1
|
Jin M, Ye K, Hu D, Chen J, Wu S, Chi S. Identification of diagnose related therapeutic targets of Danggui buxue decoction in Parkinson's disease. Brain Res 2024; 1842:149097. [PMID: 38950810 DOI: 10.1016/j.brainres.2024.149097] [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: 03/13/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is the fastest growing neurological disease. Currently, there is no disease-modifying therapy to slow the progression of the disease. Danggui buxue decoction (DBD) is widely used in the clinic because of its therapeutic effect. However, little is known about the molecular mechanism of DBD against PD. This study intends to explore the possible molecular mechanisms involved in DBD treatment of PD based on network pharmacology, and provide potential research directions for future research. METHODS Firstly, the active components and target genes of DBD were screened from the traditional Chinese medicine systems pharmacology (TCMSP), DrugBank and UniProt database. Secondly, target genes of PD were identified from the (GEO) dataset, followed by identification of common target genes of DBD and PD. Thirdly, analysis of protein-protein interaction (PPI), functional enrichment and diagnosis was performed on common target genes, followed by correlation analysis between core target genes, immune cell, miRNAs, and transcription factors (TFs). Finally, molecular docking between core target genes and active components, and real-time PCR were performed. RESULTS A total of 72 common target genes were identified between target genes of DBD and target genes of PD. Among which, 11 target genes with potential diagnostic value were further identified, including TP53, AKT1, IL1B, MMP9, NOS3, RELA, MAPK14, HMOX1, TGFB1, NOS2, and ERBB2. The combinations with the best docking binding were identified, including kaempferol-AKT1/HMOX1/NOS2/NOS3, quercetin-AKT1/ERBB2/IL1B/HMOX1/MMP9/TP53/NOS3/TGFB1. Moreover, IL1B and NOS2 respectively positively and negatively correlated with neutrophil and Type 1 T helper cell. Some miRNA-core target gene regulatory pairs were identified, such as hsa-miR-185-5p-TP53/TGFB1/RELA/MAPK14/IL1B/ERBB2/AKT1 and hsa-miR-214-3p-NOS3. These core target genes were significantly enriched in focal adhesion, TNF, HIF-1, and ErbB signaling pathway. CONCLUSION Diagnostic TP53, AKT1, IL1B, MMP9, NOS3, RELA, MAPK14, HMOX1, TGFB1, NOS2, and ERBB2 may be considered as potential therapeutic targets of DBD in the treatment of PD.
Collapse
Affiliation(s)
- Man Jin
- Department of Neurology, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310063, China
| | - Kaisheng Ye
- Department of Traditional Chinese Medicine, Hangzhou Kanghui Integrated Traditional and Western Medicine Clinic, Hangzhou, Zhejiang Province 310019, China.
| | - Defeng Hu
- Department of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310063, China
| | - Jiefang Chen
- Department of Neurology, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310063, China
| | - Sha Wu
- Intensive Care Units, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310063, China
| | - Shumei Chi
- Department of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310063, China
| |
Collapse
|
2
|
Zhang X, Yi K, Wang B, Chu K, Liu J, Zhang J, Fang J, Zhao T. EZH2 Activates HTLV-1 bZIP Factor-Mediated TGF-β Signaling in Adult T-Cell Leukemia. J Med Virol 2024; 96:e70025. [PMID: 39530290 DOI: 10.1002/jmv.70025] [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: 08/05/2024] [Revised: 09/20/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Adult T-cell leukemia (ATL) is an aggressive malignancy caused by human T-cell leukemia virus type 1 (HTLV-1) infection. Enhancer of zeste homolog 2 (EZH2) has been implicated in the development and progression of multiple cancers, including virus-induced malignancies. However, the potential function of EZH2 in HTLV-1-induced oncogenesis has not been clearly elucidated. In the present study, we showed that EZH2 was overexpressed and activated in HTLV-1-infected cell lines, potentially due to the activation of EZH2 promoter by HTLV-1 Tax and NF-κB p65 subunit. In addition, we found that EZH2 enhanced the HBZ-induced activation of TGF-β signaling in a histone methyltransferase-independent manner. As a mechanism for these actions, we found that EZH2 targeted Smad3/Smad4 to form a ternary complex, and the association between Smad3 and Smad4 was markedly enhanced in the presence of EZH2. Knockdown of EZH2 in ATL cells indeed repressed the expressions of the TGF-β target genes. In particular, EZH2 synergistically enhanced the HBZ/TGF-β-induced Foxp3 expression. Treatment of 3-Deazaneplanocin A, a specific inhibitor of EZH2 significantly inhibited the Foxp3 expression. Taken together, our results suggest that EZH2 may be involved in the differentiation of regulatory T cells through activating the HBZ-Smad3-TGF-β signaling axis, which is considered to be a key strategy for viral persistence.
Collapse
Affiliation(s)
- Xu Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Kaining Yi
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Bingbing Wang
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Kaifei Chu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Liu
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jiaqi Fang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Tiejun Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
3
|
Umeda M, Karino K, Satyam A, Yoshida N, Hisada R, Bhargava R, Vichos T, Kunzler AL, Igawa T, Ichinose K, Torigoe K, Nishino T, Maeda T, Owen CA, Abdi R, Kawakami A, Tsokos GC. Hypoxia Promotes the Expression of ADAM9 by Tubular Epithelial Cells, Which Enhances Transforming Growth Factor β1 Activation and Promotes Tissue Fibrosis in Patients With Lupus Nephritis. Arthritis Rheumatol 2024. [PMID: 39279154 DOI: 10.1002/art.42987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Enhanced expression of transforming growth factor (TGF) β in the kidneys of patients with lupus nephritis (LN) can lead to progressive fibrosis, resulting in end-organ damage. ADAM9 activates TGFβ1 by cleaving the latency-associated peptide (LAP). We hypothesized that ADAM9 in the kidney may accelerate fibrogenesis by activating TGFβ1. METHODS We assessed the expression of ADAM9 in the kidneys of mice and humans who were lupus prone. In vitro experiments were conducted using tubular epithelial cells (TECs) isolated from mice and explored the mechanisms responsible for the up-regulation of ADAM9 and the subsequent activation of TGFβ1. To assess the role of ADAM9 in the development of tubular-intestinal fibrosis in individuals with LN, we generated MRL/lpr mice who were Adam9 deficient. RESULTS ADAM9 was highly expressed in tubules from MRL/lpr mice. The transcription factor hypoxia-inducible factor-1α was found to promote the transcription of ADAM9 in TECs. TECs from mice who were Adam9 deficient and exposed to the hypoxia mimetic agent dimethyloxalylglycine failed to cleave the LAP to produce bioactive TGFβ1 from latent TGFβ1. Coculture of TECs from mice who were Adam9 deficient with fibroblasts in the presence of dimethyloxalylglycine and latent TGFβ1 produced decreased amounts of type I collagen and α-smooth muscle actin (SMA) by fibroblasts. MRL/lpr mice who were Adam9 deficient showed reduced interstitial fibrosis. At the translational level, ADAM9 expression in tissues and urine of patients with LN was found to increase. CONCLUSION Hypoxia promotes the expression of ADAM9 by TECs, which is responsible for the development of interstitial fibrosis in patients with LN by enhancing the TGFβ1 activation, which promotes fibroblasts to produce collagen and α-SMA.
Collapse
Affiliation(s)
- Masataka Umeda
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, and Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kohei Karino
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Abhigyan Satyam
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Nobuya Yoshida
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ryo Hisada
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Rhea Bhargava
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Theodoros Vichos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ana Laura Kunzler
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Takashi Igawa
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kunihiro Ichinose
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, and Shimane University Faculty of Medicine, Izumo, Japan
| | | | | | - Takahiro Maeda
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Caroline A Owen
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Reza Abdi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Atsushi Kawakami
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4
|
Boonpethkaew S, Meephansan J, Ponnikorn S, Jumlongpim O, Juntongjin P, Chakkavittumrong P, Wongpiyabovorn J, Morita A, Komine M. Exploring the role of growth factors as potential regulators in psoriatic plaque formation. Exp Dermatol 2023; 32:1924-1934. [PMID: 37665186 DOI: 10.1111/exd.14918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease in which growth activity is more prominent than inflammatory activity at the centre of lesional skin (CE skin). This growth activity is partly influenced by growth factors (GFs) that play an important role in cell growth and inflammation during the plaque development. In this study, we identified potential GFs in CE skin and predicted their regulatory functions and biological activity in mediating transcripts in the plaques. Samples of uninvolved skin (UN skin) and CE skin were biopsied from patients with psoriasis vulgaris for RNA-sequencing analysis in order to identify differentially expressed genes (DEGs). Our finding revealed that epidermal growth factor (EGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF) and hepatocyte growth factor (HGF) signalling were enriched by CE/UN skin-derived DEGs. Additionally, several EGFR ligands, namely EGF, heparin-binding EGF like growth factor (HB-EGF), amphiregulin (AREG) and transforming growth factor (TGF)-α, as well as TGF-β1, TGF-β2, vascular endothelial growth factor-A, FGFs, PDGF-B and HGF, were predicted to be GF regulators. The regulatory pattern and biological activity of these GF regulators on mediating the CE/UN skin-derived DEGs was demonstrated. This study provides a novel hypothesis regarding the overall regulatory function of GFs, which appear to modulate the expression of the transcripts involved in inflammation and growth in the CE skin. In addition, some GFs may exert anti-inflammatory effects. Further investigations on the mechanisms underlying this regulation may contribute to a deeper understanding of psoriasis and the identification of potential therapeutic targets for patients with psoriasis.
Collapse
Affiliation(s)
- Suphagan Boonpethkaew
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Thammasat University, Chonburi, Thailand
| | - Jitlada Meephansan
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Saranyoo Ponnikorn
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Thammasat University, Chonburi, Thailand
| | - Onjira Jumlongpim
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Premjit Juntongjin
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Panlop Chakkavittumrong
- Division of Dermatology, Department of Internal Medicine, Thammasat University, Pathum Thani, Thailand
| | - Jongkonnee Wongpiyabovorn
- Center of Excellence in Immunology and Immune-Mediated Disease, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mayumi Komine
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
5
|
Singh S, Gouri V, Samant M. TGF-β in correlation with tumor progression, immunosuppression and targeted therapy in colorectal cancer. Med Oncol 2023; 40:335. [PMID: 37855975 DOI: 10.1007/s12032-023-02204-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Colorectal cancer (CRC) is a complex malignancy responsible for the second-highest cancer deaths worldwide. TGF-β maintains normal cellular homeostasis by inhibiting the cell cycle and inducing apoptosis, but its elevated level is correlated with colorectal cancer progression, as TGF-β is a master regulator of the epithelial-to-mesenchymal transition, a critical step of metastasis. Tumors, including CRC, use elevated TGF-β levels to avoid immune surveillance by modulating immune cell differentiation, proliferation, and effector function. Presently, the treatment of advanced CRC is mainly based on chemotherapy, with multiple adverse effects. Thus, there is a need to develop alternate tactics because CRC continue to be mostly resistant to the present therapeutic regimen. TGF-β blockade has emerged as a promising therapeutic target in cancer therapy. Blocking TGF-β with phytochemicals and other molecules, such as antisense oligonucleotides, monoclonal antibodies, and bifunctional traps, alone or in combination, may be a safer and more effective way to treat CRC. Furthermore, combination immunotherapy comprising TGF-β blockers and immune checkpoint inhibitors is gaining popularity because both molecules work synergistically to suppress the immune system. Here, we summarize the current understanding of TGF-β as a therapeutic target for managing CRC and its context-dependent tumor-promoting or tumor-suppressing nature.
Collapse
Affiliation(s)
- Sumeet Singh
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Vinita Gouri
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
- Department of Zoology, Kumaun University, Nainital, Uttarakhand, India
| | - Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India.
| |
Collapse
|
6
|
Zhang Z, Zhao S, Sun Z, Zhai C, Xia J, Wen C, Zhang Y, Zhang Y. Enhancement of the therapeutic efficacy of mesenchymal stem cell-derived exosomes in osteoarthritis. Cell Mol Biol Lett 2023; 28:75. [PMID: 37770821 PMCID: PMC10540339 DOI: 10.1186/s11658-023-00485-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Osteoarthritis (OA), a common joint disorder with articular cartilage degradation as the main pathological change, is the major source of pain and disability worldwide. Despite current treatments, the overall treatment outcome is unsatisfactory. Thus, patients with severe OA often require joint replacement surgery. In recent years, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic option for preclinical and clinical palliation of OA. MSC-derived exosomes (MSC-Exos) carrying bioactive molecules of the parental cells, including non-coding RNAs (ncRNAs) and proteins, have demonstrated a significant impact on the modulation of various physiological behaviors of cells in the joint cavity, making them promising candidates for cell-free therapy for OA. This review provides a comprehensive overview of the biosynthesis and composition of MSC-Exos and their mechanisms of action in OA. We also discussed the potential of MSC-Exos as a therapeutic tool for modulating intercellular communication in OA. Additionally, we explored bioengineering approaches to enhance MSC-Exos' therapeutic potential, which may help to overcome challenges and achieve clinically meaningful OA therapies.
Collapse
Affiliation(s)
- Zehao Zhang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Sheng Zhao
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhaofeng Sun
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Chuanxing Zhai
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yuge Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| |
Collapse
|
7
|
Boreddy SR, Nair R, Pandey PK, Kuriakose A, Marigowda SB, Dey C, Banerjee A, Kulkarni H, Sagar M, Krishn SR, Rao S, AR M, Tiwari V, Alke B, MV PK, Shri M, Dhamne C, Patel S, Sharma P, Periyasamy S, Bhatnagar J, Kuriakose MA, Reddy RB, Suresh A, Sreenivas S, Govindappa N, Moole PR, Bughani U, Tan SL, Nair P. BCA101 Is a Tumor-Targeted Bifunctional Fusion Antibody That Simultaneously Inhibits EGFR and TGFβ Signaling to Durably Suppress Tumor Growth. Cancer Res 2023; 83:1883-1904. [PMID: 37074042 PMCID: PMC10236157 DOI: 10.1158/0008-5472.can-21-4425] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 10/12/2022] [Accepted: 03/29/2023] [Indexed: 04/20/2023]
Abstract
The EGFR and TGFβ signaling pathways are important mediators of tumorigenesis, and cross-talk between them contributes to cancer progression and drug resistance. Therapies capable of simultaneously targeting EGFR and TGFβ could help improve patient outcomes across various cancer types. Here, we developed BCA101, an anti-EGFR IgG1 mAb linked to an extracellular domain of human TGFβRII. The TGFβ "trap" fused to the light chain in BCA101 did not sterically interfere with its ability to bind EGFR, inhibit cell proliferation, or mediate antibody-dependent cellular cytotoxicity. Functional neutralization of TGFβ by BCA101 was demonstrated by several in vitro assays. BCA101 increased production of proinflammatory cytokines and key markers associated with T-cell and natural killer-cell activation, while suppressing VEGF secretion. In addition, BCA101 inhibited differentiation of naïve CD4+ T cells to inducible regulatory T cells (iTreg) more strongly than the anti-EGFR antibody cetuximab. BCA101 localized to tumor tissues in xenograft mouse models with comparable kinetics to cetuximab, both having better tumor tissue retention over TGFβ "trap." TGFβ in tumors was neutralized by approximately 90% in animals dosed with 10 mg/kg of BCA101 compared with 54% in animals dosed with equimolar TGFβRII-Fc. In patient-derived xenograft mouse models of head and neck squamous cell carcinoma, BCA101 showed durable response after dose cessation. The combination of BCA101 and anti-PD1 antibody improved tumor inhibition in both B16-hEGFR-expressing syngeneic mouse models and in humanized HuNOG-EXL mice bearing human PC-3 xenografts. Together, these results support the clinical development of BCA101 as a monotherapy and in combination with immune checkpoint therapy. SIGNIFICANCE The bifunctional mAb fusion design of BCA101 targets it to the tumor microenvironment where it inhibits EGFR and neutralizes TGFβ to induce immune activation and to suppress tumor growth.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Meena Shri
- Biofusion Therapeutics, Bengaluru, India
| | | | | | | | | | | | - Moni Abraham Kuriakose
- Integrated Head and Neck Oncology Program, MSCTR, Mazumdar Shaw Medical Foundation, Bengaluru, India
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - Ram Bhupal Reddy
- Integrated Head and Neck Oncology Program, MSCTR, Mazumdar Shaw Medical Foundation, Bengaluru, India
| | - Amritha Suresh
- Integrated Head and Neck Oncology Program, MSCTR, Mazumdar Shaw Medical Foundation, Bengaluru, India
| | | | | | | | | | | | | |
Collapse
|
8
|
Xu B, Yin M, Yang Y, Zou Y, Liu W, Qiao L, Zhang J, Wang Z, Wu Y, Shen H, Sun M, Liu W, Xue W, Fan Y, Zhang Q, Chen B, Wu X, Shi Y, Lu F, Zhao Y, Xiao Z, Dai J. Transplantation of neural stem progenitor cells from different sources for severe spinal cord injury repair in rat. Bioact Mater 2023; 23:300-313. [DOI: 10.1016/j.bioactmat.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
|
9
|
Li Y, Xiang S, Pan W, Wang J, Zhan H, Liu S. Targeting tumor immunosuppressive microenvironment for pancreatic cancer immunotherapy: Current research and future perspective. Front Oncol 2023; 13:1166860. [PMID: 37064113 PMCID: PMC10090519 DOI: 10.3389/fonc.2023.1166860] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Pancreatic cancer is one of the most malignant tumors with increased incidence rate. The effect of surgery combined with chemoradiotherapy on survival of patients is unsatisfactory. New treatment strategy such as immunotherapy need to be investigated. The accumulation of desmoplastic stroma, infiltration of immunosuppressive cells including myeloid derived suppressor cells (MDSCs), tumor associated macrophages (TAMs), cancer‐associated fibroblasts (CAFs), and regulatory T cells (Tregs), as well as tumor associated cytokine such as TGF-β, IL-10, IL-35, CCL5 and CXCL12 construct an immunosuppressive microenvironment of pancreatic cancer, which presents challenges for immunotherapy. In this review article, we explore the roles and mechanism of immunosuppressive cells and lymphocytes in establishing an immunosuppressive tumor microenvironment in pancreatic cancer. In addition, immunotherapy strategies for pancreatic cancer based on tumor microenvironment including immune checkpoint inhibitors, targeting extracellular matrix (ECM), interfering with stromal cells or cytokines in TME, cancer vaccines and extracellular vesicles (EVs) are also discussed. It is necessary to identify an approach of immunotherapy in combination with other modalities to produce a synergistic effect with increased response rates in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Ying Li
- Department of Blood Transfusion, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuai Xiang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjun Pan
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Wang
- Department of Operating Room, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hanxiang Zhan
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong, China
- *Correspondence: Shanglong Liu, ; Hanxiang Zhan,
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shanglong Liu, ; Hanxiang Zhan,
| |
Collapse
|
10
|
Wang X, Wang H, Zhang S, Shang H, Wang C, Zhou F, Gao P, Zhu R, Hu L, Wei K. The role of transforming growth factor beta-1 protein in Escherichia coli secondary infection induced by H9N2 avian influenza virus in chickens. Microb Pathog 2023; 175:105983. [PMID: 36641002 DOI: 10.1016/j.micpath.2023.105983] [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: 11/02/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
The H9N2 subtype of avian influenza virus (AIV) is common in poultry production. It causes mild clinical signs but rarely leads to poultry mortalities. However, higher mortality can occur in chickens with co-infections, especially avian pathogenic Escherichia coli (APEC), which results in huge economic losses for the poultry industry. Unfortunately, the mechanism of co-infection remains unknown. Our previous studies screened several proteins associated with bacterial adhesion, including transforming growth factor beta-1 (TGF-β1), integrins, cortactin, E-cadherin, vinculin, and fibromodulin. Herein, we investigated the contribution of TGF-β1 to APEC adhesion after H9N2 infection. We first infected H9N2 and APEC in chicken, chicken embryo and DF-1 cells, and demonstrated that H9N2 infection promotes APEC adhesion to hosts in vitro and in vivo by plate count method. Through real-time fluorescence quantification and enzyme-linked immunosorbent assay, it was demonstrated that H9N2 infection not only increases TGF-β1 expression but also its activity in a time-dependent manner. Then, through exogenous addition of TGF-β1 and overexpression, we further demonstrated that TGF-β1 can increase the adhesion of endothelial cells to DF-1 cells. Furthermore, the capacity of APEC adhesion to DF-1 cells was significantly decreased either by adding a TGF-β1 receptor inhibitor or using small interfering RNAs to interfere with the expression of TGF-β1. To sum up, H9N2 infection can promote the upregulation of TGF-β1 and then increase the adhesion ability of APEC. Targeting TGF-β1 and its associated pathway will provide valuable insights into the clinical treatment of E. coli secondary infection induced by H9N2 infection.
Collapse
Affiliation(s)
- Xiangkun Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Huan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Shuyu Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Hongqi Shang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Cheng Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Fan Zhou
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Panpan Gao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Ruiliang Zhu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Liping Hu
- Animal Disease Prevention and Control Center of Shandong Province, Animal Husbandry and Veterinary Bureau of Shandong Province, Jinan, China.
| | - Kai Wei
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, China.
| |
Collapse
|
11
|
Ursodeoxycholic acid reduces antitumor immunosuppression by inducing CHIP-mediated TGF-β degradation. Nat Commun 2022; 13:3419. [PMID: 35701426 PMCID: PMC9198048 DOI: 10.1038/s41467-022-31141-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/27/2022] [Indexed: 12/12/2022] Open
Abstract
TGF-β is essential for inducing systemic tumor immunosuppression; thus, blocking TGF-β can greatly enhance antitumor immunity. However, there are still no effective TGF-β inhibitors in clinical use. Here, we show that the clinically approved compound ursodeoxycholic acid (UDCA), by degrading TGF-β, enhances antitumor immunity through restraining Treg cell differentiation and activation in tumor-bearing mice. Furthermore, UDCA synergizes with anti-PD-1 to enhance antitumor immunity and tumor-specific immune memory in tumor-bearing mice. UDCA phosphorylates TGF-β at T282 site via TGR5-cAMP-PKA axis, causing increased binding of TGF-β to carboxyl terminus of Hsc70-interacting protein (CHIP). Then, CHIP ubiquitinates TGF-β at the K315 site, initiating p62-dependent autophagic sorting and subsequent degradation of TGF-β. Notably, results of retrospective analysis shows that combination therapy with anti-PD-1 or anti-PD-L1 and UDCA has better efficacy in tumor patients than anti-PD-1 or anti-PD-L1 alone. Thus, our results show a mechanism for TGF-β regulation and implicate UDCA as a potential TGF-β inhibitor to enhance antitumor immunity.
Collapse
|
12
|
Guo X, Dang W, Li N, Wang Y, Sun D, Nian H, Wei R. PPAR-α Agonist Fenofibrate Ameliorates Sjögren Syndrome-Like Dacryoadenitis by Modulating Th1/Th17 and Treg Cell Responses in NOD Mice. Invest Ophthalmol Vis Sci 2022; 63:12. [PMID: 35687344 PMCID: PMC9202336 DOI: 10.1167/iovs.63.6.12] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effects and mechanisms of fenofibrate, a synthetic ligand of peroxisome proliferator-activated receptor α (PPAR-α), on autoimmune dacryoadenitis in a mouse model of Sjögren syndrome (SS) dry eye. Methods Male nonobese diabetic (NOD) mice were fed chow with or without 0.03% fenofibrate for 8 weeks, and clinical scores were determined by assessing tear secretion, fluorescein, and hematoxylin and eosin staining. Intracellular IFN-γ, IL-17, and Foxp3 in CD4+ T cells were measured by flow cytometry. The expressions of Th1, Th17, and Treg cell-related transcription factors and cytokines were detected by real-time PCR. The levels of PPAR-α and liver X receptor β (LXR-β) were detected with real-time PCR and Western blotting. Results Fenofibrate efficiently diminished the lymphocytic inflammation in lacrimal glands (LGs), increased tear secretion, and decreased corneal fluorescein staining in NOD mice. Meanwhile, treatment of fenofibrate evidently reduced the proportion of Th1 and Th17 cells and increased the proportion of Treg cells in vivo and vitro, together with decreased expression of T-bet, IFN-γ, RORγt, and IL-17, as well as increased expression of Foxp3 and TGF-β1 in LGs. Furthermore, fenofibrate significantly upregulated the expressions of PPAR-α and LXR-β at the protein and mRNA levels. Conclusions Fenofibrate potently attenuated LG inflammation in a model of autoimmune dry eye, and this effect might partially result from regulating Th1/Th17/Treg cell responses by activating PPAR-α/LXR-β signaling. These data suggest that fenofibrate may be a novel class of therapeutic agent for SS-associated dacryoadenitis.
Collapse
Affiliation(s)
- Xingyi Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Weiyu Dang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Na Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Ying Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Deming Sun
- Doheny Eye Institute, And Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, United States
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
13
|
Brown ME, Peters LD, Hanbali SR, Arnoletti JM, Sachs LK, Nguyen KQ, Carpenter EB, Seay HR, Fuhrman CA, Posgai AL, Shapiro MR, Brusko TM. Human CD4 +CD25 +CD226 - Tregs Demonstrate Increased Purity, Lineage Stability, and Suppressive Capacity Versus CD4 +CD25 +CD127 lo/- Tregs for Adoptive Cell Therapy. Front Immunol 2022; 13:873560. [PMID: 35693814 PMCID: PMC9178079 DOI: 10.3389/fimmu.2022.873560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/28/2022] [Indexed: 01/21/2023] Open
Abstract
Regulatory T cell (Treg) adoptive cell therapy (ACT) represents an emerging strategy for restoring immune tolerance in autoimmune diseases. Tregs are commonly purified using a CD4+CD25+CD127lo/- gating strategy, which yields a mixed population: 1) cells expressing the transcription factors, FOXP3 and Helios, that canonically define lineage stable thymic Tregs and 2) unstable FOXP3+Helios- Tregs. Our prior work identified the autoimmune disease risk-associated locus and costimulatory molecule, CD226, as being highly expressed not only on effector T cells but also, interferon-γ (IFN-γ) producing peripheral Tregs (pTreg). Thus, we sought to determine whether isolating Tregs with a CD4+CD25+CD226- strategy yields a population with increased purity and suppressive capacity relative to CD4+CD25+CD127lo/- cells. After 14d of culture, expanded CD4+CD25+CD226- cells displayed a decreased proportion of pTregs relative to CD4+CD25+CD127lo/- cells, as measured by FOXP3+Helios- expression and the epigenetic signature at the FOXP3 Treg-specific demethylated region (TSDR). Furthermore, CD226- Tregs exhibited decreased production of the effector cytokines, IFN-γ, TNF, and IL-17A, along with increased expression of the immunoregulatory cytokine, TGF-β1. Lastly, CD226- Tregs demonstrated increased in vitro suppressive capacity as compared to their CD127lo/- counterparts. These data suggest that the exclusion of CD226-expressing cells during Treg sorting yields a population with increased purity, lineage stability, and suppressive capabilities, which may benefit Treg ACT for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Matthew E. Brown
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Leeana D. Peters
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Seif R. Hanbali
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Juan M. Arnoletti
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Lindsey K. Sachs
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Kayla Q. Nguyen
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Emma B. Carpenter
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Howard R. Seay
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- ROSALIND, Inc., San Diego, CA, United States
| | - Christopher A. Fuhrman
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- NanoString Technologies, Inc., Seattle, WA, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Melanie R. Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
14
|
Riemma MA, Cerqua I, Romano B, Irollo E, Bertolino A, Camerlingo R, Granato E, Rea G, Scala S, Terlizzi M, Spaziano G, Sorrentino R, D'Agostino B, Roviezzo F, Cirino G. Sphingosine-1-phosphate/TGF-β axis drives epithelial mesenchymal transition in asthma-like disease. Br J Pharmacol 2022; 179:1753-1768. [PMID: 34825370 PMCID: PMC9306821 DOI: 10.1111/bph.15754] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Airway remodelling is a critical feature of chronic lung diseases. Epithelial-mesenchymal transition (EMT) represents an important source of myofibroblasts, contributing to airway remodelling. Here, we investigated the sphingosine-1-phosphate (S1P) role in EMT and its involvement in asthma-related airway dysfunction. EXPERIMENTAL APPROACH A549 cells were used to assess the S1P effect on EMT and its interaction with TGF-β signalling. To assess the S1P role in vivo and its impact on lung function, two experimental models of asthma were used by exposing BALB/c mice to subcutaneous administration of either S1P or ovalbumin (OVA). KEY RESULTS Following incubation with TGF-β or S1P, A549 acquire a fibroblast-like morphology associated with an increase of mesenchymal markers and down-regulation of the epithelial. These effects are reversed by treatment with the TGF-β receptor antagonist LY2109761. Systemic administration of S1P to BALB/c mice induces asthma-like disease characterized by mucous cell metaplasia and increased levels of TGF-β, IL-33 and FGF-2 within the lung. The bronchi harvested from S1P-treated mice display bronchial hyperresponsiveness associated with overexpression of the mesenchymal and fibrosis markers and reduction of the epithelial.The S1P-induced switch from the epithelial toward the mesenchymal pattern correlates to a significant increase of lung resistance and fibroblast activation. TGF-β blockade, in S1P-treated mice, abrogates these effects. Finally, inhibition of sphingosine kinases by SK1-II in OVA-sensitized mice, abrogates EMT, pulmonary TGF-β up-regulation, fibroblasts recruitment and airway hyperresponsiveness. CONCLUSION AND IMPLICATIONS Targeting S1P/TGF-β axis may hold promise as a feasible therapeutic target to control airway dysfunction in asthma.
Collapse
Affiliation(s)
- Maria A. Riemma
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| | - Barbara Romano
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| | - Elena Irollo
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - Antonio Bertolino
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| | - Rosa Camerlingo
- RCCS INT Cellular Biology and Bioterapy‐ Research DepartmentNational Cancer Institute G. Pascale FoundationNaplesItaly
| | - Elisabetta Granato
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| | - Giuseppina Rea
- IRCCS INT Microenvironment Molecular TargetsNational Cancer Institute G. Pascale FoundationNaplesItaly
| | - Stefania Scala
- IRCCS INT Microenvironment Molecular TargetsNational Cancer Institute G. Pascale FoundationNaplesItaly
| | - Michela Terlizzi
- Department of Pharmacy (DIFARMA)University of SalernoSalernoItaly
| | - Giuseppe Spaziano
- Department of Experimental Medicine L. Donatelli, Section of Pharmacology, School of MedicineUniversity of Campania Luigi VanvitelliNaplesItaly
| | | | - Bruno D'Agostino
- Department of Experimental Medicine L. Donatelli, Section of Pharmacology, School of MedicineUniversity of Campania Luigi VanvitelliNaplesItaly
| | - Fiorentina Roviezzo
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and SurgeryUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
15
|
Sami AS, Rosh JR. Dual Biologic Therapy in an Adolescent With Camurati-Engelmann Disease and Crohn Disease. JPGN REPORTS 2022; 3:e169. [PMID: 37168741 PMCID: PMC10158315 DOI: 10.1097/pg9.0000000000000169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 12/08/2021] [Indexed: 05/13/2023]
Abstract
Camurati-Engelmann disease (CED) is a rare disorder caused by activating mutations in the TGF-β1 gene and characterized by hyperostosis of long bones and bone dysplasia. We describe a case of an adolescent with CED and moderate-severe Crohn Disease (CD). Infliximab improved gastrointestinal symptoms but was associated with worsening CED-associated bone pain. Clinical remission was successfully achieved with dual biologic therapy that included vedolizumab and ustekinumab. Possible reasons for this patient's clinical response are advanced and include speculation about the complex role of TGF-β1 signaling in the etiology of both CED and CD.
Collapse
Affiliation(s)
- Ahmad Salah Sami
- From the Department of Pediatrics, Goryeb Children’s Hospital, Morristown, NJ
| | - Joel R. Rosh
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Goryeb Children’s Hospital, Morristown, NJ
| |
Collapse
|
16
|
He Z, Khatib AM, Creemers JWM. The proprotein convertase furin in cancer: more than an oncogene. Oncogene 2022; 41:1252-1262. [PMID: 34997216 DOI: 10.1038/s41388-021-02175-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/13/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023]
Abstract
Furin is the first discovered proprotein convertase member and is present in almost all mammalian cells. Therefore, by regulating the maturation of a wide range of proproteins, Furin expression and/or activity is involved in various physiological and pathophysiological processes ranging from embryonic development to carcinogenesis. Since many of these protein precursors are involved in initiating and maintaining the hallmarks of cancer, Furin has been proposed as a potential target for treating several human cancers. In contrast, other studies have revealed that some types of cancer do not benefit from Furin inhibition. Therefore, understanding the heterogeneous functions of Furin in cancer will provide important insights into the design of effective strategies targeting Furin in cancer treatment. Here, we present recent advances in understanding how Furin expression and activity are regulated in cancer cells and their influences on the activity of Furin substrates in carcinogenesis. Furthermore, we discuss how Furin represses tumorigenic properties of several cancer cells and why Furin inhibition leads to aggressive phenotypes in other tumors. Finally, we summarize the clinical applications of Furin inhibition in treating human cancers.
Collapse
Affiliation(s)
- Zongsheng He
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Abdel-Majid Khatib
- INSERM, LAMC, UMR 1029, Allée Geoffroy St Hilaire, Pessac, France.
- Institut Bergoinié, Bordeaux, France.
| | - John W M Creemers
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
17
|
Ghosh S, Roy K, Rajalingam R, Martin S, Pal C. Cytokines in the generation and function of regulatory T cell subsets in leishmaniasis. Cytokine 2021; 147:155266. [DOI: 10.1016/j.cyto.2020.155266] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/30/2020] [Accepted: 08/24/2020] [Indexed: 01/12/2023]
|
18
|
Ramchandani R, Hossenbaccus L, Ellis AK. Immunoregulatory T cell epitope peptides for the treatment of allergic disease. Immunotherapy 2021; 13:1283-1291. [PMID: 34558985 DOI: 10.2217/imt-2021-0133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Allergic diseases are type 2 inflammatory reactions with an increasing worldwide prevalence, making the search for new therapeutic options pertinent. Allergen immunotherapy is the only disease-modifying approach for allergic rhinitis, though it can result in systemic reactions. Recently, peptide immunotherapy (PIT), involving T-cell epitope peptides that bind to major histocompatibility complexes, have been developed. It is speculated that they can induce T helper cell type 2 anergy, Treg cell upregulation or immune deviation. Promising results in cat dander, honeybee venom, Japanese cedar pollen, grass pollens, ragweed and house dust mite clinical trials have shown safety, efficacy and tolerability to PIT. Hence, PIT may hold the potential to change the treatment algorithm for allergic rhinitis.
Collapse
Affiliation(s)
- Rashi Ramchandani
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada.,Allergy Research Unit, Kingston Health Sciences Center - KGH Site, Kingston, on, K7L 2V7, Canada
| | - Lubnaa Hossenbaccus
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada.,Allergy Research Unit, Kingston Health Sciences Center - KGH Site, Kingston, on, K7L 2V7, Canada
| | - Anne K Ellis
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada.,Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada.,Allergy Research Unit, Kingston Health Sciences Center - KGH Site, Kingston, on, K7L 2V7, Canada
| |
Collapse
|
19
|
Zhang LJ, Guerrero-Juarez CF, Chen SX, Zhang X, Yin M, Li F, Wu S, Chen J, Li M, Liu Y, Jiang SIB, Hata T, Plikus MV, Gallo RL. Diet-induced obesity promotes infection by impairment of the innate antimicrobial defense function of dermal adipocyte progenitors. Sci Transl Med 2021; 13:13/577/eabb5280. [PMID: 33472955 DOI: 10.1126/scitranslmed.abb5280] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/29/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022]
Abstract
Infections are a major complication of obesity, but the mechanisms responsible for impaired defense against microbes are not well understood. Here, we found that adipocyte progenitors were lost from the dermis during diet-induced obesity (DIO) in humans and mice. The loss of adipogenic fibroblasts from mice resulted in less antimicrobial peptide production and greatly increased susceptibility to Staphylococcus aureus infection. The decrease in adipocyte progenitors in DIO mice was explained by expression of transforming growth factor-β (TGFβ) by mature adipocytes that then inhibited adipocyte progenitors and the production of cathelicidin in vitro. Administration of a TGFβ receptor inhibitor or a peroxisome proliferator-activated receptor-γ agonist reversed this inhibition in both cultured adipocyte progenitors and in mice and subsequently restored the capacity of obese mice to defend against S. aureus skin infection. Together, these results explain how obesity promotes dysfunction of the antimicrobial function of reactive dermal adipogenesis and identifies potential therapeutic targets to manage skin infection associated with obesity.
Collapse
Affiliation(s)
- Ling-Juan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China. .,Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christian F Guerrero-Juarez
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA.,Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA.,Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA.,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Stella X Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiaowei Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Meimei Yin
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shuai Wu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Joyce Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Min Li
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Yingzi Liu
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA.,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Shang I B Jiang
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tissa Hata
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA.,Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA.,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
20
|
Dutta N, Lillehoj PB, Estrela P, Dutta G. Electrochemical Biosensors for Cytokine Profiling: Recent Advancements and Possibilities in the Near Future. BIOSENSORS 2021; 11:94. [PMID: 33806879 PMCID: PMC8004910 DOI: 10.3390/bios11030094] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
Cytokines are soluble proteins secreted by immune cells that act as molecular messengers relaying instructions and mediating various functions performed by the cellular counterparts of the immune system, by means of a synchronized cascade of signaling pathways. Aberrant expression of cytokines can be indicative of anomalous behavior of the immunoregulatory system, as seen in various illnesses and conditions, such as cancer, autoimmunity, neurodegeneration and other physiological disorders. Cancer and autoimmune diseases are particularly adept at developing mechanisms to escape and modulate the immune system checkpoints, reflected by an altered cytokine profile. Cytokine profiling can provide valuable information for diagnosing such diseases and monitoring their progression, as well as assessing the efficacy of immunotherapeutic regiments. Toward this goal, there has been immense interest in the development of ultrasensitive quantitative detection techniques for cytokines, which involves technologies from various scientific disciplines, such as immunology, electrochemistry, photometry, nanotechnology and electronics. This review focusses on one aspect of this collective effort: electrochemical biosensors. Among the various types of biosensors available, electrochemical biosensors are one of the most reliable, user-friendly, easy to manufacture, cost-effective and versatile technologies that can yield results within a short period of time, making it extremely promising for routine clinical testing.
Collapse
Affiliation(s)
- Nirmita Dutta
- School of Medical Science and Technology (SMST), Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| | - Peter B. Lillehoj
- Department of Mechanical Engineering, Rice University, Houston, TX 77005, USA;
| | - Pedro Estrela
- Centre for Biosensors, Bioelectronics and Biodevices (C3Bio) and Department of Electronic & Electrical Engineering, University of Bath, Bath BA2 7AY, UK
| | - Gorachand Dutta
- School of Medical Science and Technology (SMST), Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| |
Collapse
|
21
|
Chen J, Qiao YD, Li X, Xu JL, Ye QJ, Jiang N, Zhang H, Wu XY. Intratumoral CD45 +CD71 + erythroid cells induce immune tolerance and predict tumor recurrence in hepatocellular carcinoma. Cancer Lett 2020; 499:85-98. [PMID: 33279623 DOI: 10.1016/j.canlet.2020.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023]
Abstract
CD45+CD71+ erythroid cells generated through splenic extramedullary erythropoiesis have recently been found to suppress anti-infection and tumor immunity in neonates and adults with malignances. However, their role in tumor microenvironment has not been investigated. In the present study, we found that the number of CD45+CD71+ erythroid cells was significantly elevated in hepatocellular carcinoma (HCC) tissues compared to that in paratumor region and circulation. Additionally, they were more abundant in HCC tissues compared to some immune suppressive cells as well as CD45-CD71+ erythroid cells. CD45+CD71+ erythroid cells suppressed T cells through generation of reactive oxygen species, IL-10, and TGF-β in a paracrine and cell-cell contact manner, and their suppressive effect was stronger than that of myeloid-derived suppressor cells. The abundance of CD45+CD71+ erythroid cells in tumor tissue, as illustrated via immunofluorescence, predicted disease-free survival and overall survival, and its prognostic value was better than that of Cancer of the Liver Italian Program score. This study demonstrated that accumulation of intratumoral CD45+CD71+ erythroid cells in HCC tissues could play a superior immunosuppressive role in tumor microenvironment and may serve as a valuable biomarker to predict recurrence of HCC.
Collapse
Affiliation(s)
- Jie Chen
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Yi-Dan Qiao
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Xing Li
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Jian-Liang Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Qing-Jian Ye
- Department of Gynaecology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Nan Jiang
- Department of Transplantation, The Second Affiliated Hospital of Southern University of Science and Technology and the Third People's Hospital of Shenzhen, 29th Bulan Road, Shenzhen, 510623, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
| | - Xiang-Yuan Wu
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
22
|
Zhao X, Zhao Y, Sun X, Xing Y, Wang X, Yang Q. Immunomodulation of MSCs and MSC-Derived Extracellular Vesicles in Osteoarthritis. Front Bioeng Biotechnol 2020; 8:575057. [PMID: 33251195 PMCID: PMC7673418 DOI: 10.3389/fbioe.2020.575057] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) has become recognized as a low-grade inflammatory state. Inflammatory infiltration of the synovium by macrophages, T cells, B cells, and other immune cells is often observed in OA patients and plays a key role in the pathogenesis of OA. Hence, orchestrating the local inflammatory microenvironment and tissue regeneration microenvironment is important for the treatment of OA. Mesenchymal stem cells (MSCs) offer the potential for cartilage regeneration owing to their effective immunomodulatory properties and anti-inflammatory abilities. The paracrine effect, mediated by MSC-derived extracellular vehicles (EVs), has recently been suggested as a mechanism for their therapeutic properties. In this review, we summarize the interactions between MSCs or MSC-derived EVs and OA-related immune cells and discuss their therapeutic effects in OA. Additionally, we discuss the potential of MSC-derived EVs as a novel cell-free therapy approach for the clinical treatment of OA.
Collapse
Affiliation(s)
- Xige Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
| | - Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yi Xing
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
23
|
Retinoid-Related Orphan Receptor RORγt in CD4 + T-Cell-Mediated Intestinal Homeostasis and Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1984-1999. [PMID: 32735890 DOI: 10.1016/j.ajpath.2020.07.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Retinoic acid-related orphan receptor (ROR)-γt, the master transcription factor of the Th17 subset of CD4+ Th cells, is a promising target for treating a host of autoimmune diseases. RORγt plays a vital role in the pathogenesis of inflammatory bowel diseases-Crohn disease and ulcerative colitis-caused by untoward reactivity of the immune system to the components of the intestinal microbiome. The mammalian intestinal tract is a highly complex and compartmentalized organ with specialized functions, and is a privileged site for the generation of both peripherally induced regulatory CD4+ T cells (Tregs) and effector Th17 cells. As Th17 cells can be proinflammatory in nature, the equilibrium between effector Th17 and Treg cells is crucial for balancing intestinal homeostasis and inflammation. Recent findings suggest that RORγt, in addition to Th17 cells, is also expressed in peripherally induced, colonic regulatory CD4+ T cells. Therefore, RORγt is expressed in both effector and regulatory subsets of CD4+ T cells in the intestine. The present review discusses the role of RORγt in cellular and molecular differentiation of Th17 and Treg, and examines how targeting RORγt in inflammatory bowel disease therapy could influence the development of these two diverse subsets of immune cells with opposing functions.
Collapse
|
24
|
FoxP3 + T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett 2020; 490:174-185. [PMID: 32721551 DOI: 10.1016/j.canlet.2020.07.022] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/28/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022]
Abstract
T Regulatory cells (Tregs) can have both protective and pathological roles. They maintain immune homeostasis and inhibit immune responses in various diseases, including cancer. Proportions of Tregs in the peripheral blood of some cancer patients increase by approximately two-fold, compared to those in healthy individuals. Tregs contribute to cancer development and progression by suppressing T effector cell functions, thereby compromising tumor killing and promoting tumor growth. Highly immunosuppressive Tregs express upregulated levels of the transcription factor, Forkhead box protein P3 (FoxP3). Elevated levels of FoxP3+ Tregs within the tumor microenvironment (TME) showed a positive correlation with poor prognosis in various cancer patients. Despite the success of immunotherapy, including the use of immune checkpoint inhibitors, a significant proportion of patients show low response rates as a result of primary or acquired resistance against therapy. Some of the mechanisms which underlie the development of therapy resistance are associated with Treg suppressive function. In this review, we describe Treg contribution to cancer development/progression, and the mechanisms of Treg-mediated immunosuppression. We discuss the prognostic significance of FoxP3+ Tregs in different cancers and their potential use as prognostic biomarkers. We also describe potential therapeutic strategies to target Tregs in combination with other types of immunotherapies aiming to overcome tumor resistance and improve clinical outcomes in cancer patients. Overall, understanding the prognostic significance of FoxP3+ Tregs in various cancers and their contribution to therapy resistance could help in the development of more effective targeted therapeutic strategies to enhance the clinical outcomes in cancer patients.
Collapse
|
25
|
Darlan DM, Munir D, Putra A, Jusuf NK. MSCs-released TGFβ1 generate CD4 +CD25 +Foxp3 + in T-reg cells of human SLE PBMC. J Formos Med Assoc 2020; 120:602-608. [PMID: 32718891 DOI: 10.1016/j.jfma.2020.06.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/08/2020] [Accepted: 06/28/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND/PURPOSE Regulatory T-cell (Treg) defects may cause autoreactivity of both T and B cells, leading to autoimmune disease including systemic lupus erythematosus (SLE). The immune response defects in SLE are characterized by the decreased expression of CD4, CD25, and Foxp3, known as inducible Treg (iTreg). Therefore, restoring iTreg expression can reverse autoimmunity states into immune tolerances leading to normal immune responses. Mesenchymal stem cells (MSCs) have immunomodulatory properties to control inflammatory milieu, including in SLE inflammation by releasing TGFβ1, IL10, and PGE2, thus MSCs can potentially generate iTreg cells. However, the mechanisms of MSC-released TGFβ1 to promote iTreg generation in human SLE remains unclear. This study aims to analyze the role of MSC-released TGFβ1 in generating CD4+, CD25+, and Foxp3+ expression in iTreg cells from human SLE peripheral blood mononuclear cells (PBMCs). METHODS This study used a post-test control group design. MSCs were obtained from human umbilical cord blood and characterized according to their surface antigen expression and multilineage differentiation capacities. PBMCs isolated from SLE patients were divided into five groups, including sham, control, and three treatment groups. The treatment groups were treated by co-culturing MSCs to PBMCs with ratio of 1:1 (T1), 1:25 (T2), and 1:50 (T3) for 72 h incubation. The expression of CD4, CD25, and Foxp3 in Treg was analyzed by flow cytometry assay while TGFβ1 level was determined by Cytometric Bead Array (CBA). RESULTS This study showed that the percentage of CD4+CD25+Foxp3+ iTreg cells was significantly increased in T1 and T2. This finding was aligned with the significant increase of TGFβ1 level. CONCLUSION MSCs promote iTreg cells generation from human SLE PBMCs by releasing TGFβ1 to control SLE disease.
Collapse
Affiliation(s)
- Dewi Masyithah Darlan
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Delfitri Munir
- Pusat Unggulan IPTEK Tissue Engineering, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research (SCCR), Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia; Department of Postgraduate Biomedical Science, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia; Department of Pathological Anatomy, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia.
| | - Nelva Karmila Jusuf
- Department of Dermatology and Venereology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
26
|
Chen X, Feng L, Li S, Long D, Shan J, Li Y. TGF-β1 maintains Foxp3 expression and inhibits glycolysis in natural regulatory T cells via PP2A-mediated suppression of mTOR signaling. Immunol Lett 2020; 226:31-37. [PMID: 32598969 DOI: 10.1016/j.imlet.2020.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/15/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023]
Abstract
Natural regulatory T cells (nTregs) play a dominant role in maintaining immunological homeostasis and they are known to undergo metabolic reprogramming during immune responses. Transforming growth factor-β1 (TGF-β1), an anti-inflammatory cytokine, can promote the induction of regulatory T cells. Here, we investigated the effects of TGF-β1 on the stability and metabolism of nTregs stimulated in vitro. CD4+CD25+ nTregs were isolated from mouse spleens and stimulated with anti-CD3 and anti-CD28 antibodies plus IL-2 in the presence or absence of TGF-β1. Exposure to TGF-β1 induced the activation of STAT5 and sustained the expression of the nTregs transcription factor Foxp3. In addition, TGF-β1 inhibited glycolysis, as shown by reduced lactate production and diminished expression of Glut1, Hk2, Enolase1, and Hif-1α. nTregs treated with TGF-β1 exhibited downregulated mTORC1 signaling but enhanced activation of the serine-threonine phosphatase PP2A. Moreover, treat with the PP2A inhibitor okadaic acid disrupted the maintenance of Foxp3 expression by TGF-β1. Thus, TGF-β1 serves to maintain Foxp3 expression in cultured nTregs, possibly via PP2A activation and suppression of mTORC1-regulated glycolysis.
Collapse
Affiliation(s)
- Xuelu Chen
- Key Laboratory of Transplant Engineering and Immunology of The Ministry of Health, Regenerative Medicine Research Centre, The Organ Transplantation Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Li Feng
- Key Laboratory of Transplant Engineering and Immunology of The Ministry of Health, Regenerative Medicine Research Centre, The Organ Transplantation Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Shengfu Li
- Key Laboratory of Transplant Engineering and Immunology of The Ministry of Health, Regenerative Medicine Research Centre, The Organ Transplantation Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology of The Ministry of Health, Regenerative Medicine Research Centre, The Organ Transplantation Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Juan Shan
- Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China.
| | - Youping Li
- Key Laboratory of Transplant Engineering and Immunology of The Ministry of Health, Regenerative Medicine Research Centre, The Organ Transplantation Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China; Chinese Cochrane Centre, Chinese Evidence-Based Medicine Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China.
| |
Collapse
|
27
|
Hou A, Hou K, Huang Q, Lei Y, Chen W. Targeting Myeloid-Derived Suppressor Cell, a Promising Strategy to Overcome Resistance to Immune Checkpoint Inhibitors. Front Immunol 2020. [PMID: 32508809 DOI: 10.3389/fimmu.2020.00783.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are starting to transform the treatment for patients with advanced cancer. The extensive application of these antibodies for various cancer obtains exciting anti-tumor immune response by activating T cells. Although the encouraging clinical benefit in patients receiving these immunostimulatory agents are observed, numbers of patients still derive limited response or even none for reasons unknown, sometimes at the cost of adverse reactions. Myeloid-derived suppressor cells (MDSCs) is a heterogeneous immature population of myeloid cells partly influencing the efficacy of immunotherapies. These cells not only directly suppress T cell but mediate a potently immunosuppressive network within tumor microenvironment to attenuate the anti-tumor response. The crosstalk between MDSCs and immune cells/non-immune cells generates several positive feedbacks to negatively modulate the tumor microenvironment. As such, the recruitment of immunosuppressive cells, upregulation of immune checkpoints, angiogenesis and hypoxia are induced and contributing to the acquired resistance to ICIs. Targeting MDSCs could be a potential therapy to overcome the limitation. In this review, we focus on the role of MDSCs in resistance to ICIs and summarize the therapeutic strategies targeting them to enhance ICIs efficiency in cancer patients.
Collapse
Affiliation(s)
- Aohan Hou
- Faculty of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Kaiyu Hou
- Department of Bone and Trauma, The Second People's Hospital of Yunnan Province, Kunming, China
| | - Qiubo Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| | - Wanling Chen
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| |
Collapse
|
28
|
Hou A, Hou K, Huang Q, Lei Y, Chen W. Targeting Myeloid-Derived Suppressor Cell, a Promising Strategy to Overcome Resistance to Immune Checkpoint Inhibitors. Front Immunol 2020; 11:783. [PMID: 32508809 PMCID: PMC7249937 DOI: 10.3389/fimmu.2020.00783] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are starting to transform the treatment for patients with advanced cancer. The extensive application of these antibodies for various cancer obtains exciting anti-tumor immune response by activating T cells. Although the encouraging clinical benefit in patients receiving these immunostimulatory agents are observed, numbers of patients still derive limited response or even none for reasons unknown, sometimes at the cost of adverse reactions. Myeloid-derived suppressor cells (MDSCs) is a heterogeneous immature population of myeloid cells partly influencing the efficacy of immunotherapies. These cells not only directly suppress T cell but mediate a potently immunosuppressive network within tumor microenvironment to attenuate the anti-tumor response. The crosstalk between MDSCs and immune cells/non-immune cells generates several positive feedbacks to negatively modulate the tumor microenvironment. As such, the recruitment of immunosuppressive cells, upregulation of immune checkpoints, angiogenesis and hypoxia are induced and contributing to the acquired resistance to ICIs. Targeting MDSCs could be a potential therapy to overcome the limitation. In this review, we focus on the role of MDSCs in resistance to ICIs and summarize the therapeutic strategies targeting them to enhance ICIs efficiency in cancer patients.
Collapse
Affiliation(s)
- Aohan Hou
- Faculty of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Kaiyu Hou
- Department of Bone and Trauma, The Second People's Hospital of Yunnan Province, Kunming, China
| | - Qiubo Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| | - Wanling Chen
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| |
Collapse
|
29
|
Comprehensive Molecular Profiles of Functionally Effective MSC-Derived Extracellular Vesicles in Immunomodulation. Mol Ther 2020; 28:1628-1644. [PMID: 32380062 DOI: 10.1016/j.ymthe.2020.04.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/10/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence indicates that mesenchymal stem/stromal cell-derived extracellular vesicles (MSC-EVs) exhibit immunomodulatory effects by delivering therapeutic RNAs and proteins; however, the molecular mechanism underlying the EV-mediated immunomodulation is not fully understood. In this study, we found that EVs from early-passage MSCs had better immunomodulatory potency than did EVs from late-passage MSCs in T cell receptor (TCR)- or Toll-like receptor 4 (TLR4)-stimulated splenocytes and in mice with ocular Sjögren's syndrome. Moreover, MSC-EVs were more effective when produced from 3D culture of the cells than from the conventional 2D culture. Comparative molecular profiling using proteomics and microRNA sequencing revealed the enriched factors in MSC-EVs that were functionally effective in immunomodulation. Among them, manipulation of transforming growth factor β1 (TGF-β1), pentraxin 3 (PTX3), let-7b-5p, or miR-21-5p levels in MSCs significantly affected the immunosuppressive effects of their EVs. Furthermore, there was a strong correlation between the expression levels of TGF-β1, PTX3, let-7b-5p, or miR-21-5p in MSC-EVs and their suppressive function. Therefore, our comparative strategy identified TGF-β1, PTX3, let-7b-5p, or miR-21-5p as key molecules mediating the therapeutic effects of MSC-EVs in autoimmune disease. These findings would help understand the molecular mechanism underlying EV-mediated immunomodulation and provide functional biomarkers of EVs for the development of robust EV-based therapies.
Collapse
|
30
|
Ma N, Zhang J, Reiter RJ, Ma X. Melatonin mediates mucosal immune cells, microbial metabolism, and rhythm crosstalk: A therapeutic target to reduce intestinal inflammation. Med Res Rev 2020; 40:606-632. [PMID: 31420885 DOI: 10.1002/med.21628] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
Nowadays, melatonin, previously considered only as a pharmaceutical product for rhythm regulation and sleep aiding, has shown its potential as a co-adjuvant treatment in intestinal diseases, however, its mechanism is still not very clear. A firm connection between melatonin at a physiologically relevant concentration and the gut microbiota and inflammation has recently established. Herein, we summarize their crosstalk and focus on four novelties. First, how melatonin is synthesized and degraded in the gut and exerts potentially diverse phenotypic effects through its diverse metabolites. Second, how melatonin mediates the activation and proliferation of intestinal mucosal immune cells with paracrine and autocrine properties. By modulating T/B cells, mast cells, macrophages and dendritic cells, melatonin immunomodulatory involved in regulating T-cell differentiation, intervening T/B cell interaction and attenuating the production of pro-inflammatory factors, achieving its antioxidant action via specific receptors. Third, how melatonin exerts antimicrobial action and modulates microbial components, such as lipopolysaccharide, amyloid-β peptides via nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) or signal transducers and activators of transcription (STAT1) pathway to modulate intestinal immune function in immune-pineal axis. The last, how melatonin mediates the effect of intestinal bacterial activity signals on the body rhythm system through the NF-κB pathway and influences the mucosal epithelium oscillation via clock gene expression. These processes are achieved at mitochondrial and nuclear levels to control the host immune cell development. Considering unclear mechanisms and undiscovered actions of melatonin in gut-microbiome-immune axis, it's time to reveal them and provide new insight for the outlook of melatonin as a potential therapeutic target in the treatment and management of intestinal diseases.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jie Zhang
- Animal Husbandry and Veterinary Department, Beijing Vocational College of Agriculture, Beijing, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Department of Internal Medicine and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
31
|
Singh R, Alape D, de Lima A, Ascanio J, Majid A, Gangadharan SP. Regulatory T Cells in Respiratory Health and Diseases. Pulm Med 2019; 2019:1907807. [PMID: 31827925 PMCID: PMC6886321 DOI: 10.1155/2019/1907807] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Respiratory diseases compromise the health of millions of people all over the world and are strongly linked to the immune dysfunction. CD4+FOXP3+ T regulatory cells, also known as Tregs, have a central role maintaining tissue homeostasis during immune responses. Their activity and clinical impact have been widely studied in different clinical conditions including autoimmune diseases, inflammatory conditions, and cancer, amongst others. Tregs express transcription factor forkhead box P3 (FOXP3), which allows regulation of the immune response through anti-inflammatory cytokines such as IL-10 or transforming growth factor beta (TGF-β) and direct cell-to-cell interaction. Maintenance of immune tolerance is achieved via modulation of effector CD4+ T helper 1, 2 or 17 (Th1, Th2, Th17) cells by Tregs. This review highlights the recent progress in the understanding of Tregs in different disorders of the respiratory system.
Collapse
Affiliation(s)
- Rani Singh
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel Alape
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrés de Lima
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Juan Ascanio
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Adnan Majid
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sidhu P. Gangadharan
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Regulation of the complement system and immunological tolerance in pregnancy. Semin Immunol 2019; 45:101337. [PMID: 31757607 DOI: 10.1016/j.smim.2019.101337] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/24/2019] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a serious vascular complication of the human pregnancy, whose etiology is still poorly understood. In preeclampsia, exacerbated apoptosis and fragmentation of the placental tissue occurs due to developmental qualities of the placental trophoblast cells and/or mechanical and oxidative distress to the syncytiotrophoblast, which lines the placental villi. Dysregulation of the complement system is recognized as one of the mechanisms of the disease pathology. Complement has the ability to promote inflammation and facilitate phagocytosis of placenta-derived particles and apoptotic cells by macrophages. In preeclampsia, an overload of placental cell damage or dysregulated complement system may lead to insufficient clearance of apoptotic particles and placenta-derived debris. Excess placental damage may lead to sequestration of microparticles, such as placental vesicles, to capillaries in the glomeruli of the kidney and other vulnerable tissues. This phenomenon could contribute to the manifestations of typical diagnostic symptoms of preeclampsia: proteinuria and new-onset hypertension. In this review we propose that the complement system may serve as a regulator of the complex tolerance and clearance processes that are fundamental in healthy pregnancy. It is therefore recommended that further research be conducted to elucidate the interactions between components of the complement system and immune responses in the context of complicated and healthy pregnancy.
Collapse
|
33
|
Saleh R, Elkord E. Treg-mediated acquired resistance to immune checkpoint inhibitors. Cancer Lett 2019; 457:168-179. [PMID: 31078738 DOI: 10.1016/j.canlet.2019.05.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 02/08/2023]
Abstract
T Regulatory cells (Tregs) act as a double-edged sword by regulating immune homeostasis (protective role) and inhibiting immune responses in different disease settings (pathological role). They contribute to cancer development and progression by suppressing T effector cell (Teff) functions. Decreased ratios of intratumoral CD8+ T cells to Tregs have been associated with poor prognosis in most cancer types. Targeting immune checkpoints (ICs), such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death-1 (PD-1), by immune checkpoint inhibitors (ICIs) in cancer patients has been beneficial in inducing anti-tumor immune responses and improving clinical outcomes. However, response rates remain relatively low, ranging from 15 to 40% depending on cancer type. Additionally, a significant proportion of patients who initially demonstrates a clinical response can acquire resistance overtime. This acquired resistance could occur due to the emergence of compensatory mechanisms within the tumor microenvironment (TME) to evade the anti-tumor effects of ICIs. In this review, we describe the immunosuppressive role of Tregs in the TME, the effects of currently approved ICIs on Treg phenotype and function, and the mechanisms of acquired resistance to ICIs mediated by Tregs within the TME, such as the over-expression of ICs, the up-regulation of immunosuppressive molecules, and apoptotic Treg-induced immunosuppression. We also describe potential therapeutic strategies to target Tregs in combination with ICIs aiming to overcome such resistance and improve clinical outcomes. Elucidating the Treg-mediated acquired resistance mechanisms should benefit the designing of well-targeted therapeutic strategies to overcome resistance and maximize the therapeutic efficacy in cancer patients.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| |
Collapse
|
34
|
Non-HLA Genetic Factors and Their Influence on Heart Transplant Outcomes: A Systematic Review. Transplant Direct 2019; 5:e422. [PMID: 30882026 PMCID: PMC6415970 DOI: 10.1097/txd.0000000000000859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/25/2018] [Indexed: 12/15/2022] Open
Abstract
Supplemental digital content is available in the text. Background Improvement of immunosuppressive therapies and surgical techniques has increased the survival rate after heart transplantation. Nevertheless, a large number of patients still experience complications, such as allograft rejection, vasculopathy, kidney dysfunction, and diabetes in response to immunosuppressive therapy. Variants in HLA genes have been extensively studied for their role in clinical outcomes after transplantation, whereas the knowledge about non-HLA genetic variants in this setting is still limited. Non-HLA polymorphisms are involved in the metabolism of major immunosuppressive therapeutics and may play a role in clinical outcomes after cardiac transplantation. This systematic review summarizes the existing knowledge of associations between non-HLA genetic variation and heart transplant outcomes. Methods The current evidence available on genetic polymorphisms associated with outcomes after heart transplantation was identified by a systematic search in PubMed and Embase. Studies reporting on polymorphisms significantly associated with clinical outcomes after cardiac transplantation were included. Results A total of 56 studies were included, all were candidate gene studies. These studies identified 58 polymorphisms in 36 genes that were associated with outcomes after cardiac transplantation. Variants in TGFB1, CYP3A5, and ABCB1 are consistently replicated across multiple studies for various transplant outcomes. Conclusions The research currently available supports the hypothesis that non-HLA polymorphisms are associated with clinical outcomes after heart transplantation. However, many genetic variants were only identified in a single study, questioning their true effect on the clinical outcomes tested. Further research in larger cohorts with well-defined phenotypes is warranted.
Collapse
|
35
|
Zhang LJ, Chen SX, Guerrero-Juarez CF, Li F, Tong Y, Liang Y, Liggins M, Chen X, Chen H, Li M, Hata T, Zheng Y, Plikus MV, Gallo RL. Age-Related Loss of Innate Immune Antimicrobial Function of Dermal Fat Is Mediated by Transforming Growth Factor Beta. Immunity 2018; 50:121-136.e5. [PMID: 30594464 DOI: 10.1016/j.immuni.2018.11.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/20/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022]
Abstract
Dermal fibroblasts (dFBs) resist infection by locally differentiating into adipocytes and producing cathelicidin antimicrobial peptide in response to Staphylococcus aureus (S. aureus). Here, we show that neonatal skin was enriched with adipogenic dFBs and immature dermal fat that highly expressed cathelicidin. The pool of adipogenic and antimicrobial dFBs declined after birth, leading to an age-dependent loss of dermal fat and a decrease in adipogenesis and cathelidicin production in response to infection. Transforming growth factor beta (TGF-β), which acted on uncommitted embryonic and adult dFBs and inhibited their adipogenic and antimicrobial function, was identified as a key upstream regulator of this process. Furthermore, inhibition of the TGF-β receptor restored the adipogenic and antimicrobial function of dFBs in culture and increased resistance of adult mice to S. aureus infection. These results provide insight into changes that occur in the skin innate immune system between the perinatal and adult periods of life.
Collapse
Affiliation(s)
- Ling-Juan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China; Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Stella Xiang Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yun Tong
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yuqiong Liang
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Marc Liggins
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xu Chen
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Hao Chen
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Min Li
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Tissa Hata
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ye Zheng
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
36
|
Chesneau M, Danger R, Soulillou JP, Brouard S. B cells in operational tolerance. Hum Immunol 2018; 79:373-379. [PMID: 29458071 DOI: 10.1016/j.humimm.2018.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 02/02/2018] [Accepted: 02/12/2018] [Indexed: 12/17/2022]
Abstract
Transplantation is currently the therapy of choice for endstage organ failure even though it requires long-term immunosuppresive therapy, with its numerous side effects, for acceptance of the transplanted organ. In rare cases however, patients develop operational tolerance, that is, graft survival without immunosuppression. Studies conducted on these patients reveal genetic, phenotypic, and functional signatures. They provide a better understanding of the immunological mechanisms involved in operational tolerance and define biomarkers that could be used to adapt immunosuppressive treatment to the individual, safely reduce immunosuppression doses, and ideally and safely guide immunosuppression withdrawal. This review summarizes studies that suggest a role for B cells as biomarkers of operational tolerance and discusses the use of B cells as a predictive tool for immunologic risk.
Collapse
Affiliation(s)
- M Chesneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - R Danger
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - J-P Soulillou
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France
| | - S Brouard
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France; Centre d'Investigation Clinique (CIC) Biothérapie, CHU Nantes, Nantes, France.
| |
Collapse
|
37
|
Liang M, Liwen Z, Yun Z, Yanbo D, Jianping C. The Imbalance between Foxp3 +Tregs and Th1/Th17/Th22 Cells in Patients with Newly Diagnosed Autoimmune Hepatitis. J Immunol Res 2018; 2018:3753081. [PMID: 30050955 PMCID: PMC6040251 DOI: 10.1155/2018/3753081] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/20/2018] [Indexed: 12/13/2022] Open
Abstract
This study is aimed at examining the potential role of regulatory T- (Treg-) Th1-Th17-Th22 cells in the pathogenic process of autoimmune hepatitis (AIH). The numbers of Foxp3+Tregs and Th1, Th17, and Th22 cells were measured in 32 AIH patients using flow cytometry. Moreover, a murine model of experimental autoimmune hepatitis (EAH) was also established and used to investigate the function of Treg-Th1-Th17-Th22 cells in disease progression. AIH patients undergoing an active state had significantly decreased numbers of CD3+CD4+CD25+Foxp3+Tregs and increased numbers of CD3+CD4+CD25-Foxp3+T, CD3+CD4+IFN-γ+Th1, CD3+CD4+IL-17+Th17, and CD3+CD4+IL-2+Th22 cells as well as higher levels of Th1/Th17/Th22-type cytokines compared to AIH patients in remission and HC. Additionally, the numbers of CD3+CD4+CD25+Foxp3+Tregs were negatively correlated with the numbers of Th1-Th17-Th22 cells. Also, the serum levels of IL-17A and IL-22 were correlated positively with liver injury (ALT/AST), whereas the serum levels of IL-10 were correlated negatively with hypergammaglobulinaemia (IgG, IgM) in AIH patients. Interestingly, the percentages of spleen Tregs, expression of Foxp3 mRNA, and liver IL-10 levels decreased, whereas the percentages of spleen Th1-Th17-Th22 cells, expression of T-bet/AHR/RORγt mRNA, and liver IFN-γ, IL-17, and IL-22 levels increased in the murine model of EAH. Our findings demonstrated that an imbalance between Tregs and Th1-Th17-Th22 cells might contribute to the pathogenic process of AIH.
Collapse
Affiliation(s)
- Ma Liang
- Department of Digestive Disease, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Zhang Liwen
- Department of Pediatrics, The Second People's Hospital of Changzhou, Affiliate Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Zhuang Yun
- Department of Digestive Disease, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ding Yanbo
- Department of Digestive Disease, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Chen Jianping
- Department of Digestive Disease, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
38
|
G allele at -924 A > G position of FoxP3 gene promoter as a risk factor for tuberculosis. BMC Infect Dis 2017; 17:673. [PMID: 29020928 PMCID: PMC5637085 DOI: 10.1186/s12879-017-2762-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 09/22/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Forkhead box protein 3 (FoxP3) is an important factor for development and function of Regulatory T cells (Treg). Studies have found an association between common gene polymorphisms in FoxP3 and some infectious diseases. The aim of this study was to evaluate possible associations between two Single nucleotide polymorphisms (SNPs) in the promoter of the FoxP3 gene to susceptibility to tuberculosis (TB) and the alteration of Foxp3 gene expression. METHODS The pattern distribution of genotype at two position, -3279 A > C (rs3761548) and -924 A > G (rs2232365) on the promoter of FoxP3 gene was evaluated using polymerase chain reaction-single specific primer (PCR-SSP) method in 183 tuberculosis patients and 183 healthy control. In addition the quantity of FoxP3 gene expression at mRNA level was identified by the real-time PCR. RESULTS The frequency of G allele at -924 A > G was significantly higher was higher in TB patients (59.5%) than control group (39.5%) (P ≤ 0.05). In addition, our data viewed approximately 5- folds more FoxP3 gene expression in female patients with GG genotype in comparison to female healthy cases with the same genotype (P ≤ 0.001). There was no statistically significant differences between the distribution pattern of -3279 A > C polymorphism in patients and healthy individuals along with it effect on the FoxP3 gene expression among both groups (P > 0.05). CONCLUSIONS Our outcome suggests that the -924 A > G polymorphism leads to enhance FoxP3 gene expression and susceptibility to tuberculosis in the sex dependent manner. This event may rise the count of Treg cells and modulate the immune response against tuberculosis.
Collapse
|
39
|
Functional Changes in the Gut Microbiome Contribute to Transforming Growth Factor β-Deficient Colon Cancer. mSystems 2017; 2:mSystems00065-17. [PMID: 28951889 DOI: 10.1128/msystems.00065-17] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most treatable cancers, with a 5-year survival rate of ~64%, yet over 50,000 deaths occur yearly in the United States. In 15% of cases, deficiency in mismatch repair leads to null mutations in transforming growth factor β (TGF-β) type II receptor, yet genotype alone is not responsible for tumorigenesis. Previous work in mice shows that disruptions in TGF-β signaling combined with Helicobacter hepaticus cause tumorigenesis, indicating a synergistic effect between genotype and microbial environment. Here, we examine functional shifts in the gut microbiome in CRC using integrated -omics approaches to untangle the role of host genotype, inflammation, and microbial ecology. We profile the gut microbiome of 40 mice with/without deficiency in TGF-β signaling from a Smad3 (mothers against decapentaplegic homolog-3) knockout and with/without inoculation with H. hepaticus. Clear functional differences in the microbiome tied to specific bacterial species emerge from four pathways related to human colon cancer: lipopolysaccharide (LPS) production, polyamine synthesis, butyrate metabolism, and oxidative phosphorylation (OXPHOS). Specifically, an increase in Mucispirillum schaedleri drives LPS production, which is associated with an inflammatory response. We observe a commensurate decrease in butyrate production from Lachnospiraceae bacterium A4, which could promote tumor formation. H. hepaticus causes an increase in OXPHOS that may increase DNA-damaging free radicals. Finally, multiple bacterial species increase polyamines that are associated with colon cancer, implicating not just diet but also the microbiome in polyamine levels. These insights into cross talk between the microbiome, host genotype, and inflammation could promote the development of diagnostics and therapies for CRC. IMPORTANCE Most research on the gut microbiome in colon cancer focuses on taxonomic changes at the genus level using 16S rRNA gene sequencing. Here, we develop a new methodology to integrate DNA and RNA data sets to examine functional shifts at the species level that are important to tumor development. We uncover several metabolic pathways in the microbiome that, when perturbed by host genetics and H. hepaticus inoculation, contribute to colon cancer. The work presented here lays a foundation for improved bioinformatics methodologies to closely examine the cross talk between specific organisms and the host, important for the development of diagnostics and pre/probiotic treatment.
Collapse
|
40
|
Zhang L, Zhou Y, Wu Q, Fan W, Ye J, Chen Y, Wu Y, Niu J, Gu Y. Effective prediction of preeclampsia by measuring serum angiotensin II, urinary angiotensinogen and urinary transforming growth factor β1. Exp Ther Med 2017; 14:391-397. [PMID: 28672944 PMCID: PMC5488619 DOI: 10.3892/etm.2017.4484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 01/26/2017] [Indexed: 12/19/2022] Open
Abstract
The aim of the current study was to analyze serum angiotensin II (Ang II), urinary angiotensinogen (AGT) and urinary transforming growth factor β1 (TGFβ1) levels in relation to the clinical manifestation of preeclampsia, and to explore the effects of circulating and renal renin angiotensin system (RAS) in preeclampsia patients. An enzyme-linked immunosorbent assay was used to evaluate serum Ang II, urinary AGT and urinary TGFβ1 in preeclampsia, pregnancy-induced hypertension and normotensive pregnancy patients. The correlation between urinary AGT and serum Ang II, urinary TGFβ1, blood pressure and urinary albumin/creatinine ratio (ACR) were then analyzed. Receiver operating characteristic (ROC) curves were also constructed. Negative correlations were observed between urinary AGT and blood pressure, and urinary AGT and ACR, whereas positive correlations were found between urinary AGT and serum Ang II, and urinary AFT and TGFβ1. Moreover, the area under the curve (AUC) of AGT was 0.841 [95% confidence interval (CI): 0.742–0.940, P<0.001], which was significantly higher than that of serum Ang II or urinary TGFβ1 (P<0.001). The optimal cut-off value of urinary AGT at 193 ng/l showed a high diagnostic value in preeclampsia. The AUC of combined serum Ang II, urinary AGT and urinary TGFβ1 was 0.918 (95% CI: 0.845–0.990, P<0.001), with a sensitivity of 83.9% and a specificity of 89.7%. Decreased levels of urinary AGT in preeclampsia patients suggested that local renal RAS was suppressed, and this was associated with hypertension and proteinuria. A high value preeclampsia diagnosis could be achieved by measuring urinary AGT or a combination of urinary AGT, serum Ang II and urinary TGFβ1.
Collapse
Affiliation(s)
- Lihong Zhang
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yunjiao Zhou
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Qing Wu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Weifeng Fan
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Jun Ye
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yaping Chen
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yun Wu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Jianying Niu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yong Gu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
41
|
Ren W, Liu G, Chen S, Yin J, Wang J, Tan B, Wu G, Bazer FW, Peng Y, Li T, Reiter RJ, Yin Y. Melatonin signaling in T cells: Functions and applications. J Pineal Res 2017; 62. [PMID: 28152213 DOI: 10.1111/jpi.12394] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 01/27/2017] [Indexed: 12/21/2022]
Abstract
Melatonin affects a variety of physiological processes including circadian rhythms, cellular redox status, and immune function. Importantly, melatonin significantly influences T-cell-mediated immune responses, which are crucial to protect mammals against cancers and infections, but are associated with pathogenesis of many autoimmune diseases. This review focuses on our current understanding of the significance of melatonin in T-cell biology and the beneficial effects of melatonin in T-cell response-based diseases. In addition to expressing both membrane and nuclear receptors for melatonin, T cells have the four enzymes required for the synthesis of melatonin and produce high levels of melatonin. Meanwhile, melatonin is highly effective in modulating T-cell activation and differentiation, especially for Th17 and Treg cells, and also memory T cells. Mechanistically, the influence of melatonin in T-cell biology is associated with membrane and nuclear receptors as well as receptor-independent pathways, for example, via calcineurin. Several cell signaling pathways, including ERK1/2-C/EBPα, are involved in the regulatory roles of melatonin in T-cell biology. Through modulation in T-cell responses, melatonin exerts beneficial effects in various inflammatory diseases, such as type 1 diabetes, systemic lupus erythematosus, and multiple sclerosis. These findings highlight the importance of melatonin signaling in T-cell fate determination, and T cell-based immune pathologies.
Collapse
Affiliation(s)
- Wenkai Ren
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Gang Liu
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
| | - Shuai Chen
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
| | - Jie Yin
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
| | - Jing Wang
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
| | - Bie Tan
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Tiejun Li
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha, Hunan, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Yulong Yin
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
- School of Life Science, Hunan Normal University, Changsha, China
| |
Collapse
|
42
|
Li YS, Luo W, Zhu SA, Lei GH. T Cells in Osteoarthritis: Alterations and Beyond. Front Immunol 2017; 8:356. [PMID: 28424692 PMCID: PMC5371609 DOI: 10.3389/fimmu.2017.00356] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/13/2017] [Indexed: 12/16/2022] Open
Abstract
Although osteoarthritis (OA) has been traditionally regarded as a non-inflammatory disease, reports increasingly suggest that it is inflammatory, at least in certain patients. OA patients often exhibit inflammatory infiltration of synovial membranes by macrophages, T cells, mast cells, B cells, plasma cells, natural killer cells, dendritic cells, granulocytes, etc. Although previous reviews have summarized the knowledge of inflammation in the pathogenesis of OA, as far as we know, no report review our current understanding about T cells, especially, each T cell subtype, in the biology of OA. This review highlights the current understanding of the role of T cells in the pathogenesis of OA, with attention to Th1 cells, Th2 cells, Th9 cells, Th17 cells, Th22 cells, regulatory T cells, follicular helper T cells, cytotoxic T cells, T memory cells, and even unconventional T cells (e.g., γδ T cells and cluster of differentiation 1 restricted T cells). The findings highlight the importance of T cells to the development and progression of OA and suggest new therapeutic approaches for OA patients based on the manipulation of T-cell responses.
Collapse
Affiliation(s)
- Yu-Sheng Li
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China.,Department of Orthopaedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China
| | - Shou-An Zhu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Guang-Hua Lei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
43
|
Ren W, Liu G, Yin J, Tan B, Wu G, Bazer FW, Peng Y, Yin Y. Amino-acid transporters in T-cell activation and differentiation. Cell Death Dis 2017; 8:e2655. [PMID: 28252650 PMCID: PMC5386510 DOI: 10.1038/cddis.2016.222] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/25/2022]
Abstract
T-cell-mediated immune responses aim to protect mammals against cancers and infections, and are also involved in the pathogenesis of various inflammatory or autoimmune diseases. Cellular uptake and the utilization of nutrients is closely related to the T-cell fate decision and function. Research in this area has yielded surprising findings in the importance of amino-acid transporters for T-cell development, homeostasis, activation, differentiation and memory. In this review, we present current information on amino-acid transporters, such as LAT1 (l-leucine transporter), ASCT2 (l-glutamine transporter) and GAT-1 (γ-aminobutyric acid transporter-1), which are critically important for mediating peripheral naive T-cell homeostasis, activation and differentiation, especially for Th1 and Th17 cells, and even memory T cells. Mechanically, the influence of amino-acid transporters on T-cell fate decision may largely depend on the mechanistic target of rapamycin complex 1 (mTORC1) signaling. These discoveries remarkably demonstrate the role of amino-acid transporters in T-cell fate determination, and strongly indicate that manipulation of the amino-acid transporter-mTORC1 axis could ameliorate many inflammatory or autoimmune diseases associated with T-cell-based immune responses.
Collapse
Affiliation(s)
- Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.,University of the Chinese Academy of Sciences, Beijing 10008, China
| | - Gang Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| | - Jie Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843-2471, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843-2471, USA
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage and Herbivore, College of Animal Science and Technology, Southwest University, Chongqing 400716, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| |
Collapse
|
44
|
Transforming growth factor-β1 functional polymorphisms in myeloablative sibling hematopoietic stem cell transplantation. Bone Marrow Transplant 2017; 52:739-744. [PMID: 28134923 DOI: 10.1038/bmt.2016.355] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/13/2016] [Accepted: 11/25/2016] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) with sibling donors (s.d.) is a life-saving intervention for patients with hematological malignancies. Numerous genetic factors have a role in transplant outcome. Several functional polymorphisms have been identified in TGF-β1 gene, such as single-nucleotide polymorphism (SNP) at +29C>T within exon 1. Two hundred and forty five patient/donor pairs who underwent a s.d. HSCT in our centers were genotyped for this SNP. In the myeloablative cohort, +29CC donors were associated with an increase in severe chronic GvHD (32% vs 16%, hazard ratio (HR) 9.0, P=0.02). Regarding survival outcomes, +29CC patients developed higher non relapse mortality (NRM) (1-5 years CC 28-32% vs TC/TT 7-10%; HR 5.1, P=0.01). Recipients of +29TT donors experienced a higher relapse rate (1-5 years TT 37-51% vs TC 19-25% vs CC 13%-19%; HR 2.4, P=0.01) with a decreased overall survival (OS) (1-5 years TT 69-50% vs TC/CC 77-69%; HR 1.9, P=0.05). Similar to previous myeloablative unrelated donors HSCT results, we confirmed that +29CC patients had higher NRM. In addition we found that +29TT donors might be associated with a higher relapse rate and lower OS. These results should be confirmed in larger series. Identification of these SNPs will allow personalizing transplant conditioning and immunosuppressant regimens, as well as assisting in the choice of the most appropriate donor.
Collapse
|
45
|
Impact of T-cell-specific Smad4 deficiency on the development of autoimmune diabetes in NOD mice. Immunol Cell Biol 2016; 95:287-296. [PMID: 27686408 PMCID: PMC5364321 DOI: 10.1038/icb.2016.98] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 09/12/2016] [Accepted: 09/22/2016] [Indexed: 12/27/2022]
Abstract
Type 1 diabetes results from autoimmune-mediated pancreatic beta-cell destruction and transforming growth factor-beta (TGF-β) is known to play a preventive role in type 1 diabetes in non-obese diabetic (NOD) mice. In this study, we investigated the role of Smad4, a key molecule for Smad-dependent TGF-β signaling, in T cells of NOD mice in the pathogenesis of autoimmune diabetes. We generated T-cell-specific Smad4 knockout (Smad4 tKO) NOD mice and assessed the pathological and immunological changes. Smad4 tKO showed earlier onset and increased incidence of diabetes than wild type (WT) NOD mice. Pathological features such as insulitis, anti-glutamic acid decarboxylase auto-antibody levels and serum IFN-γ levels were significantly increased in Smad4 tKO compared with WT NOD mice. Proportion and number of activated/memory CD4+ T cell were significantly increased in pancreatic lymph nodes of Smad4 tKO compared with WT NOD mice. However, the proportion and function of regulatory T cells was not different. Effector CD4+ T cells from Smad4 tKO were more resistant to suppression by regulatory T cells than effector cells from WT NOD mice. The proliferative potential of effector T cells from Smad4 tKO was significantly elevated compared with WT NOD mice, and activation of sterol regulatory element binding protein-1c (SREBP-1c) in T cells of Smad4 tKO NOD mice was correlated with this proliferative activity. We conclude that Smad4 deletion in T cells of NOD mice accelerated the development of autoimmune diabetes and increased the incidence of the disease by dysregulation of T cell activation at least in part via SREBP-1c activation.
Collapse
|
46
|
Wang T, Shi W, Fan T, Wan X, Chen YH, Ruan Q. c-Rel is Required for the Induction of pTregs in the Eye but Not in the Gut Mucosa. Immunol Invest 2016; 45:776-786. [PMID: 27224262 DOI: 10.3109/08820139.2016.1172639] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Regulatory T (Treg) cells play an integral role in maintaining immune homeostasis and preventing autoimmune diseases. Forkhead box P3 expression marks the commitment of progenitor cells to the Treg lineage. Although the essential function of the nuclear factor (NF)-κB family transcription factor c-Rel in the regulation of natural Treg cells has been firmly established, little is known about whether c-Rel is involved in the in vivo generation of peripheral Treg cells (pTregs), which develop from mature CD4+ conventional T cells outside of the thymus. We sought to answer this question through the induction of pTregs in the eye and gut mucosa using ovalbumin-specific T cell receptor transgenic mice that do or do not express c-Rel. Our results showed that Tregs can be induced in the eye in a c-Rel-dependent manner when immune-mediated inflammation occurs. However, c-Rel is dispensable for the induction of pTregs in the gut mucosa after oral antigen administration. Thus, c-Rel may play distinct roles in regulating the development of pTregs in different organs. Abbreviations ACAID: Anterior Chamber-Associated Immune Deviation; ATF: activating transcription factor; CREB: cAMP responsive element-binding protein; DMEM: Dulbecco minimum essential medium; HBSS: Hanks Balanced Salt Solution; NFAT: Nuclear Factor of Activated T cells; PBS: Phosphate-buffered saline; PE: Phycoerythrin; WT: wild type.
Collapse
Affiliation(s)
- Ting Wang
- a Shandong Eye Institute , Qingdao , People's Republic of China
| | - Weiyun Shi
- a Shandong Eye Institute , Qingdao , People's Republic of China
| | - Tingting Fan
- b Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen , People's Republic of China
| | - Xiaochun Wan
- b Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen , People's Republic of China
| | - Youhai H Chen
- c Department of Pathology and Laboratory of Medicine , University of Pennsylvania School of Medicine , Philadelphia , PA , USA
| | - Qingguo Ruan
- b Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen , People's Republic of China
| |
Collapse
|
47
|
Weinhold M, Shimabukuro-Vornhagen A, Franke A, Theurich S, Wahl P, Hallek M, Schmidt A, Schinköthe T, Mester J, von Bergwelt-Baildon M, Bloch W. Physical exercise modulates the homeostasis of human regulatory T cells. J Allergy Clin Immunol 2016; 137:1607-1610.e8. [DOI: 10.1016/j.jaci.2015.10.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 01/01/2023]
|
48
|
Hu Y, Cui Q, Ye Y, Luo Y, Tan Y, Shi J, Huang H. Reduction of Foxp3+ T cell subsets involved in incidence of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Hematol Oncol 2015; 35:118-124. [PMID: 26439896 DOI: 10.1002/hon.2255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
| | - Qu Cui
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
- Department of Hematology, Beijing Tiantan Hospital; Capital Medical University; Dongcheng District Beijing China
| | - Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
| | - Yi Luo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
| | - Yamin Tan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
| | - Jimin Shi
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine; Zhejiang University; Hangzhou China
| |
Collapse
|
49
|
Rathod SB, Tripathy AS. TGF-β1 gene - 509C > T promoter polymorphism modulates TGF-β1 levels in hepatitis E patients. Meta Gene 2015; 6:53-8. [PMID: 26504745 PMCID: PMC4576361 DOI: 10.1016/j.mgene.2015.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 02/07/2023] Open
Abstract
Elevated levels of transforming growth factor-β1 (TGF-β1) and its positive correlation with Foxp3 expression in hepatitis E patients have indicated involvement of TGF-β1 in hepatitis E pathogenesis. The current study determined polymorphisms in TGF-β1 gene, plasma TGF-β1 levels and T effector (Teff) cell proliferation and explored their association in a case control study. Polymorphisms in three selected sites (- 509C > T, + 869T > C and + 915G > C) of TGF-β1 gene by PCR & restriction fragment length polymorphism methods, plasma TGF-β1 quantitation by ELISA and Teff (CD4 + CD25 -) cell proliferation by CFSE method were carried out in 277 hepatitis E patients (HE) with self-limiting infection and 233 ethnically matched healthy controls (HCs) from western India. Frequency of CT genotype of - 509C > T site was significantly higher in hepatitis E patients compared to healthy controls (p = 0.017; OR 1.53, 95% CI 1.07-2.17). Plasma TGF-β1 levels were significantly higher in HE compared to HCs. TGF-β1 level of patient group having CT genotype of - 509C > T site was significantly higher compared to those having CC or TT genotypes. Teff cell proliferation was negatively correlated with plasma TGF-β1 levels in HE patients (r = - 0.568; p = 0.014). Influence of TGF-β1 promoter (- 509C > T) polymorphism on plasma TGF-β1 levels and inverse correlation of Teff cell proliferation with plasma TGF-β1 levels in self-limiting hepatitis E patients suggest key role of TGF-β1 in augmentation of reported T regulatory cell mediated pathogenesis in hepatitis E.
Collapse
Key Words
- ALT, alanine transaminase
- CI, confidence interval
- HBV, hepatitis B virus
- HCV, hepatitis C virus
- HCs, healthy controls
- HE, hepatitis E patients
- HEV, hepatitis E virus
- Hepatitis E
- OR, odds ratio
- PBMCs, peripheral blood mononuclear cells
- RFLP, restriction fragment length polymorphism
- Restriction fragment length polymorphism
- Single nucleotide polymorphism
- T effector cell proliferation
- TGF-β1, transforming growth factor beta 1
- Teff, T effector cells
- Transforming growth factor beta 1
- Treg, T regulatory cells
Collapse
Affiliation(s)
- Sanjay B Rathod
- Hepatitis Group, National Institute of Virology, Pune, India
| | | |
Collapse
|
50
|
Chikina MD, Gerald CP, Li X, Ge Y, Pincas H, Nair VD, Wong AK, Krishnan A, Troyanskaya OG, Raymond D, Saunders-Pullman R, Bressman SB, Yue Z, Sealfon SC. Low-variance RNAs identify Parkinson's disease molecular signature in blood. Mov Disord 2015; 30:813-21. [PMID: 25786808 DOI: 10.1002/mds.26205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 01/23/2015] [Accepted: 02/09/2015] [Indexed: 12/20/2022] Open
Abstract
The diagnosis of Parkinson's disease (PD) is usually not established until advanced neurodegeneration leads to clinically detectable symptoms. Previous blood PD transcriptome studies show low concordance, possibly resulting from the use of microarray technology, which has high measurement variation. The Leucine-rich repeat kinase 2 (LRRK2) G2019S mutation predisposes to PD. Using preclinical and clinical studies, we sought to develop a novel statistically motivated transcriptomic-based approach to identify a molecular signature in the blood of Ashkenazi Jewish PD patients, including LRRK2 mutation carriers. Using a digital gene expression platform to quantify 175 messenger RNA (mRNA) markers with low coefficients of variation (CV), we first compared whole-blood transcript levels in mouse models (1) overexpressing wild-type (WT) LRRK2, (2) overexpressing G2019S LRRK2, (3) lacking LRRK2 (knockout), and (4) and in WT controls. We then studied an Ashkenazi Jewish cohort of 34 symptomatic PD patients (both WT LRRK2 and G2019S LRRK2) and 32 asymptomatic controls. The expression profiles distinguished the four mouse groups with different genetic background. In patients, we detected significant differences in blood transcript levels both between individuals differing in LRRK2 genotype and between PD patients and controls. Discriminatory PD markers included genes associated with innate and adaptive immunity and inflammatory disease. Notably, gene expression patterns in levodopa-treated PD patients were significantly closer to those of healthy controls in a dose-dependent manner. We identify whole-blood mRNA signatures correlating with LRRK2 genotype and with PD disease state. This approach may provide insight into pathogenesis and a route to early disease detection.
Collapse
Affiliation(s)
- Maria D Chikina
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christophe P Gerald
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Xianting Li
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yongchao Ge
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hanna Pincas
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Venugopalan D Nair
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aaron K Wong
- Department of Computer Science, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Arjun Krishnan
- Department of Computer Science, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Olga G Troyanskaya
- Department of Computer Science, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Deborah Raymond
- Department of Neurology, Mount Sinai Beth Israel, New York, New York, USA
| | | | - Susan B Bressman
- Department of Neurology, Mount Sinai Beth Israel, New York, New York, USA
| | - Zhenyu Yue
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stuart C Sealfon
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|