY, USA
| | - Fareed Abdullah
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa; Office of AIDS and TB Research, South African Medical Research Council, Pretoria, South Africa; Department of Public Health Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Veronica Ueckermann
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Theresa M Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
15
|
Larson EC, Ellis AL, Rodgers MA, Gubernat AK, Gleim JL, Moriarty RV, Balgeman AJ, de Menezes YT, Ameel CL, Fillmore DJ, Pergalske SM, Juno JA, Maiello P, Chishti HB, Lin PL, Godfrey DI, Kent SJ, Pellicci DG, Ndhlovu LC, O’Connor SL, Scanga CA. Transiently boosting Vγ9+Vδ2+ γδ T cells early in Mtb coinfection of SIV-infected juvenile macaques does not improve Mtb host resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604654. [PMID: 39091843 PMCID: PMC11291075 DOI: 10.1101/2024.07.22.604654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Children living with HIV have a higher risk of developing tuberculosis (TB), a disease caused by the bacterium Mycobacterium tuberculosis (Mtb). Gamma delta (γδ) T cells in the context of HIV/Mtb coinfection have been understudied in children, despite in vitro evidence suggesting γδ T cells assist with Mtb control. We investigated whether boosting a specific subset of γδ T cells, phosphoantigen-reactive Vγ9+Vδ2+ cells, could improve TB outcome using a nonhuman primate model of pediatric HIV/Mtb coinfection. Juvenile Mauritian cynomolgus macaques (MCM), equivalent to 4-8-year-old children, were infected intravenously (i.v.) with SIV. After 6 months, MCM were coinfected with a low dose of Mtb and then randomized to receive zoledronate (ZOL), a drug that increases phosphoantigen levels, (n=5; i.v.) at 3- and 17- days after Mtb accompanied by recombinant human IL-2 (s.c.) for 5 days following each ZOL injection. A similarly coinfected MCM group (n=5) was injected with saline as a control. Vγ9+Vδ2+ γδ T cell frequencies spiked in the blood, but not airways, of ZOL+IL-2-treated MCM following the first dose, however, were refractory to the second dose. At necropsy eight weeks after Mtb, ZOL+IL-2 treatment did not reduce pathology or bacterial burden. γδ T cell subset frequencies in granulomas did not differ between treatment groups. These data show that transiently boosting peripheral γδ T cells with ZOL+IL-2 soon after Mtb coinfection of SIV-infected MCM did not improve Mtb host defense.
Collapse
Affiliation(s)
- Erica C. Larson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy L. Ellis
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Wisconsin, USA
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Abigail K. Gubernat
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Janelle L. Gleim
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ryan V. Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Wisconsin, USA
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Wisconsin, USA
| | - Yonne T. de Menezes
- Department of Immunobiology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cassaundra L. Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Daniel J. Fillmore
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Skyler M. Pergalske
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Harris B. Chishti
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Philana Ling Lin
- Department of Pediatrics, UPMC’s Children’s Hospital of the University of Pittsburgh of UPMC, Pittsburgh, PA
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Centre Clinical School, Monash University, Melbourne, VIC, Australia
| | - Daniel G. Pellicci
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Lishomwa C. Ndhlovu
- Department of Medicine, Division of Infectious Disease, Weill Cornell Medicine, New York, New York, USA
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin - Madison, Wisconsin, USA
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Litjens NHR, van der List ACJ, Klepper M, Reijerkerk D, Prevoo F, Betjes MGH. Older age is associated with a distinct and marked reduction of functionality of both alloreactive CD4+ and CD8+ T cells. Front Immunol 2024; 15:1406716. [PMID: 39044836 PMCID: PMC11263037 DOI: 10.3389/fimmu.2024.1406716] [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: 03/25/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Older recipient age is associated with a significant decreased risk for rejection after kidney transplantation which is incompletely understood. Methods In a longitudinal study, circulating alloreactive T cells were assessed of young (≤45 years) and older (≥55 years) stable kidney transplant recipients. Alloreactive T-cells were identified by CD137-expression and phenotype, cytokine producing and proliferative capacity, were evaluated using multiparameter flowcytometry. Results The results show that before transplantation frequencies of alloreactive CD4+ and CD8+ T-cells in older KT-recipients are significantly higher and shifted towards an effector memory-phenotype. However, the frequency of polyfunctional (≥2 pro-inflammatory cytokines) CD4+ T-cells was significantly lower and less IL2 was produced. The frequency of polyfunctional alloreactive CD4+ T-cells and proliferation of alloreactive T-cells donor-specifically declined after transplantation reaching a nadir at 12 months after transplantation, irrespective of age. A striking difference was observed for the proliferative response of alloreactive CD8+ T-cells. This was not only lower in older compared to younger recipients but could also not be restored by exogenous IL2 or IL15 in the majority of older recipients while the response to polyclonal stimulation was unaffected. Conclusion In conclusion, older age is associated with a distinct and marked reduction of functionality of both alloreactive CD4+ and CD8+ T-cells.
Collapse
|
17
|
He T, Hu C, Li S, Fan Y, Xie F, Sun X, Jiang Q, Chen W, Jia Y, Li W. The role of CD8 + T-cells in colorectal cancer immunotherapy. Heliyon 2024; 10:e33144. [PMID: 39005910 PMCID: PMC11239598 DOI: 10.1016/j.heliyon.2024.e33144] [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: 10/27/2023] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Immunotherapy has been an advanced and effective approach to treating various types of solid tumors in recent years, and the most successful strategy is immune checkpoint inhibitors (ICIs), which have shown beneficial effects in patients with colorectal cancer (CRC). Drug resistance to ICIs is usually associated with CD8+ T-cells targeting tumor antigens; thus, CD8+ T-cells play an important role in immunotherapy. Unfortunately, Under continuous antigen stimulation, tumor microenvironment(TME), hypoxia and other problems it leads to insufficient infiltration of CD8+ T-cells, low efficacy and mechanism exhaustion, which have become obstacles to immunotherapy. Thus, this article describes the relationship between CRC and the immune system, focuses on the process of CD8+ T-cells production, activation, transport, killing, and exhaustion, and expounds on related mechanisms leading to CD8+ T-cells exhaustion. Finally, this article summarizes the latest strategies and methods in recent years, focusing on improving the infiltration, efficacy, and exhaustion of CD8+ T-cells, which may help to overcome the barriers to immunotherapy.
Collapse
Affiliation(s)
- Tao He
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Chencheng Hu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Shichao Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yao Fan
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Fei Xie
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Xin Sun
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Qingfeng Jiang
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Weidong Chen
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yingtian Jia
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Wusheng Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| |
Collapse
|
18
|
Kemp F, Braverman EL, Byersdorfer CA. Fatty acid oxidation in immune function. Front Immunol 2024; 15:1420336. [PMID: 39007133 PMCID: PMC11240245 DOI: 10.3389/fimmu.2024.1420336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/31/2024] [Indexed: 07/16/2024] Open
Abstract
Cellular metabolism is a crucial determinant of immune cell fate and function. Extensive studies have demonstrated that metabolic decisions influence immune cell activation, differentiation, and cellular capacity, in the process impacting an organism's ability to stave off infection or recover from injury. Conversely, metabolic dysregulation can contribute to the severity of multiple disease conditions including autoimmunity, alloimmunity, and cancer. Emerging data also demonstrate that metabolic cues and profiles can influence the success or failure of adoptive cellular therapies. Importantly, immunometabolism is not one size fits all; and different immune cell types, and even subdivisions within distinct cell populations utilize different metabolic pathways to optimize function. Metabolic preference can also change depending on the microenvironment in which cells are activated. For this reason, understanding the metabolic requirements of different subsets of immune cells is critical to therapeutically modulating different disease states or maximizing cellular function for downstream applications. Fatty acid oxidation (FAO), in particular, plays multiple roles in immune cells, providing both pro- and anti-inflammatory effects. Herein, we review the major metabolic pathways available to immune cells, then focus more closely on the role of FAO in different immune cell subsets. Understanding how and why FAO is utilized by different immune cells will allow for the design of optimal therapeutic interventions targeting this pathway.
Collapse
Affiliation(s)
| | | | - Craig A. Byersdorfer
- Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular Therapies, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
19
|
Stoffel NU, Drakesmith H. Effects of Iron Status on Adaptive Immunity and Vaccine Efficacy: A Review. Adv Nutr 2024; 15:100238. [PMID: 38729263 PMCID: PMC11251406 DOI: 10.1016/j.advnut.2024.100238] [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: 01/24/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Vaccines can prevent infectious diseases, but their efficacy varies, and factors impacting vaccine effectiveness remain unclear. Iron deficiency is the most common nutrient deficiency, affecting >2 billion individuals. It is particularly common in areas with high infectious disease burden and in groups that are routinely vaccinated, such as infants, pregnant women, and the elderly. Recent evidence suggests that iron deficiency and low serum iron (hypoferremia) not only cause anemia but also may impair adaptive immunity and vaccine efficacy. A report of human immunodeficiency caused by defective iron transport underscored the necessity of iron for adaptive immune responses and spurred research in this area. Sufficient iron is essential for optimal production of plasmablasts and IgG responses by human B-cells in vitro and in vivo. The increased metabolism of activated lymphocytes depends on the high-iron acquisition, and hypoferremia, especially when occurring during lymphocyte expansion, adversely affects multiple facets of adaptive immunity, and may lead to prolonged inhibition of T-cell memory. In mice, hypoferremia suppresses the adaptive immune response to influenza infection, resulting in more severe pulmonary disease. In African infants, anemia and/or iron deficiency at the time of vaccination predict decreased response to diphtheria, pertussis, and pneumococcal vaccines, and response to measles vaccine may be increased by iron supplementation. In this review, we examine the emerging evidence that iron deficiency may limit adaptive immunity and vaccine responses. We discuss the molecular mechanisms and evidence from animal and human studies, highlight important unknowns, and propose a framework of key research questions to better understand iron-vaccine interactions.
Collapse
Affiliation(s)
- Nicole U Stoffel
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.
| | - Hal Drakesmith
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
De George DJ, Jhala G, Selck C, Trivedi P, Brodnicki TC, Mackin L, Kay TW, Thomas HE, Krishnamurthy B. Altering β Cell Antigen Exposure to Exhausted CD8+ T Cells Prevents Autoimmune Diabetes in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1658-1669. [PMID: 38587315 DOI: 10.4049/jimmunol.2300785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
Chronic destruction of insulin-producing pancreatic β cells by T cells results in autoimmune diabetes. Similar to other chronic T cell-mediated pathologies, a role for T cell exhaustion has been identified in diabetes in humans and NOD mice. The development and differentiation of exhausted T cells depends on exposure to Ag. In this study, we manipulated β cell Ag presentation to target exhausted autoreactive T cells by inhibiting IFN-γ-mediated MHC class I upregulation or by ectopically expressing the β cell Ag IGRP under the MHC class II promotor in the NOD8.3 model. Islet PD-1+TIM3+CD8+ (terminally exhausted [TEX]) cells were primary producers of islet granzyme B and CD107a, suggestive of cells that have entered the exhaustion program yet maintained cytotoxic capacity. Loss of IFN-γ-mediated β cell MHC class I upregulation correlated with a significant reduction in islet TEX cells and diabetes protection in NOD8.3 mice. In NOD.TII/8.3 mice with IGRP expression induced in APCs, IGRP-reactive T cells remained exposed to high levels of IGRP in the islets and periphery. Consequently, functionally exhausted TEX cells, with reduced granzyme B expression, were significantly increased in these mice and this correlated with diabetes protection. These results indicate that intermediate Ag exposure in wild-type NOD8.3 islets allows T cells to enter the exhaustion program without becoming functionally exhausted. Moreover, Ag exposure can be manipulated to target this key cytotoxic population either by limiting the generation of cytotoxic TIM3+ cells or by driving their functional exhaustion, with both resulting in diabetes protection.
Collapse
Affiliation(s)
- David J De George
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Gaurang Jhala
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Claudia Selck
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Prerak Trivedi
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Thomas C Brodnicki
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Leanne Mackin
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
| | - Thomas W Kay
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Helen E Thomas
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Balasubramanian Krishnamurthy
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| |
Collapse
|
21
|
Zheng X, Yang R, Zhao Y, Zhang Y, Yuan G, Li W, Xiao Z, Dong X, Ma M, Guo Y, Wang W, Zhao X, Yang H, Qiu S, Peng Z, Liu A, Yu S, Zhang Y. Alum/CpG adjuvant promotes immunogenicity of inactivated SARS-CoV-2 Omicron vaccine through enhanced humoral and cellular immunity. Virology 2024; 594:110050. [PMID: 38479071 DOI: 10.1016/j.virol.2024.110050] [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: 08/21/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/09/2024]
Abstract
The SARS-CoV-2 Omicron variant, which was classified as a variant of concern (VOC) by the World Health Organization on 26 November 2021, has attracted worldwide attention for its high transmissibility and immune evasion ability. The existing COVID-19 vaccine has been shown to be less effective in preventing Omicron variant infection and symptomatic infection, which brings new challenges to vaccine development and application. Here, we evaluated the immunogenicity and safety of an Omicron variant COVID-19 inactivated vaccine containing aluminum and CpG adjuvants in a variety of animal models. The results showed that the vaccine candidate could induce high levels of neutralizing antibodies against the Omicron variant virus and binding antibodies, and significantly promoted cellular immune response. Meanwhile, the vaccine candidate was safe. Therefore, it provided more foundation for the development of aluminum and CpG as a combination adjuvant in human vaccines.
Collapse
Affiliation(s)
- Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Rong Yang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yuxiu Zhao
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yadan Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Guangying Yuan
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Weidong Li
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Zhuangzhuang Xiao
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Xiaofei Dong
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Meng Ma
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yancen Guo
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Wei Wang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Xue Zhao
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Hongqiang Yang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Shaoting Qiu
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Zheng Peng
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Ankang Liu
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Shouzhi Yu
- Beijing Institute of Biological Products Company Limited, Beijing, China.
| | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, China; China National Biotec Group Company Limited, Beijing, China.
| |
Collapse
|
22
|
Zhang R, Zhao Y, Chen X, Zhuang Z, Li X, Shen E. Low-dose IL-2 therapy in autoimmune diseases: An update review. Int Rev Immunol 2024; 43:113-137. [PMID: 37882232 DOI: 10.1080/08830185.2023.2274574] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
Regulatory T (Treg) cells are essential for maintaining self-immune tolerance. Reduced numbers or functions of Treg cells have been involved in the pathogenesis of various autoimmune diseases and allograft rejection. Therefore, the approaches that increase the pool or suppressive function of Treg cells in vivo could be a general strategy to treat different autoimmune diseases and allograft rejection. Interleukin-2 (IL-2) is essential for the development, survival, maintenance, and function of Treg cells, constitutively expressing the high-affinity receptor of IL-2 and sensitive response to IL-2 in vivo. And low-dose IL-2 therapy in vivo could restore the imbalance between autoimmune response and self-tolerance toward self-tolerance via promoting Treg cell expansion and inhibiting follicular helper T (Tfh) and IL-17-producing helper T (Th17) cell differentiation. Currently, low-dose IL-2 treatment is receiving extensive attention in autoimmune disease and transplantation treatment. In this review, we summarize the biology of IL-2/IL-2 receptor, the mechanisms of low-dose IL-2 therapy in autoimmune diseases, the application in the progress of different autoimmune diseases, including Systemic Lupus Erythematosus (SLE), Type 1 Diabetes (T1D), Rheumatoid Arthritis (RA), Autoimmune Hepatitis (AIH), Alopecia Areata (AA), Immune Thrombocytopenia (ITP) and Chronic graft-versus-host-disease (GVHD). We also discuss the future directions to optimize low-dose IL-2 treatments.
Collapse
Affiliation(s)
- Ruizhi Zhang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Yuyang Zhao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Xiangming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Zhuoqing Zhuang
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Erxia Shen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
23
|
Cao X, Fu YX, Peng H. Promising Cytokine Adjuvants for Enhancing Tuberculosis Vaccine Immunity. Vaccines (Basel) 2024; 12:477. [PMID: 38793728 PMCID: PMC11126114 DOI: 10.3390/vaccines12050477] [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: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (M. tuberculosis), remains a formidable global health challenge, affecting a substantial portion of the world's population. The current tuberculosis vaccine, bacille Calmette-Guérin (BCG), offers limited protection against pulmonary tuberculosis in adults, underscoring the critical need for innovative vaccination strategies. Cytokines are pivotal in modulating immune responses and have been explored as potential adjuvants to enhance vaccine efficacy. The strategic inclusion of cytokines as adjuvants in tuberculosis vaccines holds significant promise for augmenting vaccine-induced immune responses and strengthening protection against M. tuberculosis. This review delves into promising cytokines, such as Type I interferons (IFNs), Type II IFN, interleukins such as IL-2, IL-7, IL-15, IL-12, and IL-21, alongside the use of a granulocyte-macrophage colony-stimulating factor (GM-CSF) as an adjuvant, which has shown effectiveness in boosting immune responses and enhancing vaccine efficacy in tuberculosis models.
Collapse
Affiliation(s)
- Xuezhi Cao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China;
- Guangzhou National Laboratory, Bio-Island, Guangzhou 510005, China
| | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hua Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China;
- Guangzhou National Laboratory, Bio-Island, Guangzhou 510005, China
| |
Collapse
|
24
|
Wang S, Prieux M, de Bernard S, Dubois M, Laubreton D, Djebali S, Zala M, Arpin C, Genestier L, Leverrier Y, Gandrillon O, Crauste F, Jiang W, Marvel J. Exogenous IL-2 delays memory precursors generation and is essential for enhancing memory cells effector functions. iScience 2024; 27:109411. [PMID: 38510150 PMCID: PMC10952031 DOI: 10.1016/j.isci.2024.109411] [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: 07/27/2023] [Revised: 11/27/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
To investigate the impact of paracrine IL-2 signals on memory precursor (MP) cell differentiation, we activated CD8 T cell in vitro in the presence or absence of exogenous IL-2 (ex-IL-2). We assessed memory differentiation by transferring these cells into virus-infected mice. Both conditions generated CD8 T cells that participate in the ongoing response and gave rise to similar memory cells. Nevertheless, when transferred into a naive host, T cells activated with ex-IL-2 generated a higher frequency of memory cells displaying increased functional memory traits. Single-cell RNA-seq analysis indicated that without ex-IL-2, cells rapidly acquire an MP signature, while in its presence they adopted an effector signature. This was confirmed at the protein level and in a functional assay. Overall, ex-IL-2 delays the transition into MP cells, allowing the acquisition of effector functions that become imprinted in their progeny. These findings may help to optimize the generation of therapeutic T cells.
Collapse
Affiliation(s)
- Shaoying Wang
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Margaux Prieux
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Laboratoire de Biologie et de Modélisation de la Cellule, Université de Lyon, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Lyon, France
| | | | - Maxence Dubois
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Daphne Laubreton
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Sophia Djebali
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Manon Zala
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Christophe Arpin
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Laboratoire de Biologie et de Modélisation de la Cellule, Université de Lyon, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Lyon, France
| | - Laurent Genestier
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Yann Leverrier
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Olivier Gandrillon
- Inria, Villeurbanne, France
- Laboratoire de Biologie et de Modélisation de la Cellule, Université de Lyon, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Lyon, France
| | - Fabien Crauste
- Laboratoire MAP5 (UMR CNRS 8145), Université Paris Cité, Paris, France
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jacqueline Marvel
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| |
Collapse
|
25
|
García-Vega M, Wan H, Reséndiz-Sandoval M, Hinojosa-Trujillo D, Valenzuela O, Mata-Haro V, Dehesa-Canseco F, Solís-Hernández M, Marcotte H, Pan-Hammarström Q, Hernández J. Comparative single-cell transcriptomic profile of hybrid immunity induced by adenovirus vector-based COVID-19 vaccines. Genes Immun 2024; 25:158-167. [PMID: 38570727 DOI: 10.1038/s41435-024-00270-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
In this study, antibody response and a single-cell RNA-seq analysis were conducted on peripheral blood mononuclear cells from five different groups: naïve subjects vaccinated with AZD1222 (AZ) or Ad5-nCoV (Cso), individuals previously infected and later vaccinated (hybrid) with AZD1222 (AZ-hb) or Ad5-nCoV (Cso-hb), and those who were infected and had recovered from COVID-19 (Inf). The results showed that AZ induced more robust neutralizing antibody responses than Cso. The single-cell RNA data revealed a high frequency of memory B cells in the Cso and Cso-hb. In contrast, AZ and AZ-hb groups exhibited the highest proportion of activated naïve B cells expressing CXCR4. Transcriptomic analysis of CD4+ and CD8+ T cells demonstrated a heterogeneous response following vaccination, hybrid immunity, or natural infection. However, a single dose of Ad5-nCoV was sufficient to strongly activate CD4+ T cells (naïve and memory) expressing ANX1 and FOS, similar to the hybrid response observed with AZ. An interesting finding was the robust activation of a subset of CD8+ T cells expressing GZMB, GZMH, and IFNG genes in the Cso-hb group. Our findings suggest that both vaccines effectively stimulated the cellular immune response; however, the Ad5-nCoV induced a more robust CD8+ T-cell response in previously infected individuals.
Collapse
Affiliation(s)
- Melissa García-Vega
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, Sonora, 83304, Mexico
| | - Hui Wan
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, SE171 65, Sweden
| | - Mónica Reséndiz-Sandoval
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, Sonora, 83304, Mexico
| | - Diana Hinojosa-Trujillo
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, Sonora, 83304, Mexico
| | - Olivia Valenzuela
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora, Hermosillo, Sonora, 83000, Mexico
| | - Verónica Mata-Haro
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, Sonora, 83304, Mexico
| | - Freddy Dehesa-Canseco
- Comisión México-Estados Unidos para la Prevención de la Fiebre Aftosa y otras Enfermedades Exóticas de los Animales (CPA), SENASICA, SADER, Ciudad de México, 05010, Mexico
| | - Mario Solís-Hernández
- Comisión México-Estados Unidos para la Prevención de la Fiebre Aftosa y otras Enfermedades Exóticas de los Animales (CPA), SENASICA, SADER, Ciudad de México, 05010, Mexico
| | - Harold Marcotte
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, SE171 65, Sweden
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, SE171 65, Sweden.
| | - Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, Sonora, 83304, Mexico.
| |
Collapse
|
26
|
Sakellariou C, Roser LA, Schiffmann S, Lindstedt M. Fine tuning of the innate and adaptive immune responses by Interleukin-2. J Immunotoxicol 2024; 21:2332175. [PMID: 38526995 DOI: 10.1080/1547691x.2024.2332175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 03/13/2024] [Indexed: 03/27/2024] Open
Abstract
Novel immunotherapies for cancer and other diseases aim to trigger the immune system to produce durable responses, while overcoming the immunosuppression that may contribute to disease severity, and in parallel considering immunosafety aspects. Interleukin-2 (IL-2) was one of the first cytokines that the FDA approved as a cancer-targeting immunotherapy. However, in the past years, IL-2 immunotherapy is not actively offered to patients, due to limited efficacy, when compared to other novel immunotherapies, and the associated severe adverse events. In order to design improved in vitro and in vivo models, able to predict the efficacy and safety of novel IL-2 alternatives, it is important to delineate the mechanistic immunological events triggered by IL-2. Particularly, in this review we will discuss the effects IL-2 has with the bridging cell type of the innate and adaptive immune responses, dendritic cells. The pathways involved in the regulation of IL-2 by dendritic cells and T-cells in cancer and autoimmune disease will also be explored.
Collapse
Affiliation(s)
| | - Luise A Roser
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Susanne Schiffmann
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| |
Collapse
|
27
|
Moreira LR, Silva AC, da Costa-Oliveira CN, da Silva-Júnior CD, Oliveira KKDS, Torres DJL, Barros MD, Rabello MCDS, de Lorena VMB. Interaction between peripheral blood mononuclear cells and Trypanosoma cruzi-infected adipocytes: implications for treatment failure and induction of immunomodulatory mechanisms in adipose tissue. Front Immunol 2024; 15:1280877. [PMID: 38533504 PMCID: PMC10963431 DOI: 10.3389/fimmu.2024.1280877] [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: 08/21/2023] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
Background/Introduction Adipose tissue (AT) has been highlighted as a promising reservoir of infection for viruses, bacteria and parasites. Among them is Trypanosoma cruzi, which causes Chagas disease. The recommended treatment for the disease in Brazil is Benznidazole (BZ). However, its efficacy may vary according to the stage of the disease, geographical origin, age, immune background of the host and sensitivity of the strains to the drug. In this context, AT may act as an ally for the parasite survival and persistence in the host and a barrier for BZ action. Therefore, we investigated the immunomodulation of T. cruzi-infected human AT in the presence of peripheral blood mononuclear cells (PBMC) where BZ treatment was added. Methods We performed indirect cultivation between T. cruzi-infected adipocytes, PBMC and the addition of BZ. After 72h of treatment, the supernatant was collected for cytokine, chemokine and adipokine assay. Infected adipocytes were removed to quantify T. cruzi DNA, and PBMC were removed for immunophenotyping. Results Our findings showed elevated secretion of interleukin (IL)-6, IL-2 and monocyte chemoattractant protein-1 (MCP-1/CCL2) in the AT+PBMC condition compared to the other controls. In contrast, there was a decrease in tumor necrosis factor (TNF) and IL-8/CXCL-8 in the groups with AT. We also found high adipsin secretion in PBMC+AT+T compared to the treated condition (PBMC+AT+T+BZ). Likewise, the expression of CD80+ and HLA-DR+ in CD14+ cells decreased in the presence of T. cruzi. Discussion Thus, our findings indicate that AT promotes up-regulation of inflammatory products such as IL-6, IL-2, and MCP-1/CCL2. However, adipogenic inducers may have triggered the downregulation of TNF and IL-8/CXCL8 through the peroxisome proliferator agonist gamma (PPAR-g) or receptor expression. On the other hand, the administration of BZ only managed to reduce inflammation in the microenvironment by decreasing adipsin in the infected culture conditions. Therefore, given the findings, we can see that AT is an ally of the parasite in evading the host's immune response and the pharmacological action of BZ.
Collapse
Affiliation(s)
- Leyllane Rafael Moreira
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | - Ana Carla Silva
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | | | - Claudeir Dias da Silva-Júnior
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | | | - Diego José Lira Torres
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | | | | | | |
Collapse
|
28
|
Na K, Lee S, Kim DK, Kim YS, Hwang JY, Kang SS, Baek S, Lee CY, Yang SM, Han YJ, Kim MH, Han H, Kim Y, Kim JH, Jeon S, Byeon Y, Lee JB, Lim SM, Hong MH, Pyo KH, Cho BC. CD81 and CD82 expressing tumor-infiltrating lymphocytes in the NSCLC tumor microenvironment play a crucial role in T-cell activation and cytokine production. Front Immunol 2024; 15:1336246. [PMID: 38515751 PMCID: PMC10954780 DOI: 10.3389/fimmu.2024.1336246] [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: 11/10/2023] [Accepted: 02/02/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction To understand the immune system within the tumor microenvironment (TME) of non-small cell lung cancer (NSCLC), it is crucial to elucidate the characteristics of molecules associated with T cell activation. Methods We conducted an in-depth analysis using single-cell RNA sequencing data obtained from tissue samples of 19 NSCLC patients. T cells were classified based on the Tumor Proportion Score (TPS) within the tumor region, and molecular markers associated with activation and exhaustion were analyzed in T cells from high TPS areas. Results Notably, tetraspanins CD81 and CD82, belonging to the tetraspanin protein family, were found to be expressed in activated T cells, particularly in cytotoxic T cells. These tetraspanins showed strong correlations with activation and exhaustion markers. In vitro experiments confirmed increased expression of CD81 and CD82 in IL-2-stimulated T cells. T cells were categorized into CD81highCD82high and CD81lowCD82low groups based on their expression levels, with CD81highCD82high T cells exhibiting elevated activation markers such as CD25 and CD69 compared to CD81lowCD82low T cells. This trend was consistent across CD3+, CD8+, and CD4+ T cell subsets. Moreover, CD81highCD82high T cells, when stimulated with anti-CD3, demonstrated enhanced secretion of cytokines such as IFN-γ, TNF-α, and IL-2, along with an increase in the proportion of memory T cells. Bulk RNA sequencing results after sorting CD81highCD82high and CD81lowCD82low T cells consistently supported the roles of CD81 and CD82. Experiments with overexpressed CD81 and CD82 showed increased cytotoxicity against target cells. Discussion These findings highlight the multifaceted roles of CD81 and CD82 in T cell activation, cytokine production, memory subset accumulation, and target cell cytolysis. Therefore, these findings suggest the potential of CD81 and CD82 as promising candidates for co-stimulatory molecules in immune therapeutic strategies for cancer treatment within the intricate TME.
Collapse
Affiliation(s)
- Kwangmin Na
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seul Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Dong Kwon Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Young Seob Kim
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Yeon Hwang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seong-San Kang
- JEUK Institute for Cancer Research, JEUK Co., Ltd., Gumi-City, Republic of Korea
| | - Sujeong Baek
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chai Young Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Min Yang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Jin Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi Hyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heekyung Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngtaek Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seunghyun Jeon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngseon Byeon
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Min Lim
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Hee Hong
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Ho Pyo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byoung Chul Cho
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
Zhang Q, Jiang L, Wang W, Huber AK, Valvo VM, Jungles KM, Holcomb EA, Pearson AN, The S, Wang Z, Parsels LA, Parsels JD, Wahl DR, Rao A, Sahai V, Lawrence TS, Green MD, Morgan MA. Potentiating the radiation-induced type I interferon antitumoral immune response by ATM inhibition in pancreatic cancer. JCI Insight 2024; 9:e168824. [PMID: 38376927 PMCID: PMC11063931 DOI: 10.1172/jci.insight.168824] [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: 01/12/2023] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
Radiotherapy induces a type I interferon-mediated (T1IFN-mediated) antitumoral immune response that we hypothesized could be potentiated by a first-in-class ataxia telangiectasia mutated (ATM) inhibitor, leading to enhanced innate immune signaling, T1IFN expression, and sensitization to immunotherapy in pancreatic cancer. We evaluated the effects of AZD1390 or a structurally related compound, AZD0156, on innate immune signaling and found that both inhibitors enhanced radiation-induced T1IFN expression via the POLIII/RIG-I/MAVS pathway. In immunocompetent syngeneic mouse models of pancreatic cancer, ATM inhibitor enhanced radiation-induced antitumoral immune responses and sensitized tumors to anti-PD-L1, producing immunogenic memory and durable tumor control. Therapeutic responses were associated with increased intratumoral CD8+ T cell frequency and effector function. Tumor control was dependent on CD8+ T cells, as therapeutic efficacy was blunted in CD8+ T cell-depleted mice. Adaptive immune responses to combination therapy provided systemic control of contralateral tumors outside of the radiation field. Taken together, we show that a clinical candidate ATM inhibitor enhances radiation-induced T1IFN, leading to both innate and subsequent adaptive antitumoral immune responses and sensitization of otherwise resistant pancreatic cancer to immunotherapy.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | - Kassidy M. Jungles
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Stephanie The
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | | | | | | | - Daniel R. Wahl
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Arvind Rao
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Vaibhav Sahai
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Division of Hematology and Oncology, Department of Internal Medicine, and
| | - Theodore S. Lawrence
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael D. Green
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Meredith A. Morgan
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
30
|
Sprent J, Boyman O. Optimising IL-2 for Cancer Immunotherapy. Immune Netw 2024; 24:e5. [PMID: 38455463 PMCID: PMC10917570 DOI: 10.4110/in.2024.24.e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
The key role of T cells in cancer immunotherapy is well established and is highlighted by the remarkable capacity of Ab-mediated checkpoint blockade to overcome T-cell exhaustion and amplify anti-tumor responses. However, total or partial tumor remission following checkpoint blockade is still limited to only a few types of tumors. Hence, concerted attempts are being made to devise new methods for improving tumor immunity. Currently, much attention is being focused on therapy with IL-2. This cytokine is a powerful growth factor for T cells and optimises their effector functions. When used at therapeutic doses for cancer treatment, however, IL-2 is highly toxic. Nevertheless, recent work has shown that modifying the structure or presentation of IL-2 can reduce toxicity and lead to effective anti-tumor responses in synergy with checkpoint blockade. Here, we review the complex interaction of IL-2 with T cells: first during normal homeostasis, then during responses to pathogens, and finally in anti-tumor responses.
Collapse
Affiliation(s)
- Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst 2010, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney 1466, Australia
- Menzies Institute of Medical Research, Hobart 7000, Australia
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich 8091, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, Zurich 8057, Switzerland
| |
Collapse
|
31
|
Petkau G, Mitchell TJ, Evans MJ, Matheson L, Salerno F, Turner M. Zfp36l1 establishes the high-affinity CD8 T-cell response by directly linking TCR affinity to cytokine sensing. Eur J Immunol 2024; 54:e2350700. [PMID: 38039407 PMCID: PMC11146077 DOI: 10.1002/eji.202350700] [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: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/03/2023]
Abstract
How individual T cells compete for and respond to IL-2 at the molecular level, and, as a consequence, how this shapes population dynamics and the selection of high-affinity clones is still poorly understood. Here we describe how the RNA binding protein ZFP36L1, acts as a sensor of TCR affinity to promote clonal expansion of high-affinity CD8 T cells. As part of an incoherent feed-forward loop, ZFP36L1 has a nonredundant role in suppressing multiple negative regulators of cytokine signaling and mediating a selection mechanism based on competition for IL-2. We suggest that ZFP36L1 acts as a sensor of antigen affinity and establishes the dominance of high-affinity T cells by installing a hierarchical response to IL-2.
Collapse
Affiliation(s)
- Georg Petkau
- The Babraham InstituteBabraham Research CampusCambridgeUnited Kingdom
| | - Twm J. Mitchell
- The Babraham InstituteBabraham Research CampusCambridgeUnited Kingdom
| | | | - Louise Matheson
- The Babraham InstituteBabraham Research CampusCambridgeUnited Kingdom
| | - Fiamma Salerno
- The Babraham InstituteBabraham Research CampusCambridgeUnited Kingdom
| | - Martin Turner
- The Babraham InstituteBabraham Research CampusCambridgeUnited Kingdom
| |
Collapse
|
32
|
Jacques C, Marchand F, Chatelais M, Brulefert A, Floris I. Understanding the Mode of Action of a Micro-Immunotherapy Formulation: Pre-Clinical Evidence from the Study of 2LEBV ® Active Ingredients. Life (Basel) 2024; 14:102. [PMID: 38255717 PMCID: PMC10821216 DOI: 10.3390/life14010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is often kept silent and asymptomatic; however, its reactivation induces a chronic and/or recurrent infection that is associated with numerous diseases, including cancer and inflammation-related disorders. As no specific treatment is currently available, the immune factors-based micro-immunotherapy (MI) medicine 2LEBV® could be considered a valuable therapeutic option to sustain the immune system in EBV reactivation. METHODS The present work aimed to investigate, for the first time, the effect of 2LEBV® in several in vitro models of uninfected immune-related cells. RESULTS 2LEBV® displayed phagocytosis-enhancing capabilities in granulocytes. In human peripheral blood mononuclear cells (PBMCs), it increased the intra- and extra-cellular expression of interleukin (IL)-2. Moreover, it modulated the secretion of other cytokines, increasing IL-4, IL-6, and tumor necrosis factor-α levels or lowering other cytokines levels such as IL-9. Finally, 2LEBV® reduced the expression of human leukocyte antigen (HLA)-II in endothelial cells and macrophages. CONCLUSIONS Although these data are still preliminary and the chosen models do not consider the underlying EBV-reactivation mechanisms, they still provide a better understanding of the mechanisms of action of 2LEBV®, both at functional and molecular levels. Furthermore, they open perspectives regarding the potential targets of 2LEBV® in its employment as a therapeutic intervention for EBV-associated diseases.
Collapse
Affiliation(s)
- Camille Jacques
- Pre-Clinical Research Department, Labo’Life France, Pescalis-Les Magnys, 79320 Moncoutant-sur-Sevre, France;
| | - Flora Marchand
- ProfileHIT, 7 rue du Buisson, 44680 Sainte-Pazanne, France; (F.M.); (M.C.)
| | - Mathias Chatelais
- ProfileHIT, 7 rue du Buisson, 44680 Sainte-Pazanne, France; (F.M.); (M.C.)
| | - Adrien Brulefert
- QIMA Life Sciences, 1 bis rue des Plantes—CS 50011, 86160 Gençay, France;
| | - Ilaria Floris
- Pre-Clinical Research Department, Labo’Life France, Pescalis-Les Magnys, 79320 Moncoutant-sur-Sevre, France;
| |
Collapse
|
33
|
Daian E Silva DSO, Cox LJ, Rocha AS, Lopes-Ribeiro Á, Souza JPC, Franco GM, Prado JLC, Pereira-Santos TA, Martins ML, Coelho-Dos-Reis JGA, Gomes-de-Pinho TM, Da Fonseca FG, Barbosa-Stancioli EF. Preclinical assessment of an anti-HTLV-1 heterologous DNA/MVA vaccine protocol expressing a multiepitope HBZ protein. Virol J 2023; 20:304. [PMID: 38115107 PMCID: PMC10731796 DOI: 10.1186/s12985-023-02264-z] [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: 06/16/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Human T-lymphotropic virus 1 (HTLV-1) is associated with the development of several pathologies and chronic infection in humans. The inefficiency of the available treatments and the challenge in developing a protective vaccine highlight the need to produce effective immunotherapeutic tools. The HTLV-1 basic leucine zipper (bZIP) factor (HBZ) plays an important role in the HTLV-1 persistence, conferring a survival advantage to infected cells by reducing the HTLV-1 proteins expression, allowing infected cells to evade immune surveillance, and enhancing cell proliferation leading to increased proviral load. METHODS We have generated a recombinant Modified Virus Vaccinia Ankara (MVA-HBZ) and a plasmid DNA (pcDNA3.1(+)-HBZ) expressing a multiepitope protein based on peptides of HBZ to study the immunogenic potential of this viral-derived protein in BALB/c mice model. Mice were immunized in a prime-boost heterologous protocol and their splenocytes (T CD4+ and T CD8+) were immunophenotyped by flow cytometry and the humoral response was evaluated by ELISA using HBZ protein produced in prokaryotic vector as antigen. RESULTS T CD4+ and T CD8+ lymphocytes cells stimulated by HBZ-peptides (HBZ42-50 and HBZ157-176) showed polyfunctional double positive responses for TNF-α/IFN-γ, and TNF-α/IL-2. Moreover, T CD8+ cells presented a tendency in the activation of effector memory cells producing granzyme B (CD44+High/CD62L-Low), and the activation of Cytotoxic T Lymphocytes (CTLs) and cytotoxic responses in immunized mice were inferred through the production of granzyme B by effector memory T cells and the expression of CD107a by CD8+ T cells. The overall data is consistent with a directive and effector recall response, which may be able to operate actively in the elimination of HTLV-1-infected cells and, consequently, in the reduction of the proviral load. Sera from immunized mice, differently from those of control animals, showed IgG-anti-HBZ production by ELISA. CONCLUSIONS Our results highlight the potential of the HBZ multiepitope protein expressed from plasmid DNA and a poxviral vector as candidates for therapeutic vaccine.
Collapse
Affiliation(s)
- D S O Daian E Silva
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - L J Cox
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - A S Rocha
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - Á Lopes-Ribeiro
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - J P C Souza
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - G M Franco
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - J L C Prado
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - T A Pereira-Santos
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - M L Martins
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
- Gerência de Desenvolvimento Técnico Científico, Fundação Centro de Hematologia e Hemoterapia do Estado de Minas Gerais - Hemominas, Belo Horizonte, Brazil
| | - J G A Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - T M Gomes-de-Pinho
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - F G Da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - E F Barbosa-Stancioli
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil.
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil.
| |
Collapse
|
34
|
Jamison BL, Lawrance M, Wang CJ, DeBerg HA, Sansom DM, Gavin MA, Walker LS, Campbell DJ. An IL-2 mutein increases IL-10 and CTLA-4-dependent suppression of dendritic cells by regulatory T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569613. [PMID: 38106196 PMCID: PMC10723345 DOI: 10.1101/2023.12.01.569613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Interleukin-2 (IL-2) variants with increased CD25 dependence that selectively expand Foxp3+ regulatory T (TR) cells are in clinical trials for treating inflammatory diseases. Using an Fc-fused IL-2 mutein (Fc.IL-2 mutein) we developed that prevents diabetes in non-obese diabetic (NOD) mice, we show that Fc.IL-2 mutein induced an activated TR population with elevated proliferation, a transcriptional program associated with Stat5- and TCR-dependent gene modules, and high IL-10 and CTLA-4 expression. Increased IL-10 signaling limited surface MHC class II upregulation during conventional dendritic cell (cDC) maturation, while increased CTLA-4-dependent transendocytosis led to the transfer of CD80 and CD86 costimulatory ligands from maturing cDCs to TR cells. In NOD mice, Fc.IL-2 mutein treatment promoted the suppression of cDCs in the inflamed pancreas and pancreatic lymph nodes resulting in T cell anergy. Thus, IL-2 mutein-expanded TR cells have enhanced functional properties and restrict cDC function, offering promise for targeted immunotherapy use in autoimmune disease.
Collapse
Affiliation(s)
- Braxton L. Jamison
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA
| | | | - Chun Jing Wang
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | | | - David M. Sansom
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | | | - Lucy S.K. Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Daniel J. Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA
- Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
35
|
Lahimchi MR, Maroufi F, Maali A. Induced Pluripotent Stem Cell-Derived Chimeric Antigen Receptor T Cells: The Intersection of Stem Cells and Immunotherapy. Cell Reprogram 2023; 25:195-211. [PMID: 37782910 DOI: 10.1089/cell.2023.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising cell-based immunotherapy applicable to various cancers. High cost of production, immune rejection, heterogeneity of cell product, limited cell source, limited expandability, and relatively long production time have created the need to achieve a universal allogeneic CAR-T cell product for "off-the-shelf" application. Since the innovation of induced pluripotent stem cells (iPSCs) by Yamanaka et al., extensive efforts have been made to prepare an unlimited cell source for regenerative medicine, that is, immunotherapy. In the autologous grafting approach, iPSCs prepare the desired cell source for generating autologous CAR-T cells through more accessible and available sources. In addition, generating iPSC-derived CAR-T cells is a promising approach to achieving a suitable source for producing an allogeneic CAR-T cell product. In brief, the first step is reprogramming somatic cells (accessible from peripheral blood, skin, etc.) to iPSCs. In the next step, CAR expression and T cell lineage differentiation should be applied in different arrangements. In addition, in an allogeneic manner, human leukocyte antigen/T cell receptor (TCR) deficiency should be applied in iPSC colonies. The allogeneic iPSC-derived CAR-T cell experiments showed that simultaneous performance of HLA/TCR deficiency, CAR expression, and T cell lineage differentiation could bring the production to the highest efficacy in generating allogeneic iPSC-derived CAR-T cells.
Collapse
Affiliation(s)
| | - Faezeh Maroufi
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
36
|
Trujillo-Cirilo L, Weiss-Steider B, Vargas-Angeles CA, Corona-Ortega MT, Rangel-Corona R. Immune microenvironment of cervical cancer and the role of IL-2 in tumor promotion. Cytokine 2023; 170:156334. [PMID: 37598478 DOI: 10.1016/j.cyto.2023.156334] [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: 08/11/2022] [Revised: 07/06/2023] [Accepted: 08/11/2023] [Indexed: 08/22/2023]
Abstract
The tumor microenvironment (TME) is a heterogeneous mixture of resident and tumor cells that maintain close communication through their secretion products. The composition of the TME is dynamic and complex among the different types of cancer, where the immune cells play a relevant role in the elimination of tumor cells, however, under certain circumstances they contribute to tumor development. In cervical cancer (CC) the human papilloma virus (HPV) shapes the microenvironment in order to mediate persistent infections that favors transformation and tumor development. Interleukin-2 (IL-2) is an important TME cytokine that induces CD8+ effector T cells and NKs to eliminate tumor cells, however, IL-2 can also suppress the immune response through Treg cells. Recent studies have shown that CC cells express the IL-2 receptor (IL-2R), that are induced to proliferate at low concentrations of exogenous IL-2 through alterations in the JAK/STAT pathway. This review provides an overview of the main immune cells that make up the TME in CC, as well as the participation of IL-2 in the tumor promotion. Finally, it is proposed that the low density of IL-2 produced by immunocompetent cells is used by tumor cells through its IL-2R as a mechanism to proliferate simultaneously depleting this molecule in order to evade immune response.
Collapse
Affiliation(s)
- Leonardo Trujillo-Cirilo
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico.
| | - Benny Weiss-Steider
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico
| | - Carlos Adrian Vargas-Angeles
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico
| | - Maria Teresa Corona-Ortega
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico
| | - Rosalva Rangel-Corona
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico
| |
Collapse
|
37
|
Thio CLP, Chang YJ. The modulation of pulmonary group 2 innate lymphoid cell function in asthma: from inflammatory mediators to environmental and metabolic factors. Exp Mol Med 2023; 55:1872-1884. [PMID: 37696890 PMCID: PMC10545775 DOI: 10.1038/s12276-023-01021-0] [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: 12/30/2022] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 09/13/2023] Open
Abstract
A dysregulated type 2 immune response is one of the fundamental causes of allergic asthma. Although Th2 cells are undoubtedly central to the pathogenesis of allergic asthma, the discovery of group 2 innate lymphoid cells (ILC2s) has added another layer of complexity to the etiology of this chronic disease. Through their inherent innate type 2 responses, ILC2s not only contribute to the initiation of airway inflammation but also orchestrate the recruitment and activation of other members of innate and adaptive immunity, further amplifying the inflammatory response. Moreover, ILC2s exhibit substantial cytokine plasticity, as evidenced by their ability to produce type 1- or type 17-associated cytokines under appropriate conditions, underscoring their potential contribution to nonallergic, neutrophilic asthma. Thus, understanding the mechanisms of ILC2 functions is pertinent. In this review, we present an overview of the current knowledge on ILC2s in asthma and the regulatory factors that modulate lung ILC2 functions in various experimental mouse models of asthma and in humans.
Collapse
Affiliation(s)
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, 115, Taiwan.
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung City, 404, Taiwan.
| |
Collapse
|
38
|
Upadhaya P, Lamenza FF, Shrestha S, Roth P, Jagadeesha S, Pracha H, Horn NA, Oghumu S. Berry Extracts and Their Bioactive Compounds Mitigate LPS and DNFB-Mediated Dendritic Cell Activation and Induction of Antigen Specific T-Cell Effector Responses. Antioxidants (Basel) 2023; 12:1667. [PMID: 37759970 PMCID: PMC10525528 DOI: 10.3390/antiox12091667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Berries have gained widespread recognition for their abundant natural antioxidant, anti-inflammatory, and immunomodulatory properties. However, there has been limited research conducted thus far to investigate the role of the active constituents of berries in alleviating contact hypersensitivity (CHS), the most prevalent occupational dermatological disease. Our study involved an ex vivo investigation aimed at evaluating the impact of black raspberry extract (BRB-E) and various natural compounds found in berries, such as protocatechuic acid (PCA), proanthocyanidins (PANT), ellagic acid (EA), and kaempferol (KMP), on mitigating the pathogenicity of CHS. We examined the efficacy of these natural compounds on the activation of dendritic cells (DCs) triggered by 2,4-dinitrofluorobenzene (DNFB) and lipopolysaccharide (LPS). Specifically, we measured the expression of activation markers CD40, CD80, CD83, and CD86 and the production of proinflammatory cytokines, including Interleukin (IL)-12, IL-6, TNF-α, and IL-10, to gain further insights. Potential mechanisms through which these phytochemicals could alleviate CHS were also investigated by investigating the role of phospho-ERK. Subsequently, DCs were co-cultured with T-cells specific to the OVA323-339 peptide to examine the specific T-cell effector responses resulting from these interactions. Our findings demonstrated that BRB-E, PCA, PANT, and EA, but not KMP, inhibited phosphorylation of ERK in LPS-activated DCs. At higher doses, EA significantly reduced expression of all the activation markers studied in DNFB- and LPS-stimulated DCs. All compounds tested reduced the level of IL-6 in DNFB-stimulated DCs in Flt3L as well as in GM-CSF-derived DCs. However, levels of IL-12 were reduced by all the tested compounds in LPS-stimulated Flt3L-derived BMDCs. PCA, PANT, EA, and KMP inhibited the activated DC-mediated Interferon (IFN)-γ and IL-17 production by T-cells. Interestingly, PANT, EA, and KMP significantly reduced T-cell proliferation and the associated IL-2 production. Our study provides evidence for differential effects of berry extracts and natural compounds on DNFB and LPS-activated DCs revealing potential novel approaches for mitigating CHS.
Collapse
Affiliation(s)
- Puja Upadhaya
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
| | - Felipe F. Lamenza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Suvekshya Shrestha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Peyton Roth
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
| | - Sushmitha Jagadeesha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
| | - Hasan Pracha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
| | - Natalie A. Horn
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.U.); (F.F.L.); (S.S.); (P.R.); (S.J.); (H.P.); (N.A.H.)
| |
Collapse
|
39
|
Ildefonso GV, Finley SD. A data-driven Boolean model explains memory subsets and evolution in CD8+ T cell exhaustion. NPJ Syst Biol Appl 2023; 9:36. [PMID: 37524735 PMCID: PMC10390540 DOI: 10.1038/s41540-023-00297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
T cells play a key role in a variety of immune responses, including infection and cancer. Upon stimulation, naïve CD8+ T cells proliferate and differentiate into a variety of memory and effector cell types; however, failure to clear antigens causes prolonged stimulation of CD8+ T cells, ultimately leading to T cell exhaustion (TCE). The functional and phenotypic changes that occur during CD8+ T cell differentiation are well characterized, but the underlying gene expression state changes are not completely understood. Here, we utilize a previously published data-driven Boolean model of gene regulatory interactions shown to mediate TCE. Our network analysis and modeling reveal the final gene expression states that correspond to TCE, along with the sequence of gene expression patterns that give rise to those final states. With a model that predicts the changes in gene expression that lead to TCE, we could evaluate strategies to inhibit the exhausted state. Overall, we demonstrate that a common pathway model of CD8+ T cell gene regulatory interactions can provide insights into the transcriptional changes underlying the evolution of cell states in TCE.
Collapse
Affiliation(s)
- Geena V Ildefonso
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA.
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
40
|
Li S, Hao L, Zhang J, Deng J, Hu X. Focus on T cell exhaustion: new advances in traditional Chinese medicine in infection and cancer. Chin Med 2023; 18:76. [PMID: 37355637 DOI: 10.1186/s13020-023-00785-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
In chronic infections and cancers, T lymphocytes (T cells) are exposed to persistent antigen or inflammatory signals. The condition is often associated with a decline in T-cell function: a state called "exhaustion". T cell exhaustion is a state of T cell dysfunction characterized by increased expression of a series of inhibitory receptors (IRs), decreased effector function, and decreased cytokine secretion, accompanied by transcriptional and epigenetic changes and metabolic defects. The rise of immunotherapy, particularly the use of immune checkpoint inhibitors (ICIs), has dramatically changed the clinical treatment paradigm for patients. However, its low response rate, single target and high immunotoxicity limit its clinical application. The multiple immunomodulatory potential of traditional Chinese medicine (TCM) provides a new direction for improving the treatment of T cell exhaustion. Here, we review recent advances that have provided a clearer molecular understanding of T cell exhaustion, revealing the characteristics and causes of T cell exhaustion in persistent infections and cancers. In addition, this paper summarizes recent advances in improving T cell exhaustion in infectious diseases and cancer with the aim of providing a comprehensive and valuable source of information on TCM as an experimental study and their role in collaboration with ICIs therapy.
Collapse
Affiliation(s)
- Shenghao Li
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Liyuan Hao
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Junli Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Jiali Deng
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
41
|
Tang Y, Fakhari S, Huntemann ED, Feng Z, Wu P, Feng WY, Lei J, Yuan F, Excoffon KJ, Wang K, Limberis MP, Kolbeck R, Yan Z, Engelhardt JF. Immunosuppression reduces rAAV2.5T neutralizing antibodies that limit efficacy following repeat dosing to ferret lungs. Mol Ther Methods Clin Dev 2023; 29:70-80. [PMID: 36950451 PMCID: PMC10025970 DOI: 10.1016/j.omtm.2023.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
The efficacy of redosing the recombinant adeno-associated virus (rAAV) vector rAAV2.5T to ferret lung is limited by AAV neutralizing antibody (NAb) responses. While immunosuppression strategies have allowed for systemic rAAV repeat dosing, their utility for rAAV lung-directed gene therapy is largely unexplored. To this end, we evaluated two immunosuppression (IS) strategies to improve repeat dosing of rAAV2.5T to ferret lungs: (1) a combination of three IS drugs (Tri-IS) with broad coverage against cellular and humoral responses (methylprednisolone [MP], azathioprine, and cyclosporine) and (2) MP alone, which is typically used in systemic rAAV applications. Repeat dosing utilized AAV2.5T-SP183-fCFTRΔR (recombinant ferret CFTR transgene), followed 28 days later by AAV2.5T-SP183-gLuc (for quantification of transgene expression). Both the Tri-IS and MP strategies significantly improved transgene expression following repeat dosing and reduced AAV2.5T NAb responses in the bronchioalveolar lavage fluid (BALF) and plasma, while AAV2.5T binding antibody subtypes and cellular immune responses by ELISpot were largely unchanged by IS. One exception was the reduction in plasma AAV2.5T binding immunoglobulin G (IgG) in both IS groups. Only the Tri-IS strategy significantly suppressed splenocyte expression of IFNA (interferon α [IFN-α]) and IL4. Our studies suggest that IS strategies may be useful in clinical application of rAAV targeting lung genetic diseases such as cystic fibrosis.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shahab Fakhari
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Eric D. Huntemann
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zehua Feng
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Peipei Wu
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - William Y. Feng
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Junying Lei
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Feng Yuan
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Kai Wang
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | | | | | - Ziying Yan
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - John F. Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
42
|
Anderson J, Jalali S, Licciardi PV, Pellicci DG. OMIP-91: A 27-color flow cytometry panel to evaluate the phenotype and function of human conventional and unconventional T-cells. Cytometry A 2023. [PMID: 37183268 DOI: 10.1002/cyto.a.24738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/13/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023]
Abstract
This 27-color panel was developed to simultaneously measure different T-cell populations (CD4, CD8, γδ T-cells, and MAIT cells) and their subsets (Memory, Th1, Th2, Th17, Tfh, and Treg) along with functional markers associated with their activation status, cytokine production and cytotoxicity. This panel will be useful for both in vivo and in vitro studies evaluating T-cells in the context of human health and disease. This panel is valuable in settings where samples are limited as a large amount of data will be generated using small volumes of blood.
Collapse
Affiliation(s)
- Jeremy Anderson
- New Vaccines, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Sedi Jalali
- New Vaccines, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Cellular Immunology, Murdoch Children's Research Institute, Melbourne, Australia
| | - Paul V Licciardi
- New Vaccines, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Daniel G Pellicci
- New Vaccines, Murdoch Children's Research Institute, Melbourne, Australia
- Cellular Immunology, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
43
|
Araujo IL, Piraine REA, Fischer G, Leite FPL. Recombinant BoHV-5 glycoprotein (rgD5) elicits long-lasting protective immunity in cattle. Virology 2023; 584:44-52. [PMID: 37244054 DOI: 10.1016/j.virol.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 03/27/2023] [Accepted: 04/13/2023] [Indexed: 05/29/2023]
Abstract
BoHV-5 is a worldwide distributed pathogen usually associated with a lethal neurological disease in dairy and beef cattle resulting in important economic losses due to the cattle industry. Using recombinant gD5, we evaluated the long-duration humoral immunity of the recombinant vaccines in a cattle model. Here we report that two doses of intramuscular immunization, particularly with the rgD5ISA vaccine, induce long-lasting antibody responses. Recombinant gD5 antigen elicited tightly mRNA transcription of the Bcl6 and the chemokine receptor CXCR5 which mediate memory B cells and long-lived plasma cells in germinal centers. In addition, using an in-house indirect ELISA we observed higher and earlier responses of rgD5-specific IgG antibody and the upregulation of mRNA transcription of IL2, IL4, IL10, IL15, and IFN-γ in rgD5 vaccinated cattle, indicating a mixed immune response. We further show that rgD5 immunization protects against both BoHV -1 and -5. Our findings indicate that the rgD5-based vaccine represents an effective vaccine strategy to induce an efficient control of herpesviruses.
Collapse
Affiliation(s)
- Itauá L Araujo
- Biotechnology Unit, Technological Development Centre, Federal University of Pelotas, Pelotas, Brazil.
| | - Renan E A Piraine
- Biotechnology Unit, Technological Development Centre, Federal University of Pelotas, Pelotas, Brazil.
| | - Geferson Fischer
- Laboratory of Virology and Immunology, Federal University of Federal de Pelotas, Pelotas, RS, Brazil.
| | - Fábio P L Leite
- Biotechnology Unit, Technological Development Centre, Federal University of Pelotas, Pelotas, Brazil; Laboratory of Virology and Immunology, Federal University of Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
44
|
Piper M, Hoen M, Darragh LB, Knitz MW, Nguyen D, Gadwa J, Durini G, Karakoc I, Grier A, Neupert B, Van Court B, Abdelazeem KNM, Yu J, Olimpo NA, Corbo S, Ross RB, Pham TT, Joshi M, Kedl RM, Saviola AJ, Amann M, Umaña P, Codarri Deak L, Klein C, D'Alessandro A, Karam SD. Simultaneous targeting of PD-1 and IL-2Rβγ with radiation therapy inhibits pancreatic cancer growth and metastasis. Cancer Cell 2023; 41:950-969.e6. [PMID: 37116489 PMCID: PMC10246400 DOI: 10.1016/j.ccell.2023.04.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/05/2023] [Accepted: 03/31/2023] [Indexed: 04/30/2023]
Abstract
In pancreatic ductal adenocarcinoma (PDAC) patients, we show that response to radiation therapy (RT) is characterized by increased IL-2Rβ and IL-2Rγ along with decreased IL-2Rα expression. The bispecific PD1-IL2v is a PD-1-targeted IL-2 variant (IL-2v) immunocytokine with engineered IL-2 cis targeted to PD-1 and abolished IL-2Rα binding, which enhances tumor-antigen-specific T cell activation while reducing regulatory T cell (Treg) suppression. Using PD1-IL2v in orthotopic PDAC KPC-driven tumor models, we show marked improvement in local and metastatic survival, along with a profound increase in tumor-infiltrating CD8+ T cell subsets with a transcriptionally and metabolically active phenotype and preferential activation of antigen-specific CD8+ T cells. In combination with single-dose RT, PD1-IL2v treatment results in a robust, durable expansion of polyfunctional CD8+ T cells, T cell stemness, tumor-specific memory immune response, natural killer (NK) cell activation, and decreased Tregs. These data show that PD1-IL2v leads to profound local and distant response in PDAC.
Collapse
Affiliation(s)
- Miles Piper
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Maureen Hoen
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Microbiology and Immunology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael W Knitz
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Greta Durini
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Idil Karakoc
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Abby Grier
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooke Neupert
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Khalid N M Abdelazeem
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Justin Yu
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicholas A Olimpo
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sophia Corbo
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Richard Blake Ross
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tiffany T Pham
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Molishree Joshi
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ross M Kedl
- Department of Microbiology and Immunology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Laura Codarri Deak
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Microbiology and Immunology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
45
|
Ko B, Takebe N, Andrews O, Makena MR, Chen AP. Rethinking Oncologic Treatment Strategies with Interleukin-2. Cells 2023; 12:cells12091316. [PMID: 37174716 PMCID: PMC10177415 DOI: 10.3390/cells12091316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
High-dose recombinant human IL-2 (rhIL-2, aldesleukin) emerged as an important treatment option for selected patients with metastatic melanoma and metastatic renal cell carcinoma, producing durable and long-lasting antitumor responses in a small fraction of patients and heralding the potential of cancer immunotherapy. However, the adoption of high-dose rhIL-2 has been restricted by its severe treatment-related adverse event (TRAE) profile, which necessitates highly experienced clinical providers familiar with rhIL-2 administration and readily accessible critical care medicine support. Given the comparatively wide-ranging successes of immune checkpoint inhibitors and chimeric antigen receptor T cell therapies, there have been concerted efforts to significantly improve the efficacy and toxicities of IL-2-based immunotherapeutic approaches. In this review, we highlight novel drug development strategies, including biochemical modifications and engineered IL-2 variants, to expand the narrow therapeutic window of IL-2 by leveraging downstream activation of the IL-2 receptor to selectively expand anti-tumor CD8-positive T cells and natural killer cells. These modified IL-2 cytokines improve single-agent activity in solid tumor malignancies beyond the established United States Food and Drug Administration (FDA) indications of metastatic melanoma and renal cell carcinoma, and may also be safer in rational combinations with established treatment modalities, including anti-PD-(L)1 and anti-CTLA-4 immunotherapy, chemotherapies, and targeted therapy approaches.
Collapse
Affiliation(s)
- Brian Ko
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| | - Naoko Takebe
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| | - Omozusi Andrews
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| | - Monish Ram Makena
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| | - Alice P Chen
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
46
|
Wu K, Lyu F, Wu SY, Sharma S, Deshpande RP, Tyagi A, Zhao D, Xing F, Singh R, Watabe K. Engineering an active immunotherapy for personalized cancer treatment and prevention of recurrence. SCIENCE ADVANCES 2023; 9:eade0625. [PMID: 37126558 DOI: 10.1126/sciadv.ade0625] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Breast cancer has been shown to be resistant to immunotherapies. To overcome this challenge, we developed an active immunotherapy for personalized treatment based on a smart nanovesicle. This is achieved by anchoring membrane-bound bioactive interleukin 2 (IL2) and enriching T cell-promoting costimulatory factors on the surface of the dendritic cell-derived small extracellular vesicles. This nanovesicle also displays major histocompatibility complex-bound antigens inherited from tumor lysate-pulsed dendritic cell. When administrated, the surface-bound IL2 is able to guide the nanovesicle to lymphoid organs and activate the IL2 receptor on lymphocytes. Furthermore, it is able to perform antigen presentation in the replacement of professional antigen-presenting cells. This nanovesicle, named IL2-ep13nsEV, induced a strong immune reaction to rescue 50% of the mice in our humanized patient-derived xenografts, sensitized cancer cells to immune checkpoint inhibitor treatment, and prevented the recurrence of resected tumors. This paradigm presents a feasible strategy for the treatment and prevention of metastatic breast cancer.
Collapse
Affiliation(s)
- Kerui Wu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Feng Lyu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Sambad Sharma
- Department of Translation Biology, Auron Therapeutics, Newton, MA 02458, USA
| | - Ravindra Pramod Deshpande
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Abhishek Tyagi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Dan Zhao
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
47
|
Liu S, Sun Q, Ren X. Novel strategies for cancer immunotherapy: counter-immunoediting therapy. J Hematol Oncol 2023; 16:38. [PMID: 37055849 PMCID: PMC10099030 DOI: 10.1186/s13045-023-01430-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/21/2023] [Indexed: 04/15/2023] Open
Abstract
The advent of immunotherapy has made an indelible mark on the field of cancer therapy, especially the application of immune checkpoint inhibitors in clinical practice. Although immunotherapy has proven its efficacy and safety in some tumors, many patients still have innate or acquired resistance to immunotherapy. The emergence of this phenomenon is closely related to the highly heterogeneous immune microenvironment formed by tumor cells after undergoing cancer immunoediting. The process of cancer immunoediting refers to the cooperative interaction between tumor cells and the immune system that involves three phases: elimination, equilibrium, and escape. During these phases, conflicting interactions between the immune system and tumor cells result in the formation of a complex immune microenvironment, which contributes to the acquisition of different levels of immunotherapy resistance in tumor cells. In this review, we summarize the characteristics of different phases of cancer immunoediting and the corresponding therapeutic tools, and we propose normalized therapeutic strategies based on immunophenotyping. The process of cancer immunoediting is retrograded through targeted interventions in different phases of cancer immunoediting, making immunotherapy in the context of precision therapy the most promising therapy to cure cancer.
Collapse
Affiliation(s)
- Shaochuan Liu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, 300060, Tianjin, China
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, 300060, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, 300060, Tianjin, China
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, 300060, Tianjin, China.
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, 300060, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, 300060, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, 300060, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China.
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, 300060, Tianjin, China.
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, 300060, Tianjin, China.
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, 300060, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, 300060, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, 300060, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China.
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, 300060, Tianjin, China.
| |
Collapse
|
48
|
Wang X, Wu X, Zhang P, Zhou Y, Cai J, Jin L. Single-cell transcriptome profiling reveals enriched memory T-cell subpopulations in hypertension. Front Cell Dev Biol 2023; 11:1132040. [PMID: 37009484 PMCID: PMC10060952 DOI: 10.3389/fcell.2023.1132040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction: The adaptive immune response mediated by T cells plays a vital role in the initiation and maintenance of blood pressure (BP) elevation. Memory T cells, which are antigen-specific T cells, can respond specifically to repeated hypertensive stimuli. Although the roles of memory T cells in animal models are well studied, their maintenance and functions in hypertensive patients are poorly understood.Method: Here, we focused on the circulating memory T cells of hypertensive patients. By using single-cell RNA sequencing technology, subsets of memory T cells were identified. Differentially expressed genes (DEGs) and functional pathways were explored for related biological functions in each population of memory T cells.Result and Discussion: Our study identified four subsets of memory T cells in the blood of hypertensive patients, with CD8 effector memory T (TEM) cells accounting for more cells and demonstrating more biological functions than CD4 TEM cells. CD8 TEM cells were further analyzed using single-cell RNA sequencing technology, and subpopulation 1 was demonstrated to contribute to BP elevation. The key marker genes CKS2, PLIN2, and CNBP were identified and validated by mass-spectrum flow cytometry. Our data suggest that CD8 TEM cells as well as the marker genes could be preventive targets for patients with hypertensive cardiovascular disease.
Collapse
Affiliation(s)
- Xiaoqi Wang
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
| | - Xiaobin Wu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Ministry of Education Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Beijing, China
| | - Pei Zhang
- Department of Hypertension, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Henan, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Ministry of Education Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Beijing, China
- *Correspondence: Yuan Zhou, ; Jun Cai, ; Ling Jin,
| | - Jun Cai
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
- *Correspondence: Yuan Zhou, ; Jun Cai, ; Ling Jin,
| | - Ling Jin
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- *Correspondence: Yuan Zhou, ; Jun Cai, ; Ling Jin,
| |
Collapse
|
49
|
Leung D, Cohen CA, Mu X, Rosa Duque J, Cheng SMS, Wang X, Wang M, Zhang W, Zhang Y, Tam I, Lam JHY, Chan SM, Chaothai S, Kwan KKH, Chan KCK, Li J, Luk LLH, Tsang LCH, Chu N, Wong WHS, Mori M, Leung W, Valkenburg S, Peiris M, Tu W, Lau YL. Immunogenicity against wild-type and Omicron SARS-CoV-2 after a third dose of inactivated COVID-19 vaccine in healthy adolescents. Front Immunol 2023; 14:1106837. [PMID: 36949953 PMCID: PMC10026957 DOI: 10.3389/fimmu.2023.1106837] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Introduction Two doses of inactivated SARS-CoV-2 vaccine CoronaVac cannot elicit high efficacy against symptomatic COVID-19, especially against the Omicron variant, but that can be improved by a third dose in adults. The use of a third dose of CoronaVac in adolescents may be supported by immunobridging studies in the absence of efficacy data. Methods With an immunobridging design, our study (NCT04800133) tested the non-inferiority of the binding and neutralizing antibodies and T cell responses induced by a third dose of CoronaVac in healthy adolescents (N=94, median age 14.2 years, 56% male) compared to adults (N=153, median age 48.1 years, 44% male). Responses against wild-type (WT) and BA.1 SARS-CoV-2 were compared in adolescents. Safety and reactogenicity were also monitored. Results A homologous third dose of CoronaVac further enhanced antibody response in adolescents compared to just 2 doses. Adolescents mounted non-inferior antibody and T cell responses compared to adults. Although S IgG and neutralizing antibody responses to BA.1 were lower than to WT, they remained detectable in 96% and 86% of adolescents. T cell responses to peptide pools spanning only the mutations of BA.1 S, N and M in adolescents were preserved, increased, and halved compared to WT respectively. No safety concerns were identified. Discussion The primary vaccination series of inactivated SARS-CoV-2 vaccines for adolescents should include 3 doses for improved humoral immunogenicity.
Collapse
Affiliation(s)
- Daniel Leung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Carolyn A. Cohen
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiaofeng Mu
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jaime S. Rosa Duque
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Samuel M. S. Cheng
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiwei Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Manni Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wenyue Zhang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yanmei Zhang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Issan Y. S. Tam
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jennifer H. Y. Lam
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sau Man Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sara Chaothai
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kelvin K. H. Kwan
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Karl C. K. Chan
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - John K. C. Li
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Leo L. H. Luk
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Leo C. H. Tsang
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nym Coco Chu
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wilfred H. S. Wong
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Masashi Mori
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Japan
| | - Wing Hang Leung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sophie Valkenburg
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Malik Peiris
- School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Centre for Immunology & Infection C2i, Hong Kong, Hong Kong SAR, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
50
|
Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neurooncol 2023; 162:93-108. [PMID: 36854924 DOI: 10.1007/s11060-023-04257-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Exosomes are nano-vesicular carriers capable of delivering cargoes for intercellular communication, which holds potential as biocompatible and high efficiency systems for drug delivery. In this study, we evaluated the potential effect of T7 peptide-decorated exosome-loaded Galectin-9 siRNA (T7-Exo/siGalectin-9) in the M1 polarization of macrophages and immunosuppression of glioblastoma (GBM). METHODS Differentially expressed genes in GBM were in silico predicted and then experimentally verified. Galectin-9 was knocked down by siRNA to assess its role in tumor-bearing mice. T7 peptide-decorated exosomes (derived from human embryonic kidney [HEK]-293T cells) targeting GBM were prepared, and loaded with Galectin-9 siRNA by electroporation to prepare nanoformulations (T7-Exo/siGalectin-9). The role of T7-Exo/siGalectin-9 in CD8+ T cell cytotoxicity to target GBM cells and polarization of macrophages was evaluated after artificial modulation of Galectin-9 expression. Anti-tumor effects of T7-Exo/siGalectin-9 were elucidated in vitro and in vivo. RESULTS Galectin-9 was highly expressed in GBM tissues and cell lines. The siRNA-mediated knockdown of Galectin-9 repressed the growth of xenografts of GBM cells in C57BL/6 mice and activated immune response in the tumor microenvironment. T7-Exo/siGalectin-9 effectively delivered siGalectin-9 to GBM cells. T7-Exo/siGalectin-9 contributed to activation of the TLR7-IRF5 pathway, which polarized macrophages to M1 phenotype. By this mechanism, phagocytosis of GBM cells by macrophages was increased, the anti-tumor effect of CD8+ T cells was enhanced and the inflammatory responses were suppressed. CONCLUSION Overall, T7-Exo/siGalectin-9 promotes macrophage repolarization and restricts the immunosuppression of GBM, thus providing novel insights into and drug delivery system of immunotherapy for GBM.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Ning Guan
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121000, Liaoning, People's Republic of China.
| |
Collapse
|