151
|
Zhang L, Feng X, Wang H, He S, Fan H, Liu D. Antibody-dependent enhancement of porcine reproductive and respiratory syndrome virus infection downregulates the levels of interferon-gamma/lambdas in porcine alveolar macrophages in vitro. Front Vet Sci 2023; 10:1150430. [PMID: 37008366 PMCID: PMC10050554 DOI: 10.3389/fvets.2023.1150430] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/24/2023] [Indexed: 03/17/2023] Open
Abstract
Fc gamma receptor-mediated antibody-dependent enhancement (ADE) can promote virus invasion of target cells, sometimes exacerbating the severity of the disease. ADE may be an enormous hurdle to developing efficacious vaccines for certain human and animal viruses. ADE of porcine reproductive and respiratory syndrome virus (PRRSV) infection has been demonstrated in vivo and in vitro. However, the effect of PRRSV-ADE infection on the natural antiviral immunity of the host cells is yet to be well investigated. Specifically, whether the ADE of PRRSV infection affects the levels of type II (interferon-gamma, IFN-γ) and III (interferon-lambdas, IFN-λs) interferons (IFNs) remains unclear. In this study, our results showed that PRRSV significantly induced the secretion of IFN-γ, IFN-λ1, IFN-λ3, and IFN-λ4 in porcine alveolar macrophages (PAMs) in early infection, and weakly inhibited the production of IFN-γ, IFN-λ1, IFN-λ3, and IFN-λ4 in PAMs in late infection. Simultaneously, PRRSV infection significantly increased the transcription of interferon-stimulated gene 15 (ISG15), ISG56, and 2′, 5′-oligoadenylate synthetase 2 (OAS2) in PAMs. In addition, our results showed that PRRSV infection in PAMs via the ADE pathway not only significantly decreased the synthesis of IFN-γ, IFN-λ1, IFN-λ3, and IFN-λ4 but also significantly enhanced the generation of transforming growth factor-beta1 (TGF-β1). Our results also showed that the ADE of PRRSV infection significantly reduced the mRNAs of ISG15, ISG56, and OAS2 in PAMs. In conclusion, our studies indicated that PRRSV-ADE infection suppressed innate antiviral response by downregulating the levels of type II and III IFNs, hence facilitating viral replication in PAMs in vitro. The ADE mechanism demonstrated in the present study furthered our understanding of persistent pathogenesis following PRRSV infection mediated by antibodies.
Collapse
|
152
|
Addressing Natural Killer Cell Dysfunction and Plasticity in Cell-Based Cancer Therapeutics. Cancers (Basel) 2023; 15:cancers15061743. [PMID: 36980629 PMCID: PMC10046032 DOI: 10.3390/cancers15061743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Natural killer (NK) cells are cytotoxic group 1 innate lymphoid cells (ILC), known for their role as killers of stressed, cancerous, and virally infected cells. Beyond this cytotoxic function, NK cell subsets can influence broader immune responses through cytokine production and have been linked to central roles in non-immune processes, such as the regulation of vascular remodeling in pregnancy and cancer. Attempts to exploit the anti-tumor functions of NK cells have driven the development of various NK cell-based therapies, which have shown promise in both pre-clinical disease models and early clinical trials. However, certain elements of the tumor microenvironment, such as elevated transforming growth factor (TGF)-β, hypoxia, and indoalemine-2,3-dioxygenase (IDO), are known to suppress NK cell function, potentially limiting the longevity and activity of these approaches. Recent studies have also identified these factors as contributors to NK cell plasticity, defined by the conversion of classical cytotoxic NK cells into poorly cytotoxic, tissue-resident, or ILC1-like phenotypes. This review summarizes the current approaches for NK cell-based cancer therapies and examines the challenges presented by tumor-linked NK cell suppression and plasticity. Ongoing efforts to overcome these challenges are discussed, along with the potential utility of NK cell therapies to applications outside cancer.
Collapse
|
153
|
Zhao Q, Li K, Jiang K, Yuan Z, Xiao M, Wei G, Zheng W, Wang X, Huang A. Proteomic approach-based comparison of metabolic pathways and functional activities of whey proteins derived from Guishan and Saanen goat milk. J Dairy Sci 2023; 106:2247-2260. [PMID: 36870847 DOI: 10.3168/jds.2022-22404] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/21/2022] [Indexed: 03/06/2023]
Abstract
Guishan goats, a unique goat breed in Yunnan Province, have a long history and representation, but their whey protein and function remain unclear. In this study, we carried out a quantitative analysis of the Guishan and Saanen goat whey proteome using a label-free proteomic approach. A total of 500 proteins were quantified from the 2 kinds of goat whey proteins, including 463 common proteins, 37 uniquely expressed whey proteins (UEWP), and 12 differentially expressed whey proteins (DEWP). Bioinformatics analysis indicated that UEWP and DEWP were mainly involved in cellular and immune system processes, membrane, and binding. In addition, UEWP and DEWP in Guishan goats participated primarily in metabolism and immune-related pathways, whereas Saanen goat whey proteins were associated mostly with environmental information processing-related pathways. Guishan goat whey promoted the growth of RAW264.7 macrophages more than Saanen goat whey, and significantly reduced the production of nitric oxide in lipopolysaccharide-stimulated RAW264.7 cells. This study provides a reference for further understanding these 2 goat whey proteins and finding functional active substances from them.
Collapse
Affiliation(s)
- Qiong Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Kunlin Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Kexin Jiang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Ziyou Yuan
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Menglin Xiao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Guangqiang Wei
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Wentao Zheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Xuefeng Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Aixiang Huang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| |
Collapse
|
154
|
Arianfar E, Khandoozi SR, Mohammadi S, Memarian A. Suppression of CD56 bright NK cells in breast cancer patients is associated with the PD-1 and TGF-βRII expression. Clin Transl Oncol 2023; 25:841-851. [PMID: 36414921 DOI: 10.1007/s12094-022-02997-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Natural killer (NK) cells, as professional cytotoxic cells, play a key role against cancer in the early and metastatic stages. Their functional defects are highly associated with the initiation or progression of breast cancer (BC). Here, we investigated the phenotypic characterization of NK cells in 26 newly diagnosed BC patients in comparison to 12 healthy counterparts. METHODS Expression of CXCR3 and PD-1, and also NKG2D, and TGF-βRII were studied on CD56dim and CD56bright NK cells from fresh peripheral blood (PB) samples using flow cytometry. The plasma levels of IFN-γ and soluble MIC-A levels were also assessed by ELISA. RESULTS Both CD56dim and CD56bright NK subtypes showed lower CXCR3 and NKG2D expression in BC patients than healthy subjects. Furthermore, patients' CD56bright NK cells significantly showed higher expression levels of TGF-βRII and PD-1. Interestingly, increased concentration of MIC-A level in plasma of BC patients was associated with the higher TGF-βRII and PD-1 expression in all NK cells, while the plasma level of IFN-γ was associated with the lower TGF-βRII expression on CD56bright NK cells in these patients. CONCLUSION Our results demonstrated phenotypically suppressed-NK cells, especially in the CD56bright subset of BC patients. It specifies their potential incompetence and outlines decrement of their anti-tumor activity, which could be interrelated with the tumor pathogenesis, TME immunosuppression, and so disease progression. The induction of compensatory mechanisms revives NK cells function and could be used in combination with the conventional treatments as a putative therapeutic approach for targeted immunotherapy.
Collapse
Affiliation(s)
- Elaheh Arianfar
- Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Saeed Mohammadi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Memarian
- Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran. .,Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
155
|
Foy SP, Jacoby K, Bota DA, Hunter T, Pan Z, Stawiski E, Ma Y, Lu W, Peng S, Wang CL, Yuen B, Dalmas O, Heeringa K, Sennino B, Conroy A, Bethune MT, Mende I, White W, Kukreja M, Gunturu S, Humphrey E, Hussaini A, An D, Litterman AJ, Quach BB, Ng AHC, Lu Y, Smith C, Campbell KM, Anaya D, Skrdlant L, Huang EYH, Mendoza V, Mathur J, Dengler L, Purandare B, Moot R, Yi MC, Funke R, Sibley A, Stallings-Schmitt T, Oh DY, Chmielowski B, Abedi M, Yuan Y, Sosman JA, Lee SM, Schoenfeld AJ, Baltimore D, Heath JR, Franzusoff A, Ribas A, Rao AV, Mandl SJ. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature 2023; 615:687-696. [PMID: 36356599 PMCID: PMC9768791 DOI: 10.1038/s41586-022-05531-1] [Citation(s) in RCA: 153] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022]
Abstract
T cell receptors (TCRs) enable T cells to specifically recognize mutations in cancer cells1-3. Here we developed a clinical-grade approach based on CRISPR-Cas9 non-viral precision genome-editing to simultaneously knockout the two endogenous TCR genes TRAC (which encodes TCRα) and TRBC (which encodes TCRβ). We also inserted into the TRAC locus two chains of a neoantigen-specific TCR (neoTCR) isolated from circulating T cells of patients. The neoTCRs were isolated using a personalized library of soluble predicted neoantigen-HLA capture reagents. Sixteen patients with different refractory solid cancers received up to three distinct neoTCR transgenic cell products. Each product expressed a patient-specific neoTCR and was administered in a cell-dose-escalation, first-in-human phase I clinical trial ( NCT03970382 ). One patient had grade 1 cytokine release syndrome and one patient had grade 3 encephalitis. All participants had the expected side effects from the lymphodepleting chemotherapy. Five patients had stable disease and the other eleven had disease progression as the best response on the therapy. neoTCR transgenic T cells were detected in tumour biopsy samples after infusion at frequencies higher than the native TCRs before infusion. This study demonstrates the feasibility of isolating and cloning multiple TCRs that recognize mutational neoantigens. Moreover, simultaneous knockout of the endogenous TCR and knock-in of neoTCRs using single-step, non-viral precision genome-editing are achieved. The manufacture of neoTCR engineered T cells at clinical grade, the safety of infusing up to three gene-edited neoTCR T cell products and the ability of the transgenic T cells to traffic to the tumours of patients are also demonstrated.
Collapse
MESH Headings
- Humans
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Biopsy
- Cell- and Tissue-Based Therapy/adverse effects
- Cell- and Tissue-Based Therapy/methods
- Cytokine Release Syndrome/complications
- Disease Progression
- Encephalitis/complications
- Gene Editing
- Gene Knock-In Techniques
- Gene Knockout Techniques
- Genes, T-Cell Receptor alpha
- Genes, T-Cell Receptor beta
- Mutation
- Neoplasms/complications
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Patient Safety
- Precision Medicine/adverse effects
- Precision Medicine/methods
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transgenes/genetics
- HLA Antigens/immunology
- CRISPR-Cas Systems
Collapse
Affiliation(s)
| | | | - Daniela A Bota
- Department of Neurology and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
| | | | - Zheng Pan
- PACT Pharma, South San Francisco, CA, USA
| | | | - Yan Ma
- PACT Pharma, South San Francisco, CA, USA
| | - William Lu
- PACT Pharma, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | - Ines Mende
- PACT Pharma, South San Francisco, CA, USA
| | | | | | | | | | | | - Duo An
- PACT Pharma, South San Francisco, CA, USA
| | | | | | | | - Yue Lu
- Institute for Systems Biology, Seattle, WA, USA
| | - Chad Smith
- PACT Pharma, South San Francisco, CA, USA
| | - Katie M Campbell
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | | - Roel Funke
- PACT Pharma, South San Francisco, CA, USA
| | | | | | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Bartosz Chmielowski
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Mehrdad Abedi
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Jeffrey A Sosman
- Department of Medicine and Robert H. Lurie Cancer Center, Northwestern University, Evanston, IL, USA
| | - Sylvia M Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Adam J Schoenfeld
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | | | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA.
| | | | | |
Collapse
|
156
|
Zhen Z, Wenwen Y, Guanghui H, Chenghua L, Zhimeng L. AjTGFβ alleviates V. splendidus-induced inflammation through SMADs pathway in Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2023; 134:108593. [PMID: 36746229 DOI: 10.1016/j.fsi.2023.108593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/05/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
The inhibition of inflammatory response is an essential process to control the development of inflammation and is an important step to protect the organism from excessive inflammatory damage. As a pleiotropic cytokine, transforming growth factor beta (TGF-β) plays a regulatory role in inhibiting inflammation in vertebrates. To investigate the role of TGF-β in the regulation of inflammation in invertebrates, we cloned and characterized the TGF-β gene from Apostichopus japonicus via rapid amplification of cDNA ends, and the sample was designated as AjTGF-β. For Vibrio splendidus-challenged sea cucumbers, the expression of AjTGF-β mRNAs in coelomocytes decreased at 96 h (0.27-fold), which was contrary to the trend of inflammation. AjTGF-β was expressed in all tissues with the highest expression in the body wall. When AjTGF-β was knocked down by using small interfering RNA (siRNA-KD) to 0.45-fold, AjSMAD 2/3 and AjSMAD6 were downregulated to 0.32- and 0.05-fold compared with the control group, respectively. Furthermore, when the damaged sea cucumber was challenged by V. splendidus co-incubated with rAjTGF-β, the damage area had no extensive inflammation, and damaged repair appeared at 72 h compared with the Vs + BSA group, in which the expression of AjSMAD 2/3 was upregulated by 1.35-fold. Under this condition, AjSMAD 2/3 silencing alleviated rAjTGF-β-induced damage recovery. Moreover, rAjTGF-β slightly induced the collagen I expression from 6.13 ng/mL to 7.84 ng/mL, and collagen III was upregulated from 6.23 ng/mL to 6.89 ng/mL compared with the Vs + BSA group. This finding indicates that AjTGF-β negatively regulated the inflammatory progress and accelerated the repair of damage by AjSMADs to regulate the collagens expression.
Collapse
Affiliation(s)
- Zhang Zhen
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Ye Wenwen
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Han Guanghui
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Li Chenghua
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Lv Zhimeng
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
157
|
Heterogeneity of Phenotypic and Functional Changes to Porcine Monocyte-Derived Macrophages Triggered by Diverse Polarizing Factors In Vitro. Int J Mol Sci 2023; 24:ijms24054671. [PMID: 36902099 PMCID: PMC10003195 DOI: 10.3390/ijms24054671] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Swine are attracting increasing attention as a biomedical model, due to many immunological similarities with humans. However, porcine macrophage polarization has not been extensively analyzed. Therefore, we investigated porcine monocyte-derived macrophages (moMΦ) triggered by either IFN-γ + LPS (classical activation) or by diverse "M2-related" polarizing factors: IL-4, IL-10, TGF-β, and dexamethasone. IFN-γ and LPS polarized moMΦ toward a proinflammatory phenotype, although a significant IL-1Ra response was observed. Exposure to IL-4, IL-10, TGF-β, and dexamethasone gave rise to four distinct phenotypes, all antithetic to IFN-γ and LPS. Some peculiarities were observed: IL-4 and IL-10 both enhanced expression of IL-18, and none of the "M2-related" stimuli induced IL-10 expression. Exposures to TGF-β and dexamethasone were characterized by enhanced levels of TGF-β2, whereas stimulation with dexamethasone, but not TGF-β2, triggered CD163 upregulation and induction of CCL23. Macrophages stimulated with IL-10, TGF-β, or dexamethasone presented decreased abilities to release proinflammatory cytokines in response to TLR2 or TLR3 ligands: IL-10 showed a powerful inhibitory activity for CXCL8 and TNF release, whereas TGF-β provided a strong inhibitory signal for IL-6 production. While our results emphasized porcine macrophage plasticity broadly comparable to human and murine macrophages, they also highlighted some peculiarities in this species.
Collapse
|
158
|
Klück V, Cabău G, Mies L, Bukkems F, van Emst L, Bakker R, van Caam A, Crişan TO, Joosten LAB. TGF-β is elevated in hyperuricemic individuals and mediates urate-induced hyperinflammatory phenotype in human mononuclear cells. Arthritis Res Ther 2023; 25:30. [PMID: 36850003 PMCID: PMC9969669 DOI: 10.1186/s13075-023-03001-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 01/29/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Soluble urate leads to a pro-inflammatory phenotype in human monocytes characterized by increased production of IL-1β and downregulation of IL-1 receptor antagonist, the mechanism of which remains to be fully elucidated. Previous transcriptomic data identified differential expression of genes in the transforming growth factor (TGF)-β pathway in monocytes exposed to urate in vitro. In this study, we explore the role of TGF-β in urate-induced hyperinflammation in peripheral blood mononuclear cells (PBMCs). METHODS TGF-β mRNA in unstimulated PBMCs and protein levels in plasma were measured in individuals with normouricemia, hyperuricemia and gout. For in vitro validation, PBMCs of healthy volunteers were isolated and treated with a dose ranging concentration of urate for assessment of mRNA and pSMAD2. Urate and TGF-β priming experiments were performed with three inhibitors of TGF-β signalling: SB-505124, 5Z-7-oxozeaenol and a blocking antibody against TGF-β receptor II. RESULTS TGF-β mRNA levels were elevated in gout patients compared to healthy controls. TGF-β-LAP levels in serum were significantly higher in individuals with hyperuricemia compared to controls. In both cases, TGF-β correlated positively to serum urate levels. In vitro, urate exposure of PBMCs did not directly induce TGF-β but did enhance SMAD2 phosphorylation. The urate-induced pro-inflammatory phenotype of monocytes was partly reversed by blocking TGF-β. CONCLUSIONS TGF-β is elevated in individuals with hyperuricemia and correlated to serum urate concentrations. In addition, the urate-induced pro-inflammatory phenotype in human monocytes is mediated by TGF-β signalling. Future studies are warranted to explore the intracellular pathways involved and to assess the clinical significance of urate-TGF-β relation.
Collapse
Affiliation(s)
- Viola Klück
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Georgiana Cabău
- Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Linda Mies
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - Femke Bukkems
- Departement of Rheumatology, Radboud UMC, Nijmegen, The Netherlands
| | - Liesbeth van Emst
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - René Bakker
- Departement of Rheumatology, Radboud UMC, Nijmegen, The Netherlands
| | - Arjan van Caam
- Departement of Rheumatology, Radboud UMC, Nijmegen, The Netherlands
| | | | - Tania O Crişan
- Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands. .,Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands. .,Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj Napoca, Romania.
| |
Collapse
|
159
|
Pan L, Hao W, Xue Y, Wang K, Zheng X, Luo J, Ba X, Xiang Y, Qin X, Bergwik J, Tanner L, Egesten A, Brasier AR, Boldogh I. 8-Oxoguanine targeted by 8-oxoguanine DNA glycosylase 1 (OGG1) is central to fibrogenic gene activation upon lung injury. Nucleic Acids Res 2023; 51:1087-1102. [PMID: 36651270 PMCID: PMC9943661 DOI: 10.1093/nar/gkac1241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 01/19/2023] Open
Abstract
Reactive oxygen species (ROS) are implicated in epithelial cell-state transition and deposition of extracellular matrix upon airway injury. Of the many cellular targets of ROS, oxidative DNA modification is a major driving signal. However, the role of oxidative DNA damage in modulation profibrotic processes has not been fully delineated. Herein, we report that oxidative DNA base lesions, 8-oxoG, complexed with 8-oxoguanine DNA glycosylase 1 (OGG1) functions as a pioneer factor, contributing to transcriptional reprogramming within airway epithelial cells. We show that TGFβ1-induced ROS increased 8-oxoG levels in open chromatin, dynamically reconfigure the chromatin state. OGG1 complexed with 8-oxoG recruits transcription factors, including phosphorylated SMAD3, to pro-fibrotic gene promoters thereby facilitating gene activation. Moreover, 8-oxoG levels are elevated in lungs of mice subjected to TGFβ1-induced injury. Pharmacologic targeting of OGG1 with the selective small molecule inhibitor of 8-oxoG binding, TH5487, abrogates fibrotic gene expression and remodeling in this model. Collectively, our study implicates that 8-oxoG substrate-specific binding by OGG1 is a central modulator of transcriptional regulation in response to tissue repair.
Collapse
Affiliation(s)
- Lang Pan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wenjing Hao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100871, China
| | - Yaoyao Xue
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ke Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xu Zheng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jixian Luo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- School of Life Sciences, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xueqing Ba
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Key Laboratory of Molecular Epigenetics of Ministry of Education, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410000, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410000, China
| | - Jesper Bergwik
- Respiratory Medicine, Allergology, & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 84 Lund, Sweden
| | - Lloyd Tanner
- Respiratory Medicine, Allergology, & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 84 Lund, Sweden
| | - Arne Egesten
- Respiratory Medicine, Allergology, & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 84 Lund, Sweden
| | - Allan R Brasier
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI 53705, USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
160
|
O’Sullivan JA, Kohlhapp FJ, Zloza A, Plaza-Rojas L, Burke B, Dulin NO, Guevara-Patiño JA. Memory Precursors and Short-Lived Effector T cell Subsets Have Different Sensitivities to TGFβ. Int J Mol Sci 2023; 24:ijms24043930. [PMID: 36835342 PMCID: PMC9966622 DOI: 10.3390/ijms24043930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
After exposure to an antigen, CD8 T cells reach a decision point about their fate: to become either short-lived effector cells (SLECs) or memory progenitor effector cells (MPECs). SLECs are specialized in providing an immediate effector function but have a shorter lifespan and lower proliferative capacity compared to MPECs. Upon encountering the cognate antigen during an infection, CD8 T cells rapidly expand and then contract to a level that is maintained for the memory phase after the peak of the response. Studies have shown that the contraction phase is mediated by TGFβ and selectively targets SLECs, while sparing MPECs. The aim of this study is to investigate how the CD8 T cell precursor stage determines TGFβ sensitivity. Our results demonstrate that MPECs and SLECs have differential responses to TGFβ, with SLECs being more sensitive to TGFβ than MPECs. This difference in sensitivity is associated with the levels of TGFβRI and RGS3, and the SLEC-related transcriptional activator T-bet binding to the TGFβRI promoter may provide a molecular basis for increased TGFβ sensitivity in SLECs.
Collapse
Affiliation(s)
- Jeremy A. O’Sullivan
- Department of Surgery, and Cancer Biology, Loyola University Chicago, IL 60153, USA
| | | | - Andrew Zloza
- Department of Surgery, and Cancer Biology, Loyola University Chicago, IL 60153, USA
| | - Lourdes Plaza-Rojas
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Brianna Burke
- Department of Surgery, and Cancer Biology, Loyola University Chicago, IL 60153, USA
| | - Nickolai O. Dulin
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - José A. Guevara-Patiño
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Correspondence:
| |
Collapse
|
161
|
Landscape of Well-Coordinated Fracture Healing in a Mouse Model Using Molecular and Cellular Analysis. Int J Mol Sci 2023; 24:ijms24043569. [PMID: 36834981 PMCID: PMC9964763 DOI: 10.3390/ijms24043569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The success of fracture healing relies on overlapping but coordinated cellular and molecular events. Characterizing an outline of differential gene regulation throughout successful healing is essential for identifying crucial phase-specific markers and may serve as the basis for engineering these in challenging healing situations. This study analyzed the healing progression of a standard closed femoral fracture model in C57BL/6N (age = 8 weeks) wild-type male mice. The fracture callus was assessed across various days post fracture (D = days 0, 3, 7, 10, 14, 21, and 28) by microarray, with D0 serving as a control. Histological analyses were carried out on samples from D7 until D28 to support the molecular findings. Microarray analysis revealed a differential regulation of immune response, angiogenesis, ossification, extracellular matrix regulation, mitochondrial and ribosomal genes during healing. In-depth analysis showed differential regulation of mitochondrial and ribosomal genes during the initial phase of healing. Furthermore, the differential gene expression showed an essential role of Serpin Family F Member 1 over the well-known Vascular Endothelial Growth Factor in angiogenesis, especially during the inflammatory phase. The significant upregulation of matrix metalloproteinase 13 and bone sialoprotein from D3 until D21 asserts their importance in bone mineralization. The study also shows type I collagen around osteocytes located in the ossified region at the periosteal surface during the first week of healing. Histological analysis of matrix extracellular phosphoglycoprotein and extracellular signal-regulated kinase stressed their roles in bone homeostasis and the physiological bone-healing process. This study reveals previously unknown and novel candidates, that could serve as a target for specific time points in healing and to remedy cases of impaired healing.
Collapse
|
162
|
Kaur S, Selden NR, Aballay A. Anti-inflammatory effects of vagus nerve stimulation in pediatric patients with epilepsy. Front Immunol 2023; 14:1093574. [PMID: 36845140 PMCID: PMC9951815 DOI: 10.3389/fimmu.2023.1093574] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/20/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction The neural control of the immune system by the nervous system is critical to maintaining immune homeostasis, whose disruption may be an underlying cause of several diseases, including cancer, multiple sclerosis, rheumatoid arthritis, and Alzheimer's disease. Methods Here we studied the role of vagus nerve stimulation (VNS) on gene expression in peripheral blood mononuclear cells (PBMCs). Vagus nerve stimulation is widely used as an alternative treatment for drug-resistant epilepsy. Thus, we studied the impact that VNS treatment has on PBMCs isolated from a cohort of existing patients with medically refractory epilepsy. A comparison of genome-wide changes in gene expression was made between the epilepsy patients treated and non-treated with vagus nerve stimulation. Results The analysis showed downregulation of genes related to stress, inflammatory response, and immunity, suggesting an anti-inflammatory effect of VNS in epilepsy patients. VNS also resulted in the downregulation of the insulin catabolic process, which may reduce circulating blood glucose. Discussion These results provide a potential molecular explanation for the beneficial role of the ketogenic diet, which also controls blood glucose, in treating refractory epilepsy. The findings indicate that direct VNS might be a useful therapeutic alternative to treat chronic inflammatory conditions.
Collapse
Affiliation(s)
- Supender Kaur
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, United States
| | - Nathan R. Selden
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Alejandro Aballay
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
163
|
Arnhold J. Host-Derived Cytotoxic Agents in Chronic Inflammation and Disease Progression. Int J Mol Sci 2023; 24:ijms24033016. [PMID: 36769331 PMCID: PMC9918110 DOI: 10.3390/ijms24033016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
At inflammatory sites, cytotoxic agents are released and generated from invading immune cells and damaged tissue cells. The further fate of the inflammation highly depends on the presence of antagonizing principles that are able to inactivate these host-derived cytotoxic agents. As long as the affected tissues are well equipped with ready-to-use protective mechanisms, no damage by cytotoxic agents occurs and resolution of inflammation is initiated. However, long-lasting and severe immune responses can be associated with the decline, exhaustion, or inactivation of selected antagonizing principles. Hence, cytotoxic agents are only partially inactivated and contribute to damage of yet-unperturbed cells. Consequently, a chronic inflammatory process results. In this vicious circle of permanent cell destruction, not only novel cytotoxic elements but also novel alarmins and antigens are liberated from affected cells. In severe cases, very low protection leads to organ failure, sepsis, and septic shock. In this review, the major classes of host-derived cytotoxic agents (reactive species, oxidized heme proteins and free heme, transition metal ions, serine proteases, matrix metalloproteases, and pro-inflammatory peptides), their corresponding protective principles, and resulting implications on the pathogenesis of diseases are highlighted.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Medical Faculty, Institute of Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
164
|
Pal-Ghosh S, Karpinski BA, Datta Majumdar H, Ghosh T, Thomasian J, Brooks SR, Sawaya AP, Morasso MI, Scholand KK, de Paiva CS, Galletti JG, Stepp MA. Molecular mechanisms regulating wound repair: Evidence for paracrine signaling from corneal epithelial cells to fibroblasts and immune cells following transient epithelial cell treatment with Mitomycin C. Exp Eye Res 2023; 227:109353. [PMID: 36539051 PMCID: PMC10560517 DOI: 10.1016/j.exer.2022.109353] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
In this paper, we use RNAseq to identify senescence and phagocytosis as key factors to understanding how mitomyin C (MMC) stimulates regenerative wound repair. We use conditioned media (CM) from untreated (CMC) and MMC treated (CMM) human and mouse corneal epithelial cells to show that corneal epithelial cells indirectly exposed to MMC secrete elevated levels of immunomodulatory proteins including IL-1α and TGFβ1 compared to cells exposed to CMC. These factors increase epithelial and macrophage phagocytosis and promote ECM turnover. IL-1α supplementation can increase phagocytosis in control epithelial cells and attenuate TGFβ1 induced αSMA expression by corneal fibroblasts. Yet, we show that epithelial cell CM contains factors besides IL-1α that regulate phagocytosis and αSMA expression by fibroblasts. Exposure to CMM also impacts the activation of bone marrow derived dendritic cells and their ability to present antigen. These in vitro studies show how a brief exposure to MMC induces corneal epithelial cells to release proteins and other factors that function in a paracrine way to enhance debris removal and enlist resident epithelial and immune cells as well as stromal fibroblasts to support regenerative and not fibrotic wound healing.
Collapse
Affiliation(s)
- Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Beverly A Karpinski
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Himani Datta Majumdar
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Trisha Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Julie Thomasian
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew P Sawaya
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maria I Morasso
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kaitlin K Scholand
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Biosciences, Rice University, TX, 77030, USA
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeremias G Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (IMEX), National Academy of Medicine/CONICET, Buenos Aires, Argentina
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA.
| |
Collapse
|
165
|
Guo ZH, Khattak S, Rauf MA, Ansari MA, Alomary MN, Razak S, Yang CY, Wu DD, Ji XY. Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders. Molecules 2023; 28:1283. [PMID: 36770950 PMCID: PMC9921752 DOI: 10.3390/molecules28031283] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023] Open
Abstract
Central nervous system disorders, especially neurodegenerative diseases, are a public health priority and demand a strong scientific response. Various therapy procedures have been used in the past, but their therapeutic value has been insufficient. The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier is two of the barriers that protect the central nervous system (CNS), but are the main barriers to medicine delivery into the CNS for treating CNS disorders, such as brain tumors, Parkinson's disease, Alzheimer's disease, and Huntington's disease. Nanotechnology-based medicinal approaches deliver valuable cargos targeting molecular and cellular processes with greater safety, efficacy, and specificity than traditional approaches. CNS diseases include a wide range of brain ailments connected to short- and long-term disability. They affect millions of people worldwide and are anticipated to become more common in the coming years. Nanotechnology-based brain therapy could solve the BBB problem. This review analyzes nanomedicine's role in medication delivery; immunotherapy, chemotherapy, and gene therapy are combined with nanomedicines to treat CNS disorders. We also evaluated nanotechnology-based approaches for CNS disease amelioration, with the intention of stimulating the immune system by delivering medications across the BBB.
Collapse
Affiliation(s)
- Zi-Hua Guo
- Department of Neurology, Kaifeng Hospital of Traditional Chinese Medicine, No. 54 East Caizhengting St., Kaifeng 475000, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Mohd Ahmar Rauf
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Sufyan Razak
- Dow Medical College, John Hopkins Medical Center, School of Medicine, Baltimore, MD 21205, USA
| | - Chang-Yong Yang
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- School of Stomatology, Henan University, Kaifeng 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
166
|
Kang K, Cui L, Zhang Q, Gao S. Leucine rich repeat containing 32 accelerates tenogenic differentiation of tendon-derived stem cells and promotes Achilles tendon repair in rats. Exp Anim 2023; 72:9-18. [PMID: 35934780 PMCID: PMC9978125 DOI: 10.1538/expanim.22-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Although many surgical or non-operative therapies have been developed to treat Achilles tendon injuries, the prognosis of which is often unsatisfactory. Recently, biologic approaches using multipotent stem cells like tendon-derived stem cells (TDSCs) pose a possible treatment option. To evaluate whether the Leucine rich repeat containing 32 (Lrrc32) affects the tenogenic differentiation of TDSCs and thus promotes Achilles tendon healing. TDSCs were infected with the recombinant Lrrc32-overexpressing lentivirus (LV-Lrrc32) and then locally injected into the injured site of rat. Four weeks after surgery, the Achilles tendon tissue (~0.5 cm) around the injured area was harvested for analysis. Pathological results showed that Lrrc32-overexpressing TDSCs significantly improved the morphological changes of the injured tendons. Specifically, the increased collagen-I expression and hydroxyproline content in extracellular matrix, and more orderly arrangement of the regenerated collagen fibers were observed in the Lrrc32 overexpression group. Moreover, 4 weeks after injection of Lrrc32-overexpressing TDSCs, the expression of tenocyte-related genes such as tenomodulin (Tnmd), scleraxis (Scx) and decorin (Dcn) were upregulated in the area of the healing tendon. These findings indicated that Lrrc32 promoted the tenogenic differentiation of TDSCs in vivo. Additionally, Lrrc32 overexpression also increased the expression of TGF-β1 and p-SMAD2/3, suggesting that the beneficial effects of Lrrc32 on tendon repair might be associated with the expression of TGF-β1 and p-SMAD2/3. Our findings collectively revealed that Lrrc32-overexpressed TDSCs promoted tendon healing more effectively than TDSCs alone.
Collapse
Affiliation(s)
- Kai Kang
- The Second Department of Joint Surgery, Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, Hebei 050051, P.R. China
| | - Lukuan Cui
- The Second Department of Joint Surgery, Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, Hebei 050051, P.R. China
| | - Qian Zhang
- The Second Department of Joint Surgery, Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, Hebei 050051, P.R. China
| | - Shijun Gao
- The Second Department of Joint Surgery, Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
167
|
Kryukov AI, Gankovskaya LV, Bondareva GP, Kunelskaya NL, Garov EV, Nasaeva ED, Martirosyan TG. [Features of innate immunity indicators in patients with recurrent exudative otitis media]. Vestn Otorinolaringol 2023; 88:4-9. [PMID: 36867137 DOI: 10.17116/otorino2022880114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
The article presents the features of the indicators of the innate immune response (TLR4, IL1B, TGFB, HBD1, and HBD2) in the exudate of the tympanic cavity in patients with recurrent exudative otitis media (EOM) with normal patency of the auditory tube and its dysfunction. The results of the study demonstrate changes in the indices of the innate immune response characteristic of the inflammatory process in patients with recurrent EOM against the background of dysfunction of the auditory tube in comparison with the group where it is absent. The data obtained can be used to clarify the pathogenesis of otitis media with dysfunction of the auditory tube, to develop new methods of diagnosis, prevention and therapy.
Collapse
Affiliation(s)
- A I Kryukov
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - L V Gankovskaya
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - G P Bondareva
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - N L Kunelskaya
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - E V Garov
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - E D Nasaeva
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - T G Martirosyan
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| |
Collapse
|
168
|
Soliman AM, Barreda DR. Acute Inflammation in Tissue Healing. Int J Mol Sci 2022; 24:ijms24010641. [PMID: 36614083 PMCID: PMC9820461 DOI: 10.3390/ijms24010641] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
There are well-established links between acute inflammation and successful tissue repair across evolution. Innate immune reactions contribute significantly to pathogen clearance and activation of subsequent reparative events. A network of molecular and cellular regulators supports antimicrobial and tissue repair functions throughout the healing process. A delicate balance must be achieved between protection and the potential for collateral tissue damage associated with overt inflammation. In this review, we summarize the contributions of key cellular and molecular components to the acute inflammatory process and the effective and timely transition toward activation of tissue repair mechanisms. We further discuss how the disruption of inflammatory responses ultimately results in chronic non-healing injuries.
Collapse
Affiliation(s)
- Amro M. Soliman
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Daniel R. Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence: ; Tel.: +1-(780)492-0375
| |
Collapse
|
169
|
Gómez-Bernal F, Quevedo-Abeledo JC, García-González M, Fernández-Cladera Y, González-Rivero AF, de Vera-González A, Martín-González C, González-Gay MÁ, Ferraz-Amaro I. Serum Levels of Transforming Growth Factor Beta 1 in Systemic Lupus Erythematosus Patients. Biomolecules 2022; 13:biom13010073. [PMID: 36671458 PMCID: PMC9855909 DOI: 10.3390/biom13010073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a highly pleiotropic cytokine that has broad anti-inflammatory and immunosuppressive effects. In patients with systemic lupus erythematosus (SLE), the immunosuppressive effect of TGF-β1 is thought to be dysfunctional. In the present work, we aimed to study the relationship between the serum levels of TGF-β1 with the characteristics of the disease as well as with the patterns of activity, damage, or severity of the disease. Two hundred and eighty-four patients with well-characterized SLE were recruited. The serum levels of TGF-β1 were assessed. A multivariable linear regression analysis was performed to analyze the relation of disease characteristics to TGF-β1. The Katz severity index (beta coefficient 179 [95% confidence interval 7-350] pg/mL, p = 0.041) and SLEDAI activity index (beta coefficient 96 [95% CI 20-171] pg/mL, p = 0.014) were associated with higher serum levels of TGF-β1 after the multivariable analysis. When the disease-specific features were studied, ocular and cardiovascular manifestations were positively associated with serum TGF-β1 levels. In contrast, gastrointestinal and musculoskeletal involvements were associated with lower levels of circulating TGF-β1. Among patients with SLE, the serum levels of TGF-β1 were highly associated with disease-related manifestations.
Collapse
Affiliation(s)
- Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain
| | | | | | | | | | | | - Candelaria Martín-González
- Division of Internal Medicine, Hospital Universitario de Canarias, 38320 Tenerife, Spain
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Miguel Á. González-Gay
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39011 Santander, Spain
- Division of Rheumatology, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, 39011 Santander, Spain
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
- Correspondence: (M.Á.G.-G.); (I.F.-A.)
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
- Correspondence: (M.Á.G.-G.); (I.F.-A.)
| |
Collapse
|
170
|
Wang D, Sun Z, Zhu X, Zheng X, Zhou Y, Lu Y, Yan P, Wang H, Liu H, Jin J, Zhu H, Sun R, Wang Y, Fu B, Tian Z, Wei H. GARP-mediated active TGF-β1 induces bone marrow NK cell dysfunction in AML patients with early relapse post-allo-HSCT. Blood 2022; 140:2788-2804. [PMID: 35981475 PMCID: PMC10653097 DOI: 10.1182/blood.2022015474] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/22/2022] [Accepted: 08/04/2022] [Indexed: 01/05/2023] Open
Abstract
Relapse is a leading cause of death after allogeneic hematopoietic stem cell transplantation (allo-HSCT) for acute myeloid leukemia (AML). However, the underlying mechanisms remain poorly understood. Natural killer (NK) cells play a crucial role in tumor surveillance and cancer immunotherapy, and NK cell dysfunction has been observed in various tumors. Here, we performed ex vivo experiments to systematically characterize the mechanisms underlying the dysfunction of bone marrow-derived NK (BMNK) cells isolated from AML patients experiencing early relapse after allo-HSCT. We demonstrated that higher levels of active transforming growth factor β1 (TGF-β1) were associated with impaired effector function of BMNK cells in these AML patients. TGF-β1 activation was induced by the overexpression of glycoprotein A repetitions predominant on the surface of CD4+ T cells. Active TGF-β1 significantly suppressed mTORC1 activity, mitochondrial oxidative phosphorylation, the proliferation, and cytotoxicity of BMNK cells. Furthermore, pretreatment with the clinical stage TGF-β1 pathway inhibitor, galunisertib, significantly restored mTORC1 activity, mitochondrial homeostasis, and cytotoxicity. Importantly, the blockade of the TGF-β1 signaling improved the antitumor activity of NK cells in a leukemia xenograft mouse model. Thus, our findings reveal a mechanism explaining BMNK cell dysfunction and suggest that targeted inhibition of TGF-β1 signaling may represent a potential therapeutic intervention to improve outcomes in AML patients undergoing allo-HSCT or NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Dongyao Wang
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Zimin Sun
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoyu Zhu
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaohu Zheng
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yonggang Zhou
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yichen Lu
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Peidong Yan
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Huiru Wang
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Huilan Liu
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Jing Jin
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Huaiping Zhu
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Wang
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Binqing Fu
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiming Wei
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
171
|
Zhang Q, Geng M, Li K, Gao H, Jiao X, Ai K, Wei X, Yang J. TGF-β1 suppresses the T-cell response in teleost fish by initiating Smad3- and Foxp3-mediated transcriptional networks. J Biol Chem 2022; 299:102843. [PMID: 36581209 PMCID: PMC9860442 DOI: 10.1016/j.jbc.2022.102843] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) can suppress the activation, proliferation, and function of many T-cell subsets, protecting organisms from inflammatory and autoimmune disease caused by an overexuberant immune response. However, whether and how TGF-β1 regulates T-cell immunity in early vertebrates remain unknown. Here, using a Nile tilapia (Oreochromis niloticus) model, we investigated suppression of the T-cell response by TGF-β1 in teleost species. Tilapia encodes an evolutionarily conserved TGF-β1, the expression of which in lymphocytes is significantly induced during the immune response following Edwardsiella piscicida infection. Once activated, tilapia T cells increase TGF-β1 production, which in turn suppresses proinflammatory cytokine expression and inhibits T-cell activation. Notably, we found administration of TGF-β1 cripples the proliferation of tilapia T cells, reduces the potential capacity of Th1/2 differentiation, and impairs the cytotoxic function, rendering the fish more vulnerable to bacterial infection. Mechanistically, TGF-β1 initiates the TGF-βR/Smad signaling pathway and triggers the phosphorylation and nuclear translocation of Smad2/3. Smad3 subsequently interacts with several transcriptional partners to repress transcription of cytokines IL-2 and IFN-γ but promote transcription of immune checkpoint regulator CTLA4 and transcription factor Foxp3. Furthermore, TGF-β1/Smad signaling further utilizes Foxp3 to achieve the cascade regulation of these T-cell genes. Taken together, our findings reveal a detailed mechanism by which TGF-β1 suppresses the T cell-based immunity in Nile tilapia and support the notion that TGF-β1 had already been employed to inhibit the T-cell response early in vertebrate evolution, thus providing novel insights into the evolution of the adaptive immune system.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ming Geng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Haiyou Gao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Kete Ai
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
172
|
Kareva EN, Fedotcheva TA, Semeikin AV, Kochina NA, Krasnoshchok EV, Shimanovskii NL. The mechanisms of anti-inflammatory action of enisamium iodide. TERAPEVT ARKH 2022; 94:1262-1267. [PMID: 37167164 DOI: 10.26442/00403660.2022.11.201961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 12/27/2022]
Abstract
Aim. The role of cyclooxygenases (COX-1 and/or COX-2), transcription nuclear factor NF-B, anti-inflammatory cytokines TGF1b, IL-4, IL-10 and pro-inflammatory cytokines IL-1, IL-6 were studied to substantiate the expediency of antiviral agent enisamium iodide (Nobazit) using to regulate key inflammatory components in acute respiratory infections, IL-8, TNF-alpha in the realization of the pharmacological activity of this drug.
Materials and methods. Gene expression was determined by real-time RT-PCR, the concentration of interleukins was determined by ELISA, and the viability of peripheral blood mononuclear cells (PBMC) was assessed by the MTT spectrophotometric method. The chemiluminescence method was used to assess PBMC oxidant activity.
Results. Enisamium iodide (10 M) reduced mRNA levels of COX-1, COX-2, NF-B, TGF1b, IL-1, IL-6 in stimulated PBMC of healthy donors by an average of 48% (p0.05). At 5 times higher concentration, 50 M, enisamium iodide suppressed the expression of these genes by an average of 43% (p0.05). At a concentration of 100 M, enisamium iodide reduced the expression of COX-2, TGF1b, IL-1, IL-6 by an average of 47% (p0.05). At a concentration of 10 M, enisamium iodide stimulated the secretion of IL-10 by mononuclear cells by 1.2 times, p0.05. The tested drug at a concentration of 50 M did not affected on the concentration of IL-1, IL-4, IL-8 and TNF-alpha, but significantly stimulated the production of IL-10 by 1.5 times, p0.05. The chemiluminescence method revealed that enisamium iodide in the entire concentration range (10100 M) does not reduce the viability of macrophages, but inhibits their oxidative activity (maximum value of CL intensity) by an average of 55% (p0.05).
Conclusion. The anti-inflammatory effect of enisamium iodide at a concentration of 10 M may be associated with inhibition of the expression of COX-1, 2, NF-B, IL-1, IL-6, TGF1b and an increase in the expression and production of IL-10. An additional contribution to the anti-inflammatory activity of enisamium iodide is made by its antioxidant and antiradical activity. The absence of the effect of enisamium iodide (10100 M) on the viability of PBMC indicates its safety for the cells of the immune system and the expediency of using it to suppress inflammatory reactions in acute respiratory infections, restore the quality of life of patients and the possibility of using Nobazit as an effective agent for treatment of these infections of various etiologies.
Collapse
|
173
|
Kubo E, Shibata S, Shibata T, Sasaki H, Singh DP. Role of Decorin in the Lens and Ocular Diseases. Cells 2022; 12:cells12010074. [PMID: 36611867 PMCID: PMC9818407 DOI: 10.3390/cells12010074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Decorin is an archetypal member of the small leucine-rich proteoglycan gene family and is involved in various biological functions and many signaling networks, interacting with extra-cellular matrix (ECM) components, growth factors, and receptor tyrosine kinases. Decorin also modulates the growth factors, cell proliferation, migration, and angiogenesis. It has been reported to be involved in many ischemic and fibrotic eye diseases, such as congenital stromal dystrophy of the cornea, anterior subcapsular fibrosis of the lens, proliferative vitreoretinopathy, et al. Furthermore, recent evidence supports its role in secondary posterior capsule opacification (PCO) after cataract surgery. The expression of decorin mRNA in lens epithelial cells in vitro was found to decrease upon transforming growth factor (TGF)-β-2 addition and increase upon fibroblast growth factor (FGF)-2 addition. Wound healing of the injured lens in mice transgenic for lens-specific human decorin was promoted by inhibiting myofibroblastic changes. Decorin may be associated with epithelial-mesenchymal transition and PCO development in the lens. Gene therapy and decorin administration have the potential to serve as excellent therapeutic approaches for modifying impaired wound healing, PCO, and other eye diseases related to fibrosis and angiogenesis. In this review, we present findings regarding the roles of decorin in the lens and ocular diseases.
Collapse
Affiliation(s)
- Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
- Correspondence: ; Tel.: +81-76-286-2211 (ext. 3412); Fax: +81-76-286-1010
| | - Shinsuke Shibata
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
| | - Teppei Shibata
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
| | - Hiroshi Sasaki
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 920-0293, Ishikawa, Japan
| | - Dhirendra P. Singh
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
174
|
Rana PS, Soler DC, Kort J, Driscoll JJ. Targeting TGF-β signaling in the multiple myeloma microenvironment: Steering CARs and T cells in the right direction. Front Cell Dev Biol 2022; 10:1059715. [PMID: 36578789 PMCID: PMC9790996 DOI: 10.3389/fcell.2022.1059715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) remains a lethal hematologic cancer characterized by the expansion of transformed plasma cells within the permissive bone marrow (BM) milieu. The emergence of relapsed and/or refractory MM (RRMM) is provoked through clonal evolution of malignant plasma cells that harbor genomic, metabolic and proteomic perturbations. For most patients, relapsed disease remains a major cause of overall mortality. Transforming growth factors (TGFs) have pleiotropic effects that regulate myelomagenesis as well as the emergence of drug resistance. Moreover, TGF-β modulates numerous cell types present with the tumor microenvironment, including many immune cell types. While numerous agents have been FDA-approved over the past 2 decades and significantly expanded the treatment options available for MM patients, the molecular mechanisms responsible for drug resistance remain elusive. Multiple myeloma is uniformly preceded by a premalignant state, monoclonal gammopathy of unknown significance, and both conditions are associated with progressive deregulation in host immunity characterized by reduced T cell, natural killer (NK) cell and antigen-presenting dendritic cell (DC) activity. TGF-β promotes myelomagenesis as well as intrinsic drug resistance by repressing anti-myeloma immunity to promote tolerance, drug resistance and disease progression. Hence, repression of TGF-β signaling is a prerequisite to enhance the efficacy of current and future immunotherapeutics. Novel strategies that incorporate T cells that have been modified to express chimeric antigen receptor (CARs), T cell receptors (TCRs) and bispecific T cell engagers (BiTEs) offer promise to block TGF-β signaling, overcome chemoresistance and enhance anti-myeloma immunity. Here, we describe the effects of TGF-β signaling on immune cell effectors in the bone marrow and emerging strategies to overcome TGF-β-mediated myeloma growth, drug resistance and survival.
Collapse
Affiliation(s)
- Priyanka S. Rana
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States,Case Comprehensive Cancer Center, Cleveland, OH, United States
| | - David C. Soler
- The Brain Tumor and Neuro-Oncology Center, The Center of Excellence for Translational Neuro-Oncology, Department of Neurosurgery, Case Western Reserve University, Cleveland, OH, United States
| | - Jeries Kort
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States,Case Comprehensive Cancer Center, Cleveland, OH, United States,Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - James J. Driscoll
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States,Case Comprehensive Cancer Center, Cleveland, OH, United States,Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States,*Correspondence: James J. Driscoll,
| |
Collapse
|
175
|
Biering SB, Gomes de Sousa FT, Tjang LV, Pahmeier F, Zhu C, Ruan R, Blanc SF, Patel TS, Worthington CM, Glasner DR, Castillo-Rojas B, Servellita V, Lo NTN, Wong MP, Warnes CM, Sandoval DR, Clausen TM, Santos YA, Fox DM, Ortega V, Näär AM, Baric RS, Stanley SA, Aguilar HC, Esko JD, Chiu CY, Pak JE, Beatty PR, Harris E. SARS-CoV-2 Spike triggers barrier dysfunction and vascular leak via integrins and TGF-β signaling. Nat Commun 2022; 13:7630. [PMID: 36494335 PMCID: PMC9734751 DOI: 10.1038/s41467-022-34910-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 11/09/2022] [Indexed: 12/13/2022] Open
Abstract
Severe COVID-19 is associated with epithelial and endothelial barrier dysfunction within the lung as well as in distal organs. While it is appreciated that an exaggerated inflammatory response is associated with barrier dysfunction, the triggers of vascular leak are unclear. Here, we report that cell-intrinsic interactions between the Spike (S) glycoprotein of SARS-CoV-2 and epithelial/endothelial cells are sufficient to induce barrier dysfunction in vitro and vascular leak in vivo, independently of viral replication and the ACE2 receptor. We identify an S-triggered transcriptional response associated with extracellular matrix reorganization and TGF-β signaling. Using genetic knockouts and specific inhibitors, we demonstrate that glycosaminoglycans, integrins, and the TGF-β signaling axis are required for S-mediated barrier dysfunction. Notably, we show that SARS-CoV-2 infection caused leak in vivo, which was reduced by inhibiting integrins. Our findings offer mechanistic insight into SARS-CoV-2-triggered vascular leak, providing a starting point for development of therapies targeting COVID-19.
Collapse
Affiliation(s)
- Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| | | | - Laurentia V Tjang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Felix Pahmeier
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Chi Zhu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Richard Ruan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sophie F Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Trishna S Patel
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | | | - Dustin R Glasner
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Bryan Castillo-Rojas
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Venice Servellita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Nicholas T N Lo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Colin M Warnes
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Daniel R Sandoval
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Thomas Mandel Clausen
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Yale A Santos
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Douglas M Fox
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Victoria Ortega
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Anders M Näär
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah A Stanley
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Charles Y Chiu
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - John E Pak
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - P Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
176
|
Zhao Q, Dai H, Hu Y, Jiang H, Feng Z, Liu W, Dong Z, Tang X, Hou F, Rui H, Liu B. Cytokines network in primary membranous nephropathy. Int Immunopharmacol 2022; 113:109412. [DOI: 10.1016/j.intimp.2022.109412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
|
177
|
VandenBroek MM, Skebo SI, Ormiston ML. Targeting BMPR-II in pulmonary arterial hypertension: a case of Hercules versus the Hydra? Expert Opin Ther Targets 2022; 26:1027-1030. [PMID: 36638064 DOI: 10.1080/14728222.2022.2168188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/10/2023] [Indexed: 01/14/2023]
Affiliation(s)
| | - Sofia I Skebo
- Department of Biology, Queen's University, Kingston, K7L, Canada
| | - Mark L Ormiston
- Department of Medicine, Queen's University, Kingston, K7L, Canada
- Departments of Biomedical and Molecular Sciences and Surgery, Queen's University, Kingston, K7L, Canada
| |
Collapse
|
178
|
Salem MOA, Taştan Y, Bilen S, Terzi E, Sönmez AY. Effects of white mustard (Sinapis alba) oil on growth performance, immune response, blood parameters, digestive and antioxidant enzyme activities in rainbow trout (Oncorhynchusmykiss). FISH & SHELLFISH IMMUNOLOGY 2022; 131:283-299. [PMID: 36210002 DOI: 10.1016/j.fsi.2022.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
A study was conducted to evaluate the effects of dietary supplementation of white mustard (Sinapis alba) oil (WMO) on growth performance, immune responses, digestive and antioxidant enzyme activities in juvenile rainbow trout (Oncorhynchus mykiss). For this purpose, fish (initial weight: 25.77 ± 0.13 g) were divided into four experimental groups in triplicate and fed ad libitum twice a day with diets containing WMO at 0 (control), 0.5, 1, and 1.5% of diet for 9 weeks. Three fish from each tank (n:9 per treatment) were sampled on 21st, 42nd, and 63rd days for further analyses. At the end of the feeding period, fish were challenged with Aeromonas hydrophila and Yersinia ruckeri in two separate experimental setups. Results showed that final weight, weight gain, and specific growth rate were significantly increased in all experimental groups compared to the control. Feed conversion ratio was similar among treatments. Respiratory burst and potential killing activity decreased in all experimental groups compared to the control (P < 0.05). Lysozyme and myeloperoxidase activities were elevated in all experimental groups at the end of the experiment compared to the control (P < 0.05). Cytokine gene expressions in the head kidney and intestine were elevated in all experimental groups compared to that of the control in general (P < 0.05). Hematological responses of the experimental fish groups were similar to that of the control (P > 0.05). Pepsin and trypsin levels decreased in all experimental groups (P < 0.05). In terms of antioxidant enzyme activities, significant improvement in liver superoxide dismutase, catalase, and glutathione s-transferase activities in all treatment groups were determined (P < 0.05). In addition, a significant decline in liver lipid peroxidation levels was recorded in all treated groups at all sampling times compared to the control (P < 0.05). At the end of this feeding trial, no significant differences (P > 0.05) were observed in survival against A. hydrophila among experimental groups compared to the control (P > 0.05). However, increased survival against Y. ruckeri was determined in experimental fish groups (P < 0.05). This study suggests that white mustard oil had a favorable effect on the overall health and growth of rainbow trout.
Collapse
Affiliation(s)
- Mohamed Omar Abdalla Salem
- Kastamonu University Institute of Science, Department of Aquaculture, Kastamonu, Turkey; Bani Waleed University, Faculty of Education, Department of Biology, Bani Walid, Libya
| | - Yiğit Taştan
- Kastamonu University, Faculty of Fisheries, Department of Aquaculture, Kastamonu, Turkey
| | - Soner Bilen
- Kastamonu University, Faculty of Fisheries, Department of Basic Sciences, Kastamonu, Turkey
| | - Ertugrul Terzi
- Kastamonu University, Faculty of Fisheries, Department of Aquaculture, Kastamonu, Turkey
| | - Adem Yavuz Sönmez
- Kastamonu University, Faculty of Fisheries, Department of Basic Sciences, Kastamonu, Turkey.
| |
Collapse
|
179
|
Liu W, Zhang J, Liang X, Wang Y, Liu R, Zhang R, Zha J, Martyniuk CJ. Environmental concentrations of 2, 4-DTBP cause immunotoxicity in zebrafish (Danio rerio) and may elicit ecological risk to wildlife. CHEMOSPHERE 2022; 308:136465. [PMID: 36126734 DOI: 10.1016/j.chemosphere.2022.136465] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/29/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
Synthetic phenolic antioxidant 2,4-di-tert-butylphenol (2,4-DTBP) has gained growing concerns due to relatively high concentrations in aquatic ecosystems. There are, however, significant knowledge gaps regarding its potential toxicity to aquatic organisms. In this study, zebrafish (Danio rerio) larvae were exposed to 0.01, 0.1, or 1 μM 2,4-DTBP for 6 d. Transcriptomic analysis of larvae revealed that biological processes related to anti-inflammatory function of macrophage M2 lineage were inhibited by 0.01 μM 2,4-DTBP. Decreases of transcripts related to the IL1B-MYD88-NF-κB pathway (i.e., il1b, il1rl1, myd88, irak4, irak1, traf6, ikbkg, nfkbia, nfkb) and protein levels of NF-κB in larvae intestine confirmed anti-inflammatory effects of 2,4-DTBP. Subsequently, larvae exposed to 2,4-DTBP were challenged with E. coli and showed higher survival rate, suggesting sustained activation of inflammation via LPS can be attenuated by 2,4-DTBP. Moreover, histological examination revealed that intestine barrier was compromised and there was an imbalance of intestine macrophage homeostasis. Food intake was also reduced following exposure to 0.1 and 1 μM 2,4-DTBP. In addition, a risk assessment revealed that 2,4-DTBP in surface water pose low to high ecological risks to aquatic organisms. Taken together, exposure to environmentally relevant concentrations of 2,4-DTBP could negatively affect immune response in zebrafish and may elicit ecological risk in fish population.
Collapse
Affiliation(s)
- Wang Liu
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010021, China; Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Jiye Zhang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010021, China
| | - Xuefang Liang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010021, China.
| | - Yuchen Wang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010021, China
| | - Ruimin Liu
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010021, China
| | - Ruiqing Zhang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, School of Ecology and Environment, Inner Mongolia University, Hohhot, 010021, China
| | - Jinmiao Zha
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
180
|
TGF-β Inhibitors for Therapeutic Management of Kidney Fibrosis. Pharmaceuticals (Basel) 2022; 15:ph15121485. [PMID: 36558936 PMCID: PMC9783223 DOI: 10.3390/ph15121485] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 11/26/2022] [Indexed: 11/30/2022] Open
Abstract
Kidney fibrosis is a common pathophysiological mechanism of chronic kidney disease (CKD) progression caused by several underlying kidney diseases. Among various contributors to kidney fibrosis, transforming growth factor-β1 (TGF-β1) is the major factor driving fibrosis. TGF-β1 exerts its profibrotic attributes via the activation of canonical and non-canonical signaling pathways, which induce proliferation and activation of myofibroblasts and subsequent accumulation of extracellular matrix. Over the past few decades, studies have determined the TGF-β1 signaling pathway inhibitors and evaluated whether they could ameliorate the progression of CKD by hindering kidney fibrosis. However, therapeutic strategies that block TGF-β1 signaling have usually demonstrated unsatisfactory results. Herein, we discuss the therapeutic concepts of the TGF-β1 signaling pathway and its inhibitors and review the current state of the art regarding regarding TGF-β1 inhibitors in CKD management.
Collapse
|
181
|
Wu F, Xu X, Li W, Hong Y, Lai H, Zhang J, Wu X, Zhou K, Hu N. Nanoparticle-Delivered Transforming Growth Factor-β1 siRNA Induces PD-1 against Gastric Cancer by Transforming the Phenotype of the Tumor Immune Microenvironment. Pharmaceuticals (Basel) 2022; 15:ph15121487. [PMID: 36558938 PMCID: PMC9787292 DOI: 10.3390/ph15121487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Immune checkpoint blockade (ICB) is currently considered to be an important therapeutic method, which obtained FDA approval for clinical use in gastric cancer in 2017. As a new mechanism, it was found that the effect of αPDL1 could be improved by blocking the TGF-β1 signaling pathway, which converts the tumor immune microenvironment from the "immune-excluded phenotype" to the "immune-inflamed phenotype". Based on this phenomenon, this project was designed to prepare TGF-β1-siRNA-loaded PEG-PCL nanoparticles conjugated to αPDL1 (siTGF-β1-αPDL1-PEG-PCL) since we have linked similar antibodies to PEG-PCL previously. Therefore, MFC tumor-engrafted mice were established to simulate the biological characteristics of converting the phenotype of the immune microenvironment, and to study the anti-tumor effect and possible molecular mechanism. In this study, αPDL1 antibody conjugates markedly increased the cell uptake of NPs. The produced αPDL1-PEG-PCL NPs efficiently reduced the amounts of TGF-β1 mRNA in MFC cells, converting the immune microenvironment of MFC tumors engrafted mice from the "immune-excluded phenotype" to the "immune-inflamed phenotype". PDL1-harboring gastric cancer had increased susceptibility to αPDL1. The value of this drug-controlled release system targeting the tumor microenvironment in immune checkpoint therapy of gastric cancer would provide a scientific basis for clinically applying nucleic acid drugs.
Collapse
Affiliation(s)
- Fenglei Wu
- Department of Oncology, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
| | - Xiujuan Xu
- Department of Radiation Oncology, Lianyungang Second People’s Hospital (Lianyungang Cancer Hospital), Lianyungang 222023, China
| | - Wei Li
- Center of Research Laboratory, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
| | - Yidong Hong
- Department of Oncology, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
| | - Huan Lai
- Department of Oncology, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
| | - Jingzhou Zhang
- Department of Oncology, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
| | - Xueyu Wu
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang 222002, China
| | - Kangjie Zhou
- Department of Oncology, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
| | - Nan Hu
- Department of Oncology, The First Affiliated Hospital of Kangda College of Nanjing Medical University (The Affiliated Lianyungang Hospital of Xuzhou Medical University), Xuzhou 221004, China
- Correspondence:
| |
Collapse
|
182
|
Alilou M, Khorrami M, Prasanna P, Bera K, Gupta A, Viswanathan VS, Patil P, Velu PD, Fu P, Velcheti V, Madabhushi A. A tumor vasculature-based imaging biomarker for predicting response and survival in patients with lung cancer treated with checkpoint inhibitors. SCIENCE ADVANCES 2022; 8:eabq4609. [PMID: 36427313 PMCID: PMC9699671 DOI: 10.1126/sciadv.abq4609] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 10/06/2022] [Indexed: 05/30/2023]
Abstract
Tumor vasculature is a key component of the tumor microenvironment that can influence tumor behavior and therapeutic resistance. We present a new imaging biomarker, quantitative vessel tortuosity (QVT), and evaluate its association with response and survival in patients with non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitor (ICI) therapies. A total of 507 cases were used to evaluate different aspects of the QVT biomarkers. QVT features were extracted from computed tomography imaging of patients before and after ICI therapy to capture the tortuosity, curvature, density, and branching statistics of the nodule vasculature. Our results showed that QVT features were prognostic of OS (HR = 3.14, 0.95% CI = 1.2 to 9.68, P = 0.0006, C-index = 0.61) and could predict ICI response with AUCs of 0.66, 0.61, and 0.67 on three validation sets. Our study shows that QVT imaging biomarker could potentially aid in predicting and monitoring response to ICI in patients with NSCLC.
Collapse
Affiliation(s)
- Mehdi Alilou
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11790, USA
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Amit Gupta
- University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Pradnya Patil
- Department of Solid Tumor Oncology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Priya Darsini Velu
- Pathology and Laboratory Medicine, Weill Cornell Medicine Physicians, New York, NY 10021, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, CWRU, Cleveland, OH 44106, USA
| | - Vamsidhar Velcheti
- Department of Hematology and Oncology, NYU Langone Health, New York, NY 10016, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
- Atlanta Veterans Administration Medical Center, Atlanta, GA 30322, USA
| |
Collapse
|
183
|
Chang CY, You R, Armstrong D, Bandi A, Cheng YT, Burkhardt PM, Becerra-Dominguez L, Madison MC, Tung HY, Zeng Z, Wu Y, Song L, Phillips PE, Porter P, Knight JM, Putluri N, Yuan X, Marcano DC, McHugh EA, Tour JM, Catic A, Maneix L, Burt BM, Lee HS, Corry DB, Kheradmand F. Chronic exposure to carbon black ultrafine particles reprograms macrophage metabolism and accelerates lung cancer. SCIENCE ADVANCES 2022; 8:eabq0615. [PMID: 36383649 PMCID: PMC9668323 DOI: 10.1126/sciadv.abq0615] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Chronic exposure to airborne carbon black ultrafine (nCB) particles generated from incomplete combustion of organic matter drives IL-17A-dependent emphysema. However, whether and how they alter the immune responses to lung cancer remains unknown. Here, we show that exposure to nCB particles increased PD-L1+ PD-L2+ CD206+ antigen-presenting cells (APCs), exhausted T cells, and Treg cells. Lung macrophages that harbored nCB particles showed selective mitochondrial structure damage and decreased oxidative respiration. Lung macrophages sustained the HIF1α axis that increased glycolysis and lactate production, culminating in an immunosuppressive microenvironment in multiple mouse models of non-small cell lung cancers. Adoptive transfer of lung APCs from nCB-exposed wild type to susceptible mice increased tumor incidence and caused early metastasis. Our findings show that nCB exposure metabolically rewires lung macrophages to promote immunosuppression and accelerates the development of lung cancer.
Collapse
Affiliation(s)
- Cheng-Yen Chang
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ran You
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dominique Armstrong
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ashwini Bandi
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yi-Ting Cheng
- Developmental Biology Program, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Philip M. Burkhardt
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luis Becerra-Dominguez
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew C. Madison
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui-Ying Tung
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhimin Zeng
- Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yifan Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lizhen Song
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Patricia E. Phillips
- Cytometry and Cell Sorting Core, Baylor College of Medicine, Houston TX 77030, USA
| | - Paul Porter
- Cytometry and Cell Sorting Core, Baylor College of Medicine, Houston TX 77030, USA
| | - John M. Knight
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| | - Daniela C. Marcano
- Department of Chemistry and Smalley-Curl Institute, NanoCarbon Center, The Welch Institute for Advanced Materials, and Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005 USA
| | - Emily A. McHugh
- Department of Chemistry and Smalley-Curl Institute, NanoCarbon Center, The Welch Institute for Advanced Materials, and Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005 USA
| | - James M. Tour
- Department of Chemistry and Smalley-Curl Institute, NanoCarbon Center, The Welch Institute for Advanced Materials, and Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005 USA
| | - Andre Catic
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Developmental Biology Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Laure Maneix
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bryan M. Burt
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Division of Thoracic Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hyun-Sung Lee
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Division of Thoracic Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - David B. Corry
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Baylor College of Medicine, Houston, TX 77030, USA
| | - Farrah Kheradmand
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
184
|
Amer SA, Farahat M, Khamis T, Abdo SA, Younis EM, Abdel-Warith AWA, Reda R, Ali SA, Davies SJ, Ibrahim RE. Evaluation of Spray-Dried Bovine Hemoglobin Powder as a Dietary Animal Protein Source in Nile Tilapia, Oreochromis niloticus. Animals (Basel) 2022; 12:ani12223206. [PMID: 36428433 PMCID: PMC9687044 DOI: 10.3390/ani12223206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
The present study evaluated the potential effects of dietary inclusion of spray-dried bovine hemoglobin powder (SDBH) on the growth, gene expression of peptide and amino acid transporters, insulin growth factor-1 (IGF-1) and myostatin, digestive enzymes activity, intestinal histomorphology and immune status, immune-related gene expression, and economic efficiency in Nile tilapia, Oreochromis niloticus. Two hundred twenty-five fingerlings (32.38 ± 0.05 g/fish) were distributed into five treatments with five dietary inclusion levels of SDBH: 0, 2.5, 5, 7.5, and 10% for a ten-week feeding period. Dietary inclusion of SDBH linearly increased the final body weight (FBW), total weight gain (TWG), specific growth rate (SGR), and protein efficiency ratio (PER). Additionally, a linear decrease in feed conversion ratio (FCR) and daily feed intake relative to the daily BW was reported in the highest inclusion levels (7.5 and 10%). Dietary inclusion of SDBH was associated with a significant increase in the intestinal villous height (VH), villous width (VW), villous height: crypt depth ratio (VH: CD), and muscle coat thickness (MCT), with the highest values reported in SDBH7.5 group. Increased serum growth hormone levels and decreased serum leptin hormone levels were also reported by increasing the SDBH level. The serum glucose level was decreased in the SDBH7.5 and SDBH10 groups. The digestive enzymes' activity (amylase and protease) was increased by increasing the SDBH inclusion level. An up-regulation in the expression of peptide and amino acid transporters, IGF-1, and down-regulation of myostatin was reported in the SDBH2.5 to SDBH7.5 groups. Spleen sections showed more lymphoid elements, especially in the SDBH2.5 and SDBH7.5 groups. The SDBH inclusion increased the serum lysozyme activity, nitric oxide (NO), and complement 3 (C3) levels, with the highest values recorded in the SDBH5 group. The phagocytic % and the phagocytic index were increased by increasing the SDBH inclusion %. The expressions of immune-related genes (transforming growth factor-beta (TGF-β), Toll-like receptor 2 (TLR2), and interleukin 10 (IL10)) were up-regulated by SDBH inclusion with the highest expression in the SDBH5 group. Economically, the feed costs and feed costs/kg gain were linearly decreased in the SDBH7.5 and SDBH10 diets. In conclusion, spray-dried bovine hemoglobin powder could be used as a protein source for up to 10% of the diets of Nile tilapia for better growth and immune status of fish.
Collapse
Affiliation(s)
- Shimaa A. Amer
- Department of Nutrition & Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
- Correspondence: (S.A.A.); (R.E.I.)
| | - Mahmoud Farahat
- Department of Nutrition & Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Samar A. Abdo
- Biochemistry Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Elsayed M. Younis
- Department of Zoology, College of Science, King Saudi University, Riyadh 11451, Saudi Arabia
| | | | - Rehab Reda
- Animal Wealth Development Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Sozan A. Ali
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Simon J. Davies
- School of Science and Engineering, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Rowida E. Ibrahim
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
- Correspondence: (S.A.A.); (R.E.I.)
| |
Collapse
|
185
|
Lucarini V, Melaiu O, D’Amico S, Pastorino F, Tempora P, Scarsella M, Pezzullo M, De Ninno A, D’Oria V, Cilli M, Emionite L, Infante P, Di Marcotullio L, De Ioris MA, Barillari G, Alaggio R, Businaro L, Ponzoni M, Locatelli F, Fruci D. Combined mitoxantrone and anti-TGFβ treatment with PD-1 blockade enhances antitumor immunity by remodelling the tumor immune landscape in neuroblastoma. J Exp Clin Cancer Res 2022; 41:326. [PMID: 36397148 PMCID: PMC9670422 DOI: 10.1186/s13046-022-02525-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background Poor infiltration of functioning T cells renders tumors unresponsive to checkpoint-blocking immunotherapies. Here, we identified a combinatorial in situ immunomodulation strategy based on the administration of selected immunogenic drugs and immunotherapy to sensitize poorly T-cell-infiltrated neuroblastoma (NB) to the host antitumor immune response. Methods 975A2 and 9464D NB cell lines derived from spontaneous tumors of TH-MYCN transgenic mice were employed to study drug combinations able of enhancing the antitumor immune response using in vivo and ex vivo approaches. Migration of immune cells towards drug-treated murine-derived organotypic tumor spheroids (MDOTS) were assessed by microfluidic devices. Activation status of immune cells co-cultured with drug-treated MDOTS was evaluated by flow cytometry analysis. The effect of drug treatment on the immune content of subcutaneous or orthotopic tumors was comprehensively analyzed by flow-cytometry, immunohistochemistry and multiplex immunofluorescence. The chemokine array assay was used to detect soluble factors released into the tumor microenvironment. Patient-derived organotypic tumor spheroids (PDOTS) were generated from human NB specimens. Migration and activation status of autologous immune cells to drug-treated PDOTS were performed. Results We found that treatment with low-doses of mitoxantrone (MTX) recalled immune cells and promoted CD8+ T and NK cell activation in MDOTS when combined with TGFβ and PD-1 blockade. This combined immunotherapy strategy curbed NB growth resulting in the enrichment of a variety of both lymphoid and myeloid immune cells, especially intratumoral dendritic cells (DC) and IFNγ- and granzyme B-expressing CD8+ T cells and NK cells. A concomitant production of inflammatory chemokines involved in remodelling the tumor immune landscape was also detected. Interestingly, this treatment induced immune cell recruitment against PDOTS and activation of CD8+ T cells and NK cells. Conclusions Combined treatment with low-dose of MTX and anti-TGFβ treatment with PD-1 blockade improves antitumor immunity by remodelling the tumor immune landscape and overcoming the immunosuppressive microenvironment of aggressive NB. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02525-9.
Collapse
|
186
|
Samer S, Thomas Y, Araínga M, Carter C, Shirreff LM, Arif MS, Avita JM, Frank I, McRaven MD, Thuruthiyil CT, Heybeli VB, Anderson MR, Owen B, Gaisin A, Bose D, Simons LM, Hultquist JF, Arthos J, Cicala C, Sereti I, Santangelo PJ, Lorenzo-Redondo R, Hope TJ, Villinger FJ, Martinelli E. Blockade of TGF-β signaling reactivates HIV-1/SIV reservoirs and immune responses in vivo. JCI Insight 2022; 7:e162290. [PMID: 36125890 PMCID: PMC9675457 DOI: 10.1172/jci.insight.162290] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022] Open
Abstract
TGF-β plays a critical role in maintaining immune cells in a resting state by inhibiting cell activation and proliferation. Resting HIV-1 target cells represent the main cellular reservoir after long-term antiretroviral therapy (ART). We hypothesized that releasing cells from TGF-β-driven signaling would promote latency reversal. To test our hypothesis, we compared HIV-1 latency models with and without TGF-β and a TGF-β type 1 receptor inhibitor, galunisertib. We tested the effect of galunisertib in SIV-infected, ART-treated macaques by monitoring SIV-env expression via PET/CT using the 64Cu-DOTA-F(ab')2 p7D3 probe, along with plasma and tissue viral loads (VLs). Exogenous TGF-β reduced HIV-1 reactivation in U1 and ACH-2 models. Galunisertib increased HIV-1 latency reversal ex vivo and in PBMCs from HIV-1-infected, ART-treated, aviremic donors. In vivo, oral galunisertib promoted increased total standardized uptake values in PET/CT images in gut and lymph nodes of 5 out of 7 aviremic, long-term ART-treated, SIV-infected macaques. This increase correlated with an increase in SIV RNA in the gut. Two of the 7 animals also exhibited increases in plasma VLs. Higher anti-SIV T cell responses and antibody titers were detected after galunisertib treatment. In summary, our data suggest that blocking TGF-β signaling simultaneously increases retroviral reactivation events and enhances anti-SIV immune responses.
Collapse
Affiliation(s)
- Sadia Samer
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yanique Thomas
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mariluz Araínga
- New Iberia Research Center (NIRC), University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Crystal Carter
- New Iberia Research Center (NIRC), University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Lisa M. Shirreff
- New Iberia Research Center (NIRC), University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Muhammad S. Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Juan M. Avita
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ines Frank
- Center for Biomedical Research, Population Council, New York, New York, USA
| | - Michael D. McRaven
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Christopher T. Thuruthiyil
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Veli B. Heybeli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Meegan R. Anderson
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Benjamin Owen
- Integrated Molecular Structure Education and Research (IMSERC), Northwestern University, Evanston, Illinois, USA
| | - Arsen Gaisin
- Integrated Molecular Structure Education and Research (IMSERC), Northwestern University, Evanston, Illinois, USA
| | - Deepanwita Bose
- New Iberia Research Center (NIRC), University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Lacy M. Simons
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health Northwestern University, Chicago, Illinois, USA
| | - Judd F. Hultquist
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health Northwestern University, Chicago, Illinois, USA
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Irini Sereti
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Philip J. Santangelo
- WH Coulter Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health Northwestern University, Chicago, Illinois, USA
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francois J. Villinger
- New Iberia Research Center (NIRC), University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
187
|
Mirjačić Martinović K, Vuletić A, Mališić E, Srdić-Rajić T, Tišma Miletić N, Babović N, Jurišić V. Increased circulating TGF-β1 is associated with impairment in NK cell effector functions in metastatic melanoma patients. Growth Factors 2022; 40:231-239. [PMID: 36129407 DOI: 10.1080/08977194.2022.2124915] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Transforming growth factor beta (TGF-β) plays a complex role in carcinogenesis. In 30 melanoma patients and 20 healthy controls (HC) we analysed functional and phenotypic characteristics of NK cells by Flow cytometry, gene expression of TGF-β1 in peripheral blood mononuclear cells by qPCR and serum and supernatant level of free TGF-β1 by ELISA. Melanoma patients had significantly higher serum level of circulatingTGF-β1 compared to HC, especially those with metastasis into the central nervous system (subclass M1d) and high LDH serum values. Melanoma patients compared to HC had significantly higher level of TGF-β1 gene in PBMC. TGF-β1 serum values negatively correlate with NK cell activity analysed by CD107a (degranulation marker), IFN-γ, NKG2D, and NKp46 in patients. Study shows the association of high level of TGF-β1 with NK cell inhibition in patients represents the main mechanism of tumour immune evasion. Targeting TGF-β may become an important cancer treatment for improving antitumor immunity.
Collapse
Affiliation(s)
| | - Ana Vuletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Emina Mališić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Nevena Tišma Miletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Nada Babović
- Department of Medical Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
188
|
Lähteenmäki Taalas T, Järvelä L, Niinikoski H, Huurre A, Harila‐Saari A. Inflammatory biomarkers after an exercise intervention in childhood acute lymphoblastic leukemia survivors. EJHAEM 2022; 3:1188-1200. [PMID: 36467791 PMCID: PMC9713025 DOI: 10.1002/jha2.588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 06/17/2023]
Abstract
Cancer survivors show increased risk for non-communicable diseases and chronic low-grade inflammation characterizes the development of such diseases. We investigated inflammatory plasma protein profiles of survivors of childhood acute lymphoblastic leukemia (ALL) in comparison to healthy controls and after an intervention with a home-based exercise program. Survivors of childhood ALL aged 16-30 years (n = 21) with a median age at diagnosis 4.9 (1.6-12.9) years and a median time of 15.9 years from diagnosis, and sex- and age-matched healthy controls (n = 21) were studied. Stored plasma samples were analyzed with Olink's 92-protein-wide Inflammation panel in 21 ALL long-term survivors at baseline, after a previous 16-week home-based exercise intervention (n = 17) and in 21 age- and sex-matched controls at baseline. Protein expression levels were compared between the groups. Inflammatory protein levels did not differ between the survivors and controls at baseline. Significantly reduced levels after the intervention were found in 11 proteins related to either vascular inflammation, insulin resistance, or both: tumor necrosis factor superfamily member 14 (TNFSF14), oncostatin M (OSM), monocyte chemoattractant protein 1 (MCP-1), MCP-2, fibroblast growth factor 21 (FGF-21), chemokine (C-C motif) ligand 4 (CCL4), transforming growth factor alpha (TGF-α), tumor necrosis factor-related apoptosis-inducing ligand 10 (TRAIL), adenosine deaminase (ADA), chemokine (C-X-C motif) ligand 6 (CXCL6), and latency-associated peptide transforming growth factor beta 1 (LAP TGF-β1). The ALL survivors were not significantly more affected by inflammation than controls at baseline. The survivors' 16-week exercise intervention led to significant reduction in inflammatory protein levels. Physical exercise should be promoted for survivors of childhood cancer.
Collapse
Affiliation(s)
- Tuomas Lähteenmäki Taalas
- University of TurkuTurkuFinland
- Department of Women's and Children's HealthUppsala UniversityUppsalaSweden
| | - Liisa Järvelä
- University of TurkuTurkuFinland
- Department of Pediatrics and Adolescent MedicineTurku University HospitalTurkuFinland
| | - Harri Niinikoski
- University of TurkuTurkuFinland
- Department of Pediatrics and Adolescent MedicineTurku University HospitalTurkuFinland
| | - Anu Huurre
- University of TurkuTurkuFinland
- Department of Pediatrics and Adolescent MedicineTurku University HospitalTurkuFinland
| | - Arja Harila‐Saari
- Department of Women's and Children's HealthUppsala UniversityUppsalaSweden
| |
Collapse
|
189
|
Understanding fibrosis pathogenesis via modeling macrophage-fibroblast interplay in immune-metabolic context. Nat Commun 2022; 13:6499. [PMID: 36310236 PMCID: PMC9618579 DOI: 10.1038/s41467-022-34241-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/18/2022] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is a progressive biological condition, leading to organ dysfunction in various clinical settings. Although fibroblasts and macrophages are known as key cellular players for fibrosis development, a comprehensive functional model that considers their interaction in the metabolic/immunologic context of fibrotic tissue has not been set up. Here we show, by transcriptome-based mathematical modeling in an in vitro system that represents macrophage-fibroblast interplay and reflects the functional effects of inflammation, hypoxia and the adaptive immune context, that irreversible fibrosis development is associated with specific combinations of metabolic and inflammatory cues. The in vitro signatures are in good alignment with transcriptomic profiles generated on laser captured glomeruli and cortical tubule-interstitial area, isolated from human transplanted kidneys with advanced stages of glomerulosclerosis and interstitial fibrosis/tubular atrophy, two clinically relevant conditions associated with organ failure in renal allografts. The model we describe here is validated on tissue based quantitative immune-phenotyping of biopsies from transplanted kidneys, demonstrating its feasibility. We conclude that the combination of in vitro and in silico modeling represents a powerful systems medicine approach to dissect fibrosis pathogenesis, applicable to specific pathological conditions, and develop coordinated targeted approaches.
Collapse
|
190
|
Kuppa SS, Kim HK, Kang JY, Lee SC, Seon JK. Role of Mesenchymal Stem Cells and Their Paracrine Mediators in Macrophage Polarization: An Approach to Reduce Inflammation in Osteoarthritis. Int J Mol Sci 2022; 23:13016. [PMID: 36361805 PMCID: PMC9658630 DOI: 10.3390/ijms232113016] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA) is a low-grade inflammatory disorder of the joints that causes deterioration of the cartilage, bone remodeling, formation of osteophytes, meniscal damage, and synovial inflammation (synovitis). The synovium is the primary site of inflammation in OA and is frequently characterized by hyperplasia of the synovial lining and infiltration of inflammatory cells, primarily macrophages. Macrophages play a crucial role in the early inflammatory response through the production of several inflammatory cytokines, chemokines, growth factors, and proteinases. These pro-inflammatory mediators are activators of numerous signaling pathways that trigger other cytokines to further recruit more macrophages to the joint, ultimately leading to pain and disease progression. Very few therapeutic alternatives are available for treating inflammation in OA due to the condition's low self-healing capacity and the lack of clear diagnostic biomarkers. In this review, we opted to explore the immunomodulatory properties of mesenchymal stem cells (MSCs) and their paracrine mediators-dependent as a therapeutic intervention for OA, with a primary focus on the practicality of polarizing macrophages as suppression of M1 macrophages and enhancement of M2 macrophages can significantly reduce OA symptoms.
Collapse
Affiliation(s)
- Sree Samanvitha Kuppa
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Hyung Keun Kim
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Ju Yeon Kang
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Seok Cheol Lee
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Jong Keun Seon
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| |
Collapse
|
191
|
Yan W, Zhang Y, Cao J, Yan H. TGF-β2 levels in the aqueous humor are elevated in the second eye of high myopia within two weeks after sequential cataract surgery. Sci Rep 2022; 12:17974. [PMID: 36289352 PMCID: PMC9606117 DOI: 10.1038/s41598-022-22746-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 10/19/2022] [Indexed: 01/24/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is the critical regulator of physiological and pathological conditions in lens. The TGF-β signaling pathway is closely associated with high myopia patients. Thirty eyes from fifteen patients with high myopia who received sequential cataract surgery were enrolled in this prospective study. Ten cataract patients with non-myopia were chosen as a control group. Aqueous humor (AH) samples were used to detect the levels of TGF-β1, TGF-β2, and TGF-β3 in both groups. Compared with the non-myopic cataracts patient group, the highly myopic cataracts group had a significantly higher TGF-β2 (P = 0.019). Besides, the level of TGF-β2 of the second eye was significantly higher than that in the first eye in high myopia cataract patients group (P = 0.037). And TGF-β1 showed significant differences with age and axial length of high myopia cataract patients. Therefore, TGF-β2 may contribute to the development of high myopia and cataract surgery increased the expression of TGF-β2.
Collapse
Affiliation(s)
- Weijia Yan
- grid.440588.50000 0001 0307 1240Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated Northwestern Polytechnical University, Xi’an, 710004 Shaanxi China ,grid.7700.00000 0001 2190 4373Department of Ophthalmology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Yaping Zhang
- grid.440588.50000 0001 0307 1240Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated Northwestern Polytechnical University, Xi’an, 710004 Shaanxi China
| | - Junguo Cao
- grid.7700.00000 0001 2190 4373Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hong Yan
- grid.440588.50000 0001 0307 1240Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated Northwestern Polytechnical University, Xi’an, 710004 Shaanxi China
| |
Collapse
|
192
|
Transforming Growth Factor-β Concerning Malarial Infection and Severity: A Systematic Review and Meta-Analysis. Trop Med Infect Dis 2022; 7:tropicalmed7100299. [PMID: 36288040 PMCID: PMC9612234 DOI: 10.3390/tropicalmed7100299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/07/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is important in the pathophysiology of malaria, but its role in acute and severe malaria is largely unknown. As a result, this study used a meta-analysis approach to investigate the difference in TGF-β levels between several groups of malaria patients and healthy controls. The systematic review protocol was registered at PROSPERO (ID: CRD42022318864). From inception to 7 March 2022, studies that reported TGF-β levels in patients with uncomplicated and healthy controls and patients with severe and uncomplicated malaria were searched in PubMed, Scopus and Embase. The assessment of the quality of the included studies was conducted according to the Strengthening the Reporting of Observational Studies in Epidemiology guidelines. Qualitative and quantitative syntheses were performed to narratively describe and quantitatively pool the mean difference (MD) in TGF-β levels between uncomplicated malaria and healthy controls, and between severe and uncomplicated malaria, using a random-effects model. A total of 1027 relevant articles were identified, and 13 studies were included for syntheses. The meta-analysis results show 233 patients with uncomplicated malaria and 239 healthy controls. Patients with uncomplicated malaria (233 cases) had lower mean TGF-β levels than healthy controls (239 cases; p < 0.01, pooled MD = −14.72 pg/mL, 95% confidence interval (95% CI) = −20.46 to 8.99 pg/mL, I2 = 98.82%, seven studies). The meta-analysis found no difference in mean TGF-β levels between patients with severe malaria (367 cases) and patients with uncomplicated malaria (180 cases; p = 0.11, pooled MD = −6.07 pg/mL, 95% CI = −13.48 to 1.35 pg/mL, I2 = 97.73%, six studies). The meta-analysis demonstrated decreased TGF-β levels in patients with uncomplicated malaria compared to healthy controls. In addition, no difference in TGF-β levels was found between patients with severe and uncomplicated malaria. More research is needed to determine whether TGF-β levels could be a candidate marker for malarial infection or disease severity.
Collapse
|
193
|
Mihoubi E, Amroun H, Bouldjennet F, Azzouz M, Touil-Boukoffa C, Raache R, Attal N. Polymorphismes 869C> T et 915 G>C du TGF-β dans la rétinopathie du diabète de type 1 chez la population algérienne. J Fr Ophtalmol 2022; 45:908-914. [DOI: 10.1016/j.jfo.2022.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 10/18/2022]
|
194
|
Chauché C, Rasid O, Donachie A, McManus CM, Löser S, Campion T, Richards J, Smyth DJ, McSorley HJ, Maizels RM. Suppression of airway allergic eosinophilia by Hp-TGM, a helminth mimic of TGF-β. Immunology 2022; 167:197-211. [PMID: 35758054 PMCID: PMC9885513 DOI: 10.1111/imm.13528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/20/2022] [Indexed: 02/02/2023] Open
Abstract
Type 2-high asthma is a chronic inflammatory disease of the airways which is increasingly prevalent in countries where helminth parasite infections are rare, and characterized by T helper 2 (Th2)-dependent accumulation of eosinophils in the lungs. Regulatory cytokines such as TGF-β can restrain inflammatory reactions, dampen allergic Th2 responses, and control eosinophil activation. The murine helminth parasite Heligmosomoides polygyrus releases a TGF-β mimic (Hp-TGM) that replicates the biological and functional properties of TGF-β despite bearing no structural similarity to the mammalian protein. Here, we investigated if Hp-TGM could alleviate allergic airway inflammation in mice exposed to Alternaria alternata allergen, house dust mite (HDM) extract or alum-adjuvanted ovalbumin protein (OVA). Intranasal administration of Hp-TGM during Alternaria exposure sharply reduced airway and lung tissue eosinophilia along with bronchoalveolar lavage fluid IL-5 and lung IL-33 cytokine levels at 24 h. The protective effect of Hp-TGM on airway eosinophilia was also obtained in the longer T-cell mediated models of HDM or OVA sensitisation with significant inhibition of eotaxin-1, IL-4 and IL-13 responses depending on the model and time-point. Hp-TGM was also protective when administered parenterally either when given at the time of allergic sensitisation or during airway allergen challenge. This project has taken the first steps in identifying the role of Hp-TGM in allergic asthma and highlighted its ability to control lung inflammation and allergic pathology. Future research will investigate the mode of action of Hp-TGM against airway allergic eosinophilia, and further explore its potential to be developed as a biotherapeutic in allergic asthma.
Collapse
Affiliation(s)
- Caroline Chauché
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK,Centre for Inflammation ResearchUniversity of Edinburgh, Queen's Medical Research InstituteEdinburghUK
| | - Orhan Rasid
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| | - Anne‐Marie Donachie
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| | - Caitlin M. McManus
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| | - Stephan Löser
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| | - Tiffany Campion
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| | - Josh Richards
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK,Division of Cell Signalling and ImmunologySchool of Life Sciences, Wellcome Trust Building, University of DundeeDundeeUK
| | - Danielle J. Smyth
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK,Division of Cell Signalling and ImmunologySchool of Life Sciences, Wellcome Trust Building, University of DundeeDundeeUK
| | - Henry J. McSorley
- Division of Cell Signalling and ImmunologySchool of Life Sciences, Wellcome Trust Building, University of DundeeDundeeUK
| | - Rick M. Maizels
- Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| |
Collapse
|
195
|
Shukla N, Naik A, Moryani K, Soni M, Shah J, Dave H. TGF-β at the crossroads of multiple prognosis in breast cancer, and beyond. Life Sci 2022; 310:121011. [PMID: 36179816 DOI: 10.1016/j.lfs.2022.121011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 10/25/2022]
Abstract
Transforming growth factor β (TGF-β), a pluripotent cytokine and a multifunctional growth factor has a crucial role in varied biological mechanisms like invasion, migration, epithelial-mesenchymal transition, apoptosis, wound healing, and immunosuppression. Moreover, it also has an imperative role both in normal mammary gland development as well as breast carcinogenesis. TGF-β has shown to have a paradoxical role in breast carcinogenesis, by transitioning from a growth inhibitor to a growth promoter with the disease advancement. The inter-communication and crosstalk of TGF-β with different signaling pathways has strengthened the likelihood to explore it as a comprehensive biomarker. In the last two decades, TGF-β has been studied extensively and has been found to be a promising biomarker for early detection, disease monitoring, treatment selection, and tumor progression making it beneficial for disease management. In this review, we focus on the signaling pathways and biological activities of the TGF-β family in breast cancer pathogenesis and its role as a circulatory and independent biomarker for breast cancer progression and metastasis. Moreover, this review highlights TGF-β as a drug target, and the underlying mechanisms through which it is involved in tumorigenesis that will aid in the development of varied therapies targeting the different stages of breast cancer.
Collapse
Affiliation(s)
- Nirali Shukla
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Ankit Naik
- Ahmedabad University, Ahmedabad, Gujarat 390009, India
| | - Kamlesh Moryani
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Molisha Soni
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Jigna Shah
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Heena Dave
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
196
|
Chen L, Huang H, Zheng X, Li Y, Chen J, Tan B, Liu Y, Sun R, Xu B, Yang M, Li B, Wu C, Lu B, Jiang J. IL1R2 increases regulatory T cell population in the tumor microenvironment by enhancing MHC-II expression on cancer-associated fibroblasts. J Immunother Cancer 2022. [PMCID: PMC9438093 DOI: 10.1136/jitc-2022-004585] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Regulatory T cells (Treg) are an integral part of the tumor immune tolerance. Carcinoma-associated fibroblasts (CAFs) is a pivotal driver for accumulation of Treg cells in the tumor microenvironment (TME). The molecular nature underpinning Treg cells and CAFs coupling needs to be further defined. Methods The Il1r2flox/floxFoxp3Cre mice were generated to establish the conditional knock-out of Il1r2 in Foxp3+ Tregs in vivo. Using the MC38 tumor model, we evaluated the antitumor efficacy of immune checkpoint inhibitors (ICIs) and further analyzed the immune profiling of the TME by multicolor flow cytometry. Single-cell RNA sequencing of the whole tumor tissues, TCR repertoire analysis of sorted CD3+ TILs were also performed. Results We showed that IL1 receptor 2 (IL1R2), a decoy receptor that neutralizes IL1, was highly expressed in Treg cells in the TME. In addition, we found that Il1r1 was largely expressed in the CAFs, suggesting IL1R2 plays a role in modulating crosstalk between Tregs and CAFs. We further demonstrated that Il1r2 deficiency in Treg cells led to greater antitumor efficacy of ICI, decreased Tregs and increased CD8+ T cells in the TME, as well as reduced levels of T cell dysfunction. Mechanistically, we showed that IL1 inhibited major histocompatibility complex class II (MHC-II) expression on fibroblasts and Treg-specific Il1r2 deletion led to a decrease in genes associated with MHC-II antigen presentation in CAFs. Conclusions Our study established a critical role of IL1 signaling in inhibiting Treg-mediated tumor immune suppression through downregulating MHC-II antigen presentation in CAFs.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Hao Huang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Yuan Li
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bo Tan
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Yingting Liu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Runzi Sun
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Min Yang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Department of Nephrology, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changping Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| |
Collapse
|
197
|
Medina JM, Abbas MN, Bensaoud C, Hackenberg M, Kotsyfakis M. Bioinformatic Analysis of Ixodes ricinus Long Non-Coding RNAs Predicts Their Binding Ability of Host miRNAs. Int J Mol Sci 2022; 23:ijms23179761. [PMID: 36077158 PMCID: PMC9456184 DOI: 10.3390/ijms23179761] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Ixodes ricinus ticks are distributed across Europe and are a vector of tick-borne diseases. Although I. ricinus transcriptome studies have focused exclusively on protein coding genes, the last decade witnessed a strong increase in long non-coding RNA (lncRNA) research and characterization. Here, we report for the first time an exhaustive analysis of these non-coding molecules in I. ricinus based on 131 RNA-seq datasets from three different BioProjects. Using this data, we obtained a consensus set of lncRNAs and showed that lncRNA expression is stable among different studies. While the length distribution of lncRNAs from the individual data sets is biased toward short length values, implying the existence of technical artefacts, the consensus lncRNAs show a more homogeneous distribution emphasizing the importance to incorporate data from different sources to generate a solid reference set of lncRNAs. KEGG enrichment analysis of host miRNAs putatively targeting lncRNAs upregulated upon feeding showed that these miRNAs are involved in several relevant functions for the tick-host interaction. The possibility that at least some tick lncRNAs act as host miRNA sponges was further explored by identifying lncRNAs with many target regions for a given host miRNA or sets of host miRNAs that consistently target lncRNAs together. Overall, our findings suggest that lncRNAs that may act as sponges have diverse biological roles related to the tick–host interaction in different tissues.
Collapse
Affiliation(s)
- José María Medina
- Departamentode Genética, Facultad de Ciencias, Universidad de Granada, Campus de Fuentenueva s/n, 18071 Granada, Spain
- Laboratorio de Bioinformática, Centro de Investigación Biomédica, PTS, Instituto de Biotecnología, Avda. del Conocimiento s/n, 18016 Granada, Spain
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400715, China
| | - Chaima Bensaoud
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Michael Hackenberg
- Departamentode Genética, Facultad de Ciencias, Universidad de Granada, Campus de Fuentenueva s/n, 18071 Granada, Spain
- Laboratorio de Bioinformática, Centro de Investigación Biomédica, PTS, Instituto de Biotecnología, Avda. del Conocimiento s/n, 18016 Granada, Spain
- Correspondence: (M.H.); (M.K.)
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
- Correspondence: (M.H.); (M.K.)
| |
Collapse
|
198
|
Hsieh MCW, Wang WT, Lin CY, Kuo YR, Lee SS, Hou MF, Wu YC. Stem Cell-Based Therapeutic Strategies in Diabetic Wound Healing. Biomedicines 2022; 10:biomedicines10092085. [PMID: 36140185 PMCID: PMC9495374 DOI: 10.3390/biomedicines10092085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/10/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Impaired wound healing and especially the “all-too-common” occurrence of associated diabetic foot ulcers (DFU) are becoming an increasingly urgent and deteriorating healthcare issue, which drastically impact the quality of life and further heighten the risks of infection and amputation in patients with diabetes mellitus. Amongst the multifactorial wound healing determinants, glycemic dysregulation has been identified to be the primary casual factor of poor wound healing. Unfortunately, current therapeutic modalities merely serve as moderate symptomatic relieves but often fail to completely restore the wound site to its pre-injury state and prevent further recurrence. Stem cell-based therapeutics have been employed for its promising potential to address the root of the problem as they not only exhibit the capacity for self-renewal and differentiation towards multiple lineages, but also have been disclosed to participate in mediating variant growth factors and cytokines. Herein we review the current literatures on the therapeutic benefits of using various kinds of stem cells, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and adipose-derived stem cells (ASCs) in diabetic wound healing by searching on the PubMed® Database for publications. This study shall serve as an overview of the current body of research with particular focus on autologous ASCs and the laboratory expandable iPSCs in hope of shedding more light on this attractive therapy so as to elevate the efficacy of wound healing that is almost always compromised in diabetic patients.
Collapse
Affiliation(s)
- Meng-Chien Willie Hsieh
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Plastic Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Wei-Ting Wang
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chuang-Yu Lin
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yur-Ren Kuo
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Su-Shin Lee
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Division of Breast Oncology and Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Yi-Chia Wu
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Plastic Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Division of Breast Oncology and Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 7675)
| |
Collapse
|
199
|
Martin NM, Griffin DE. Effect of IL-10 Deficiency on TGFβ Expression during Fatal Alphavirus Encephalomyelitis in C57Bl/6 Mice. Viruses 2022; 14:1791. [PMID: 36016413 PMCID: PMC9416572 DOI: 10.3390/v14081791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
Sindbis virus (SINV) causes viral encephalitis in mice with strain-dependent virulence. Fatal encephalomyelitis in C57Bl/6 mice infected with a neuroadapted strain of SINV (NSV) is an immunopathogenic process that involves Th17 cells modulated by the regulatory cytokine IL-10. To further characterize the pathogenic immune response to NSV, we analyzed the regulation of transforming growth factor (TGF)-b in both wild-type (WT) and IL-10-deficient mice. NSV infection upregulated the expression of TGFb1 and TGFb3 in the central nervous system (CNS). In the absence of IL-10, levels of brain Tgfb1 mRNA and brain and spinal cord mature active TGFβ1 and TGFβ3 proteins were higher than in WT mice. Compared to WT mice, IL-10-deficient mice had more TGFβ1-expressing type 3 innate lymphoid cells (ILC3s) and CD4+ T cells infiltrating the CNS, but similar numbers in the cervical lymph nodes. Expression of glycoprotein A repetitions predominant protein (GARP) that binds pro-TGFb on the surface of regulatory T cells was decreased on CNS cells from IL-10-deficient mice. Higher CNS TGFb was accompanied by more expression of TGFbRII receptor, activation of SMAD transcription factors, increased PCKα mRNA, and more RORγt-positive and IL-17A-expressing cells. These results suggest a compensatory role for TGFβ in the absence of IL-10 that fosters Th17-related immunopathology and more rapid death after NSV infection.
Collapse
Affiliation(s)
| | - Diane E. Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
200
|
Arketamine, a new rapid-acting antidepressant: A historical review and future directions. Neuropharmacology 2022; 218:109219. [PMID: 35977629 DOI: 10.1016/j.neuropharm.2022.109219] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 12/16/2022]
Abstract
The N-methyl-d-aspartate receptor (NMDAR) antagonist (R,S)-ketamine causes rapid onset and sustained antidepressant actions in treatment-resistant patients with major depressive disorder (MDD) and other psychiatric disorders, such as bipolar disorder and post-traumatic stress disorder. (R,S)-ketamine is a racemic mixture consisting of (R)-ketamine (or arketamine) and (S)-ketamine (or esketamine), with (S)-enantiomer having greater affinity for the NMDAR. In 2019, an esketamine nasal spray by Johnson & Johnson was approved in the USA and Europe for treatment-resistant depression. In contrast, an increasing number of preclinical studies show that arketamine has greater potency and longer-lasting antidepressant-like effects than esketamine in rodents, despite the lower binding affinity of arketamine for the NMDAR. Importantly, the side effects, i.e., psychotomimetic and dissociative effects and abuse liability, of arketamine are less than those of (R,S)-ketamine and esketamine in animals and humans. An open-label study demonstrated the rapid and sustained antidepressant effects of arketamine in treatment-resistant patients with MDD. A phase 2 clinical trial of arketamine in treatment-resistant patients with MDD is underway. This study was designed to review the brief history of the novel antidepressant arketamine, the molecular mechanisms underlying its antidepressant actions, and future directions.
Collapse
|