1
|
Grimus S, Sarangova V, Welzel PB, Ludwig B, Seissler J, Kemter E, Wolf E, Ali A. Immunoprotection Strategies in β-Cell Replacement Therapy: A Closer Look at Porcine Islet Xenotransplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401385. [PMID: 38884159 PMCID: PMC11336975 DOI: 10.1002/advs.202401385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/28/2024] [Indexed: 06/18/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by absolute insulin deficiency primarily due to autoimmune destruction of pancreatic β-cells. The prevailing treatment for T1DM involves daily subcutaneous insulin injections, but a substantial proportion of patients face challenges such as severe hypoglycemic episodes and poorly controlled hyperglycemia. For T1DM patients, a more effective therapeutic option involves the replacement of β-cells through allogeneic transplantation of either the entire pancreas or isolated pancreatic islets. Unfortunately, the scarcity of transplantable human organs has led to a growing list of patients waiting for an islet transplant. One potential alternative is xenotransplantation of porcine pancreatic islets. However, due to inter-species molecular incompatibilities, porcine tissues trigger a robust immune response in humans, leading to xenograft rejection. Several promising strategies aim to overcome this challenge and enhance the long-term survival and functionality of xenogeneic islet grafts. These strategies include the use of islets derived from genetically modified pigs, immunoisolation of islets by encapsulation in biocompatible materials, and the creation of an immunomodulatory microenvironment by co-transplanting islets with accessory cells or utilizing immunomodulatory biomaterials. This review concentrates on delineating the primary obstacles in islet xenotransplantation and elucidates the fundamental principles and recent breakthroughs aimed at addressing these challenges.
Collapse
Affiliation(s)
- Sarah Grimus
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| | - Victoria Sarangova
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Petra B. Welzel
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Barbara Ludwig
- Department of Medicine IIIUniversity Hospital Carl Gustav CarusTechnische Universität DresdenD‐01307DresdenGermany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the Technische Universität DresdenD‐01307DresdenGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
- DFG‐Center for Regenerative Therapies DresdenTechnische Universität DresdenD‐01307DresdenGermany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IVDiabetes Zentrum – Campus InnenstadtKlinikum der Ludwig‐Maximilians‐Universität MünchenD‐80336MunichGermany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Asghar Ali
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| |
Collapse
|
2
|
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
|
3
|
Cafaro A, Schietroma I, Sernicola L, Belli R, Campagna M, Mancini F, Farcomeni S, Pavone-Cossut MR, Borsetti A, Monini P, Ensoli B. Role of HIV-1 Tat Protein Interactions with Host Receptors in HIV Infection and Pathogenesis. Int J Mol Sci 2024; 25:1704. [PMID: 38338977 PMCID: PMC10855115 DOI: 10.3390/ijms25031704] [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: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Each time the virus starts a new round of expression/replication, even under effective antiretroviral therapy (ART), the transactivator of viral transcription Tat is one of the first HIV-1 protein to be produced, as it is strictly required for HIV replication and spreading. At this stage, most of the Tat protein exits infected cells, accumulates in the extracellular matrix and exerts profound effects on both the virus and neighbor cells, mostly of the innate and adaptive immune systems. Through these effects, extracellular Tat contributes to the acquisition of infection, spreading and progression to AIDS in untreated patients, or to non-AIDS co-morbidities in ART-treated individuals, who experience inflammation and immune activation despite virus suppression. Here, we review the role of extracellular Tat in both the virus life cycle and on cells of the innate and adaptive immune system, and we provide epidemiological and experimental evidence of the importance of targeting Tat to block residual HIV expression and replication. Finally, we briefly review vaccine studies showing that a therapeutic Tat vaccine intensifies ART, while its inclusion in a preventative vaccine may blunt escape from neutralizing antibodies and block early events in HIV acquisition.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (L.S.); (R.B.); (M.C.); (F.M.); (S.F.); (M.R.P.-C.); (A.B.); (P.M.)
| | | | | | | | | | | | | | | | | | | | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (L.S.); (R.B.); (M.C.); (F.M.); (S.F.); (M.R.P.-C.); (A.B.); (P.M.)
| |
Collapse
|
4
|
Sizova O, John LS, Ma Q, Molldrem JJ. Multi-faceted role of LRP1 in the immune system. Front Immunol 2023; 14:1166189. [PMID: 37020553 PMCID: PMC10069629 DOI: 10.3389/fimmu.2023.1166189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Graft versus host disease (GVHD) represents the major complication after allogeneic hematopoietic stem cell transplantation (Allo-SCT). GVHD-prone patients rely on GVHD prophylaxis (e.g. methotrexate) and generalized anti-GVHD medical regimen (glucocorticoids). New anti-GVHD therapy strategies are being constantly explored, however there is an urgent need to improve current treatment, since GVHD-related mortality reaches 22% within 5 years in patients with chronic GVHD. This review is an attempt to describe a very well-known receptor in lipoprotein studies - the low-density lipoprotein receptor related protein 1 (LRP1) - in a new light, as a potential therapeutic target for GVHD prevention and treatment. Our preliminary studies demonstrated that LRP1 deletion in donor murine T cells results in significantly lower GVHD-related mortality in recipient mice with MHC (major histocompatibility complex) -mismatched HSCT. Given the importance of T cells in the development of GVHD, there is a significant gap in scientific literature regarding LRP1's role in T cell biology. Furthermore, there is limited research interest and publications on this classical receptor molecule in other immune cell types. Herein, we endeavor to summarize existing knowledge about LRP1's role in various immune cells to demonstrate the possibility of this receptor to serve as a novel target for anti-GVHD treatment.
Collapse
Affiliation(s)
- Olga Sizova
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lisa St. John
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qing Ma
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jeffrey J. Molldrem
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- ECLIPSE, Therapeutic Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Jeffrey J. Molldrem,
| |
Collapse
|
5
|
PRSS2 remodels the tumor microenvironment via repression of Tsp1 to stimulate tumor growth and progression. Nat Commun 2022; 13:7959. [PMID: 36575174 PMCID: PMC9794699 DOI: 10.1038/s41467-022-35649-9] [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: 06/11/2021] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
The progression of cancer from localized to metastatic disease is the primary cause of morbidity and mortality. The interplay between the tumor and its microenvironment is the key driver in this process of tumor progression. In order for tumors to progress and metastasize they must reprogram the cells that make up the microenvironment to promote tumor growth and suppress endogenous defense systems, such as the immune and inflammatory response. We have previously demonstrated that stimulation of Tsp-1 in the tumor microenvironment (TME) potently inhibits tumor growth and progression. Here, we identify a novel tumor-mediated mechanism that represses the expression of Tsp-1 in the TME via secretion of the serine protease PRSS2. We demonstrate that PRSS2 represses Tsp-1, not via its enzymatic activity, but by binding to low-density lipoprotein receptor-related protein 1 (LRP1). These findings describe a hitherto undescribed activity for PRSS2 through binding to LRP1 and represent a potential therapeutic strategy to treat cancer by blocking the PRSS2-mediated repression of Tsp-1. Based on the ability of PRSS2 to reprogram the tumor microenvironment, this discovery could lead to the development of therapeutic agents that are indication agnostic.
Collapse
|
6
|
Lei L, Huang D, Gao H, He B, Cao J, Peppas NA. Hydrogel-guided strategies to stimulate an effective immune response for vaccine-based cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eadc8738. [PMID: 36427310 PMCID: PMC9699680 DOI: 10.1126/sciadv.adc8738] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/07/2022] [Indexed: 05/25/2023]
Abstract
Cancer vaccines have attracted widespread interest in tumor therapy because of the potential to induce an effective antitumor immune response. However, many challenges including weak immunogenicity, off-target effects, and immunosuppressive microenvironments have prevented their broad clinical translation. To overcome these difficulties, effective delivery systems have been designed for cancer vaccines. As carriers in cancer vaccine delivery systems, hydrogels have gained substantial attention because they can encapsulate a variety of antigens/immunomodulators and protect them from degradation. This enables hydrogels to simultaneously reverse immunosuppression and stimulate the immune response. Meanwhile, the controlled release properties of hydrogels allow for precise temporal and spatial release of loads in situ to further enhance the immune response of cancer vaccines. Therefore, this review summarizes the classification of cancer vaccines, highlights the strategies of hydrogel-based cancer vaccines, and provides some insights into the future development of hydrogel-based cancer vaccines.
Collapse
Affiliation(s)
- Lei Lei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Dennis Huang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, USA
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
- Departments of Pediatrics, Surgery, and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
7
|
Tombácz I, Laczkó D, Shahnawaz H, Muramatsu H, Natesan A, Yadegari A, Papp TE, Alameh MG, Shuvaev V, Mui BL, Tam YK, Muzykantov V, Pardi N, Weissman D, Parhiz H. Highly efficient CD4+ T cell targeting and genetic recombination using engineered CD4+ cell-homing mRNA-LNP. Mol Ther 2021; 29:3293-3304. [PMID: 34091054 PMCID: PMC8571164 DOI: 10.1016/j.ymthe.2021.06.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/30/2021] [Accepted: 06/01/2021] [Indexed: 11/23/2022] Open
Abstract
Nucleoside-modified messenger RNA (mRNA)-lipid nanoparticles (LNPs) are the basis for the first two EUA (Emergency Use Authorization) COVID-19 vaccines. The use of nucleoside-modified mRNA as a pharmacological agent opens immense opportunities for therapeutic, prophylactic and diagnostic molecular interventions. In particular, mRNA-based drugs may specifically modulate immune cells, such as T lymphocytes, for immunotherapy of oncologic, infectious and other conditions. The key challenge, however, is that T cells are notoriously resistant to transfection by exogenous mRNA. Here, we report that conjugating CD4 antibody to LNPs enables specific targeting and mRNA interventions to CD4+ cells, including T cells. After systemic injection in mice, CD4-targeted radiolabeled mRNA-LNPs accumulated in spleen, providing ∼30-fold higher signal of reporter mRNA in T cells isolated from spleen as compared with non-targeted mRNA-LNPs. Intravenous injection of CD4-targeted LNPs loaded with Cre recombinase-encoding mRNA provided specific dose-dependent loxP-mediated genetic recombination, resulting in reporter gene expression in about 60% and 40% of CD4+ T cells in spleen and lymph nodes, respectively. T cell phenotyping showed uniform transfection of T cell subpopulations, with no variability in uptake of CD4-targeted mRNA-LNPs in naive, central memory, and effector cells. The specific and efficient targeting and transfection of mRNA to T cells established in this study provides a platform technology for immunotherapy of devastating conditions and HIV cure.
Collapse
Affiliation(s)
- István Tombácz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dorottya Laczkó
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamna Shahnawaz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ambika Natesan
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amir Yadegari
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler E Papp
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohamad-Gabriel Alameh
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir Shuvaev
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Vladimir Muzykantov
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Norbert Pardi
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamideh Parhiz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Tan S, Li S, Min Y, Gisterå A, Moruzzi N, Zhang J, Sun Y, Andersson J, Malmström RE, Wang M, Berggren PO, Schlisio S, Liao W, Ketelhuth DFJ, Ma C, Li N. Platelet factor 4 enhances CD4 + T effector memory cell responses via Akt-PGC1α-TFAM signaling-mediated mitochondrial biogenesis. J Thromb Haemost 2020; 18:2685-2700. [PMID: 32671959 DOI: 10.1111/jth.15005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/10/2020] [Accepted: 07/08/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cell metabolism drives T cell functions, while platelets regulate overall CD4+ T cell immune responses. OBJECTIVE To investigate if platelets influence cell metabolism and thus regulate CD4+ T effector memory cell (Tem) responses. METHODS Human CD4+ Tem cells were activated with αCD3/αCD28 and cultured without or with platelets or platelet-derived mediators. RESULTS Polyclonal stimulation induced rapid and marked Th1 and Treg cell activation of CD4+ Tem cells. Platelet co-culture enhanced Th1 response transiently, while it persistently enhanced Treg cell activation of Tem cells, with an enhancement that plateaued by day 3. Platelet factor 4 (PF4) was the key platelet-derived mediator regulating CD4+ Tem cell responses, which involved cellular metabolisms as indicated by mass spectrometric analyses. PF4 exerted its effects via its receptor CXCR3, attenuated Akt activity, and reduced PGC1α phosphorylation, and resulted in elevations of PGC1α function and mitochondrial transcription factor A (TFAM) synthesis. The latter increased mitochondrial biogenesis, and subsequently enhanced Th1 and Treg responses. Consistent with these observations, inhibition of mitochondrial function by rotenone counteracted the enhancements by recombinant PF4, and TFAM overexpression by TFAM-adenovirus infection mimicked PF4 effects. Furthermore, increased mitochondrial mass elevated oxygen consumption, and enhanced adenosine triphosphate and reactive oxygen species production, which, in turn, stimulated Th1 (T-bet) and Treg (FoxP3) transcription factor expression and corresponding CD4+ T effector cell responses. CONCLUSIONS Platelets enhance CD4+ T cell responses of Tem cells through PF4-dependent and Akt-PGC1α-TFAM signaling-mediated mitochondrial biogenesis. Hence, PF4 may be a promising intervention target of platelet-regulated immune responses.
Collapse
Affiliation(s)
- Shuai Tan
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - Shuijie Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yanan Min
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - Anton Gisterå
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Noah Moruzzi
- Department of Molecular Medicine and Surgery, Rolf Ruft Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Junhao Zhang
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Sun
- Shandong University Cheeloo Medical College, Institute of Immunology, Jinan, China
| | - John Andersson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rickard E Malmström
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Clinical Pharmacology, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Miao Wang
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Per-Olof Berggren
- Department of Molecular Medicine and Surgery, Rolf Ruft Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Schlisio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Daniel F J Ketelhuth
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chunhong Ma
- Shandong University Cheeloo Medical College, Institute of Immunology, Jinan, China
| | - Nailin Li
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|