1
|
Papanikolaou M, Paul J, Nattkemper LA, Kirsner RS, Yosipovitch G. Prevalence and Mechanisms of Itch in Chronic Wounds: A Narrative Review. J Clin Med 2025; 14:2877. [PMID: 40363908 PMCID: PMC12072805 DOI: 10.3390/jcm14092877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Itch is a commonly experienced problem by individuals with chronic wounds and greatly compromises their quality of life. Scratching can further hinder the wound healing process. Despite this being a clinically recognized issue, our knowledge of its exact prevalence in chronic wounds of different types and the molecular mechanisms driving it is limited. The multifactorial nature of wound itch makes its characterization particularly challenging. The present review is based on a thorough PubMed search, and it aims to provide an overview of existing evidence on the epidemiology, impact, and pathophysiology of wound itch, along with general recommendations on its management. Importantly, our work highlights the merit of screening chronic wound patients for associated pruritus and incorporating anti-itch measures in mainstream wound care.
Collapse
Affiliation(s)
- Marieta Papanikolaou
- Department of Dermatology, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (L.A.N.); (G.Y.)
| | - Julia Paul
- School of Nursing, Oakland University, Rochester, MI 48309, USA;
| | - Leigh A. Nattkemper
- Department of Dermatology, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (L.A.N.); (G.Y.)
| | - Robert S. Kirsner
- Dr. Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Gil Yosipovitch
- Department of Dermatology, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (L.A.N.); (G.Y.)
| |
Collapse
|
2
|
Li C, Wei Y, Lei J. Quantitative cancer-immunity cycle modeling for predicting disease progression in advanced metastatic colorectal cancer. NPJ Syst Biol Appl 2025; 11:33. [PMID: 40221414 PMCID: PMC11993626 DOI: 10.1038/s41540-025-00513-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Patients with advanced metastatic colorectal cancer (mCRC) typically exhibit significant interindividual differences in treatment responses and face poor survival outcomes. To systematically analyze the heterogeneous tumor progression and recurrence observed in advanced mCRC patients, we developed a quantitative cancer-immunity cycle (QCIC) model. The QCIC model employs differential equations to capture the biological mechanisms underlying the cancer-immunity cycle and predicts tumor evolution dynamics under various treatment strategies through stochastic computational methods. We introduce the treatment response index (TRI) to quantify disease progression in virtual clinical trials and the death probability function (DPF) to estimate overall survival. Additionally, we investigate the impact of predictive biomarkers on survival prognosis in advanced mCRC patients, identifying tumor-infiltrating CD8+ cytotoxic T lymphocytes (CTLs) as key predictors of disease progression and the tumor-infiltrating CD4+ Th1/Treg ratio as a significant determinant of survival outcomes. This study presents an approach that bridges the gap between diverse clinical data sources and the generation of virtual patient cohorts, providing valuable insights into interindividual treatment variability and survival forecasting in mCRC patients.
Collapse
Affiliation(s)
- Chenghang Li
- School of Mathematical Sciences, Tiangong University, Tianjin, 300387, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China.
| | - Jinzhi Lei
- School of Mathematical Sciences, Tiangong University, Tianjin, 300387, China.
- Center for Applied Mathematics, Tiangong University, Tianjin, 300387, China.
| |
Collapse
|
3
|
Zhang G, Yao Q, Long C, Yi P, Song J, Wu L, Wan W, Rao X, Lin Y, Wei G, Ying J, Hua F. Infiltration by monocytes of the central nervous system and its role in multiple sclerosis: reflections on therapeutic strategies. Neural Regen Res 2025; 20:779-793. [PMID: 38886942 PMCID: PMC11433895 DOI: 10.4103/nrr.nrr-d-23-01508] [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: 09/06/2023] [Revised: 12/12/2023] [Accepted: 02/18/2024] [Indexed: 06/20/2024] Open
Abstract
Mononuclear macrophage infiltration in the central nervous system is a prominent feature of neuroinflammation. Recent studies on the pathogenesis and progression of multiple sclerosis have highlighted the multiple roles of mononuclear macrophages in the neuroinflammatory process. Monocytes play a significant role in neuroinflammation, and managing neuroinflammation by manipulating peripheral monocytes stands out as an effective strategy for the treatment of multiple sclerosis, leading to improved patient outcomes. This review outlines the steps involved in the entry of myeloid monocytes into the central nervous system that are targets for effective intervention: the activation of bone marrow hematopoiesis, migration of monocytes in the blood, and penetration of the blood-brain barrier by monocytes. Finally, we summarize the different monocyte subpopulations and their effects on the central nervous system based on phenotypic differences. As activated microglia resemble monocyte-derived macrophages, it is important to accurately identify the role of monocyte-derived macrophages in disease. Depending on the roles played by monocyte-derived macrophages at different stages of the disease, several of these processes can be interrupted to limit neuroinflammation and improve patient prognosis. Here, we discuss possible strategies to target monocytes in neurological diseases, focusing on three key aspects of monocyte infiltration into the central nervous system, to provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangyong Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Qing Yao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Chubing Long
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jiali Song
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Luojia Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Wei Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
4
|
Miura M, Kitaura H, Ohori F, Narita K, Ren J, Noguchi T, Marahleh A, Ma J, Lin A, Fan Z, Mizoguchi I. Role of CXCL10 released from osteocytes in response to TNF-α stimulation on osteoclasts. Sci Rep 2025; 15:3040. [PMID: 39856227 PMCID: PMC11760356 DOI: 10.1038/s41598-025-87092-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) is a significant cytokine that regulates bone resorption under inflammatory conditions. However, its mechanism of action in osteocytes remains unclear. In this study, highly purified osteocytes were isolated from dentin matrix protein 1 (DMP1)-Topaz mice using cell sorter. RNA sequencing (RNA-seq) revealed that TNF-α stimulation increased C-X-C motif chemokine ligand 10 (CXCL10) gene expression in osteocytes. Although CXCL10 did not affect osteoclast differentiation in vitro, it enhanced the migration of osteoclast precursors. Additionally, in the transwell co-culture system, TNF-α induced the migration of osteoclast precursors. However, this effect was attenuated by a CXCL10-neutralizing antibody. In vivo, mice were administered supracalvarial injections of TNF-α with or without the CXCL10-neutralizing antibody for 5 days. The percentage of CXCL10-positive osteocytes increased after TNF-α administration. Additionally, osteoclast formation and bone resorption were assessed. CXCL10-neutralizing antibody-treated calvariae exhibited a significantly lower number of osteoclasts and bone resorption than those treated with TNF-α alone. These results indicated that TNF-α-induced CXCL10, which affects the migration of osteocyte-derived osteoclast precursors, may enhance TNF-α-triggered osteoclast formation and bone resorption in vivo.
Collapse
Affiliation(s)
- Mariko Miura
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Hideki Kitaura
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Fumitoshi Ohori
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Kohei Narita
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Jiayi Ren
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Takahiro Noguchi
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Aseel Marahleh
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- Creative Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3 Aramaki Aza Aoba, Aoba-ku, Sendai, 980-8578, Japan
| | - Jinghan Ma
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Angyi Lin
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Ziqiu Fan
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Itaru Mizoguchi
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
5
|
Jones HN, Davenport BN, Wilson RL. Maternal-fetal interfaces transcriptome changes associated with placental insufficiency and a novel gene therapy intervention. Physiol Genomics 2025; 57:8-15. [PMID: 39374081 PMCID: PMC11918312 DOI: 10.1152/physiolgenomics.00131.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
The etiology of fetal growth restriction (FGR) is multifactorial, although many cases often involve placental insufficiency. Placental insufficiency is associated with inadequate trophoblast invasion, resulting in high resistance to blood flow, decreased availability of nutrients, and increased hypoxia. We have developed a nonviral, polymer-based nanoparticle that facilitates delivery and transient gene expression of human insulin-like 1 growth factor (hIGF1) in placental trophoblast for the treatment of placenta insufficiency and FGR. Using the established guinea pig maternal nutrient restriction (MNR) model of placental insufficiency and FGR, the aim of the study was to identify novel pathways in the subplacenta/decidua that provide insight into the underlying mechanism driving placental insufficiency and may be corrected with hIGF1 nanoparticle treatment. Pregnant guinea pigs underwent ultrasound-guided sham or hIGF1 nanoparticle treatment at midpregnancy, and subplacenta/decidua tissue was collected 5 days later. Transcriptome analysis was performed using RNA Sequencing on the Illumina platform. The MNR subplacenta/decidua demonstrated fewer maternal spiral arteries lined by trophoblast, shallower trophoblast invasion, and downregulation of genelists involved in the regulation of cell migration. hIGF1 nanoparticle treatment resulted in marked changes to transporter activity in the MNR + hIGF1 subplacenta/decidua when compared with sham MNR. Under normal growth conditions however, hIGF1 nanoparticle treatment decreased genelists enriched for kinase signaling pathways and increased genelists enriched for proteolysis, indicative of homeostasis. Overall, this study identified changes to the subplacenta/decidua transcriptome that likely result in inadequate trophoblast invasion and increases our understanding of pathways that hIGF1 nanoparticle treatment acts on to restore or maintain appropriate placenta function.NEW & NOTEWORTHY Placental insufficiency at midpregnancy, established through moderate maternal nutrient restriction, is characterized with fewer maternal spiral arteries lined by trophoblast, shallower trophoblast invasion, and downregulation of genelists involved in the regulation of cell migration. Treatment of placenta insufficiency with a hIGF1 nanoparticle results in marked changes to transporter activity and increases our mechanistic understanding of how therapies designed to improve fetal growth may impact the placenta.
Collapse
Affiliation(s)
- Helen N Jones
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Baylea N Davenport
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Rebecca L Wilson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
6
|
Roozitalab MR, Prekete N, Allen M, Grose RP, Louise Jones J. The Microenvironment in DCIS and Its Role in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:211-235. [PMID: 39821028 DOI: 10.1007/978-3-031-70875-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Ductal carcinoma in situ (DCIS) accounts for ~20% of all breast cancer diagnoses but whilst known to be a precursor of invasive breast cancer (IBC), evidence suggests only one in six patients will ever progress. A key challenge is to distinguish between those lesions that will progress and those that will remain indolent. Molecular analyses of neoplastic epithelial cells have not identified consistent differences between lesions that progressed and those that did not, and this has focused attention on the tumour microenvironment (ME).The DCIS ME is unique, complex and dynamic. Myoepithelial cells form the wall of the ductal-lobular tree and exhibit broad tumour suppressor functions. However, in DCIS they acquire phenotypic changes that bestow them with tumour promoter properties, an important evolution since they act as the primary barrier for invasion. Changes in the peri-ductal stromal environment also arise in DCIS, including transformation of fibroblasts into cancer-associated fibroblasts (CAFs). CAFs orchestrate other changes in the stroma, including the physical structure of the extracellular matrix (ECM) through altered protein synthesis, as well as release of a plethora of factors including proteases, cytokines and chemokines that remodel the ECM. CAFs can also modulate the immune ME as well as impact on tumour cell signalling pathways. The heterogeneity of CAFs, including recognition of anti-tumourigenic populations, is becoming evident, as well as heterogeneity of immune cells and the interplay between these and the adipocyte and vascular compartments. Knowledge of the impact of these changes is more advanced in IBC but evidence is starting to accumulate for a role in DCIS. Detailed in vitro, in vivo and tissue studies focusing on the interplay between DCIS epithelial cells and the ME should help to define features that can better predict DCIS behaviour.
Collapse
Affiliation(s)
- Mohammad Reza Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Niki Prekete
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Michael Allen
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK.
| |
Collapse
|
7
|
Shuai XU, Dandan F, Liang XU. Study on the changes and significance of CXCL10 level in serum of isolated polymyalgia rheumatica. Clin Rheumatol 2024; 43:3993-3998. [PMID: 39476056 PMCID: PMC11582166 DOI: 10.1007/s10067-024-07209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/02/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE To investigate the significance of CXC chemokine ligand 10 (CXCL10) in the pathogenesis of isolated polymyalgia rheumatica (PMR). METHODS The serum of six PMR patients diagnosed and treated at the First Affiliated Hospital of Wannan Medical College from September 2019 to December 2020 before treatment and after remission was collected, and the serum of six active rheumatoid arthritis (RA) patients and six healthy medical checkups were also collected, and protein microarray technology was used to detect 24 cytokines, including IL-6, IL-4, CXCL10, CXCL8, and CXCL2. Subsequently, serum was collected from other 28 patients with active PMR, 26 patients with PMR in remission, 24 patients with active RA, and 24 healthy medical checkups who were diagnosed and treated at the First Affiliated Hospital of Wannan Medical College from January 2021 to July 2023, and the enzyme-linked immunosorbent assay (ELISA) was used to validate and compare the levels of CXCL10 in each group and analyze the correlation between the levels of serum CXCL10 and the parameters of the clinical activities of PMR. RESULTS Protein microarray screening revealed significant differences in CXCL10 before and after PMR treatment, and ELISA validation revealed that peripheral serum CXCL10 levels were significantly higher in the PMR-active group than in the remission group (P < 0.001), and also significantly higher than in the RA-active group (P = 0.003) and in the healthy control group (P < 0.001); correlation analysis showed a significant positive correlation between serum CXCL10 levels and serum ferritin in PMR patients (r = 0.450, P = 0.024). In the ROC curve for distinguishing PMR and RA, the area under the curve is 0.741, sensitivity = 0.643, and specificity = 0.792. CONCLUSION CXCL10 may play a role in the pathogenesis of isolated PMR and its level might contribute to the differential diagnosis of PMR and RA. Key Points • The concentration of CXCL10 was higher in peripheral blood of isolated PMR patients. • CXCL10 is a potential diagnostic biomarker for isolated PMR patients. • The level of CXCL10 might contribute to the differential diagnosis of PMR and RA.
Collapse
Affiliation(s)
- X U Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Feng Dandan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - X U Liang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.
| |
Collapse
|
8
|
Mačionienė E, Simanavičius M, Vitkauskaitė M, Vickienė A, Staučė R, Vinikovas A, Miglinas M. Urinary Chemokines CXCL9 and CXCL10 Are Non-Invasive Biomarkers of Kidney Transplant Rejection. Ann Transplant 2024; 29:e944762. [PMID: 39402819 PMCID: PMC11490196 DOI: 10.12659/aot.944762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Rejection is the main cause of kidney allograft failure, and kidney biopsy is the criterion standard method to diagnose it. However, non-invasive techniques to detect kidney transplant rejection are necessary. This study aimed to evaluate urinary chemokines CXCL9 and CXCL10 as potential biomarkers of kidney transplant rejection and to analyze chemokine association with allograft prognosis. MATERIAL AND METHODS We collected 117 urine samples from kidney transplant recipients undergoing allograft biopsy. CXCL9 and CXCL10 levels were measured by ELISA and the ratio to urine creatinine was calculated. Histology and other clinical data were collected from medical records. RESULTS The diagnostic performance of urinary CXCL9/cre in discriminating rejection from all other histological groups showed an ROC AUC value of 0.728 (95% CI 0.632-0.824, p<0.001), and a cut-off value 0.11 ng/mmol had the best sensitivity (76.9%) and specificity (73.1%). The ability of CXCL10/cre to discriminate transplant rejection from all other histological groups had ROC AUC value 0.73 (95% CI 0.63-0.84, P<0.001), the cut-off value 0.42 ng/mmol with best sensitivity (71.4%) and specificity (84.6%). CXCL9 and CXCL10 levels were also increased in patients with polyoma BK virus, recurrent AA amyloidosis, and thrombotic microangiopathy. Patients with higher CXCL9/cre (≥0.11 ng/mmol) and CXCL10/cre (≥0.42 ng/mmol) levels were at increased risk of transplant progression to ESRD (HR 3.25, 95% CI=1.27-8.36, P=0.01), irrespective of serum creatinine at the time of biopsy. CONCLUSIONS Urinary CXCL9/cre and CXCL10/cre were able to distinguished between patients with transplant rejection and those without rejection. High levels of urinary CXCL9/cre and CXCL10/cre were associated with worse allograft survival.
Collapse
Affiliation(s)
- Ernesta Mačionienė
- Institute of Clinical Medicine, Gastroenterology, Nephro-Urology and Surgery Clinic, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Martynas Simanavičius
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Monika Vitkauskaitė
- Institute of Clinical Medicine, Gastroenterology, Nephro-Urology and Surgery Clinic, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Alvita Vickienė
- Institute of Clinical Medicine, Gastroenterology, Nephro-Urology and Surgery Clinic, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Roberta Staučė
- Nephrology Center, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Artūras Vinikovas
- Nephrology Center, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Marius Miglinas
- Institute of Clinical Medicine, Gastroenterology, Nephro-Urology and Surgery Clinic, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
9
|
Than NG, Romero R, Fitzgerald W, Gudicha DW, Gomez-Lopez N, Posta M, Zhou F, Bhatti G, Meyyazhagan A, Awonuga AO, Chaiworapongsa T, Matthies D, Bryant DR, Erez O, Margolis L, Tarca AL. Proteomic Profiles of Maternal Plasma Extracellular Vesicles for Prediction of Preeclampsia. Am J Reprod Immunol 2024; 92:e13928. [PMID: 39347565 DOI: 10.1111/aji.13928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 10/01/2024] Open
Abstract
PROBLEM Preeclampsia is a heterogeneous syndrome of diverse etiologies and molecular pathways leading to distinct clinical subtypes. Herein, we aimed to characterize the extracellular vesicle (EV)-associated and soluble fractions of the maternal plasma proteome in patients with preeclampsia and to assess their value for disease prediction. METHOD OF STUDY This case-control study included 24 women with term preeclampsia, 23 women with preterm preeclampsia, and 94 healthy pregnant controls. Blood samples were collected from cases on average 7 weeks before the diagnosis of preeclampsia and were matched to control samples. Soluble and EV fractions were separated from maternal plasma; EVs were confirmed by cryo-EM, NanoSight, and flow cytometry; and 82 proteins were analyzed with bead-based, multiplexed immunoassays. Quantile regression analysis and random forest models were implemented to evaluate protein concentration differences and their predictive accuracy. Preeclampsia subgroups defined by molecular profiles were identified by hierarchical cluster analysis. Significance was set at p < 0.05 or false discovery rate-adjusted q < 0.1. RESULTS In preterm preeclampsia, PlGF, PTX3, and VEGFR-1 displayed differential abundance in both soluble and EV fractions, whereas angiogenin, CD40L, endoglin, galectin-1, IL-27, CCL19, and TIMP1 were changed only in the soluble fraction (q < 0.1). The direction of changes in the EV fraction was consistent with that in the soluble fraction for nine proteins. In term preeclampsia, CCL3 had increased abundance in both fractions (q < 0.1). The combined EV and soluble fraction proteomic profiles predicted preterm and term preeclampsia with an AUC of 78% (95% CI, 66%-90%) and 68% (95% CI, 56%-80%), respectively. Three clusters of preeclampsia featuring distinct clinical characteristics and placental pathology were identified based on combined protein data. CONCLUSIONS Our findings reveal distinct alterations of the maternal EV-associated and soluble plasma proteome in preterm and term preeclampsia and identify molecular subgroups of patients with distinct clinical and placental histopathologic features.
Collapse
Affiliation(s)
- Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Dereje W Gudicha
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology & Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University Doctoral School, Budapest, Hungary
| | - Fei Zhou
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Gaurav Bhatti
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Arun Meyyazhagan
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Awoniyi O Awonuga
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Doreen Matthies
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - David R Bryant
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Department of Obstetrics and Gynecology, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Leonid Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Adi L Tarca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| |
Collapse
|
10
|
Noack D, van Haperen A, van den Hout MCGN, Marshall EM, Koutstaal RW, van Duinen V, Bauer L, van Zonneveld AJ, van IJcken WFJ, Koopmans MPG, Rockx B. A three-dimensional vessel-on-chip model to study Puumala orthohantavirus pathogenesis. LAB ON A CHIP 2024. [PMID: 39292495 DOI: 10.1039/d4lc00543k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Puumala orthohantavirus (PUUV) infection in humans can result in hemorrhagic fever with renal syndrome. Endothelial cells (ECs) are primarily infected with increased vascular permeability as a central aspect of pathogenesis. Historically, most studies included ECs cultured under static two-dimensional (2D) conditions, thereby not recapitulating the physiological environment due to their lack of flow and inherent pro-inflammatory state. Here, we present a high-throughput model for culturing primary human umbilical vein ECs in 3D vessels-on-chip in which we compared host responses of these ECs to those of static 2D-cultured ECs on a transcriptional level. The phenotype of ECs in vessels-on-chip more closely resembled the in vivo situation due to higher similarity in expression of genes encoding described markers for disease severity and coagulopathy, including IDO1, LGALS3BP, IL6 and PLAT, and more diverse endothelial-leukocyte interactions in the context of PUUV infection. In these vessels-on-chip, PUUV infection did not directly increase vascular permeability, but increased monocyte adhesion. This platform can be used for studying pathogenesis and assessment of possible therapeutics for other endotheliotropic viruses even in high biocontainment facilities.
Collapse
Affiliation(s)
- Danny Noack
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Anouk van Haperen
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Mirjam C G N van den Hout
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Biomics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eleanor M Marshall
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Rosanne W Koutstaal
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Vincent van Duinen
- Department of Internal Medicine, Division of Nephrology and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Lisa Bauer
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Anton Jan van Zonneveld
- Department of Internal Medicine, Division of Nephrology and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Biomics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| |
Collapse
|
11
|
Košćak Lukač J, Baronica KB, Šućur A, Sremec J, Tomasović S, Baronica R, Kelava T, Grčević D, Kovačić N. Serum Concentrations of Chemokines CCL20, CXCL8 and CXCL10 in Relapsing-Remitting Multiple Sclerosis and Their Association with Presence of Antibodies against Epstein-Barr Virus. Int J Mol Sci 2024; 25:8064. [PMID: 39125633 PMCID: PMC11311471 DOI: 10.3390/ijms25158064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Epstein-Barr virus (EBV) infection and various chemokines, including CCL20, CXCL8 and CXCL10 are considered to participate in the pathogenesis of multiple sclerosis (MS), and several studies point to a direct regulatory effect of EBV on the expression of these chemokines. In our study we hypothesized that serum concentrations of CCL20, CXCL8 and CXCL0 are induced in patients with relapsing-remitting MS (RRMS) in comparison to healthy individuals, and that they are associated with EBV infection. Serum concentrations of CXCL8 and CXCL10 were lower in RRMS patients in relapse in comparison to healthy controls. Although potential effects of corticosteroid therapy introduced in a subgroup of RRMS patients prior to sampling were excluded by subgroup comparison, this possibility has to be considered while interpreting the results. We found an inverse association between serum concentrations of CXCL8 and anti-Epstein-Barr Virus Nuclear Antigen (EBNA) IgG and decreased expression of CXCL8 in peripheral blood mononuclear cells (PBMC) in relapse compared to remission. Lower serum concentrations of CXCL8 and CXCL10 in RRMS patients and decreased peripheral production of CXCL8 in relapse may indicate compensatory anti-inflammatory counter-regulation in MS.
Collapse
Affiliation(s)
- Jelena Košćak Lukač
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
| | - Koraljka Bačić Baronica
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
- Neurology Clinic, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31 000 Osijek, Croatia
| | - Alan Šućur
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Šalata 3, 10 000 Zagreb, Croatia
| | - Josip Sremec
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
| | - Sanja Tomasović
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
- Neurology Clinic, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31 000 Osijek, Croatia
| | - Robert Baronica
- Department of Anesthesiology, Reanimatology, Intensive Medicine and Pain Therapy, University Hospital Centre Zagreb, Kišpatićeva 12, 10 000 Zagreb, Croatia;
| | - Tomislav Kelava
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Šalata 3, 10 000 Zagreb, Croatia
| | - Danka Grčević
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Šalata 3, 10 000 Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Anatomy, University of Zagreb School of Medicine, Šalata 11, 10 000 Zagreb, Croatia
| |
Collapse
|
12
|
Akasaka K, Amano M, Nakamura T, Nishizawa T, Yamakawa H, Sato S, Aoki A, Shima K, Matsushima H, Takada T. Cytokine profiles associated with disease severity and prognosis of autoimmune pulmonary alveolar proteinosis. Respir Investig 2024; 62:610-616. [PMID: 38705133 DOI: 10.1016/j.resinv.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Pulmonary alveolar proteinosis (PAP) is characterized by an abnormal accumulation of surfactants in the alveoli. Most cases are classified as autoimmune PAP (APAP) because they are associated with autoantibodies against granulocyte-macrophage colony-stimulating factor (GM-CSF). However, GM-CSF autoantibody levels are unlikely to correlate with the disease severity or prognosis of APAP. METHODS We collected clinical records and measured 38 serum cytokine concentrations for consecutive patients with APAP. After exclusion of 21 cytokines because of undetectable levels, 17 cytokine levels were compared between low and high disease severity scores (DSSs). We also compared whole lung lavage (WLL)-free survival with cut-off values defined by receiver operating characteristic (ROC) curves of cytokine levels and WLL administration at 11 months. RESULTS Nineteen patients with APAP were enrolled in the study. Five were classified as DSS 1 or 2, while the others were classified as DSS 4 or 5. Comparison between DSS 1-2 and 4-5 revealed that the concentrations of IP-10 and GRO increased in the latter groups (p < 0.05). Fifteen patients underwent WLL. Comparison between those who underwent WLL within 11 months and the others showed that IP-10 and TNF-α were tended to be elevated in the former group (p = 0.082 and 0.057, respectively). The cut-off values of IP-10, 308.8 pg/mL and TNF-α, 19.1 pg/mL, defined by the ROC curves, significantly separated WLL-free survivals with log-rank analyses (p = 0.005). CONCLUSIONS The concentrations of IP-10 and GRO may reflect the DSSs of APAP. A combination of IP-10 and TNF-α levels could be a biomarker to predict WLL-free survival.
Collapse
Affiliation(s)
- Keiichi Akasaka
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan; Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, 4132 Urasa, Minami Uonuma-shi, Niigata, 949-7302, Japan
| | - Masako Amano
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan
| | - Tomohiko Nakamura
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan
| | - Tomotaka Nishizawa
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan
| | - Hideaki Yamakawa
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan
| | - Shintaro Sato
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan
| | - Ami Aoki
- Division of Respiratory Medicine, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata, 951-8520, Japan
| | - Kenjiro Shima
- Division of Respiratory Medicine, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata, 951-8520, Japan
| | - Hidekazu Matsushima
- Department of Respiratory Medicine, Saitama Red Cross Hospital, 1-5 Shintoshin, Chuo-ku, Saitama, 330-8553, Japan
| | - Toshinori Takada
- Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, 4132 Urasa, Minami Uonuma-shi, Niigata, 949-7302, Japan; Division of Respiratory Medicine, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata, 951-8520, Japan.
| |
Collapse
|
13
|
Neyton LPA, Patel RK, Sarma A, Willmore A, Haller SC, Kangelaris KN, Eckalbar WL, Erle DJ, Krummel MF, Hendrickson CM, Woodruff PG, Langelier CR, Calfee CS, Fragiadakis GK. Distinct pulmonary and systemic effects of dexamethasone in severe COVID-19. Nat Commun 2024; 15:5483. [PMID: 38942804 PMCID: PMC11213873 DOI: 10.1038/s41467-024-49756-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. Here we perform bulk and single-cell RNA sequencing of samples from the lower respiratory tract and blood, and assess plasma cytokine profiling to study the effects of dexamethasone on both systemic and pulmonary immune cell compartments. In blood samples, dexamethasone is associated with decreased expression of genes associated with T cell activation, including TNFSFR4 and IL21R. We also identify decreased expression of several immune pathways, including major histocompatibility complex-II signaling, selectin P ligand signaling, and T cell recruitment by intercellular adhesion molecule and integrin activation, suggesting these are potential mechanisms of the therapeutic benefit of steroids in COVID-19. We identify additional compartment- and cell- specific differences in the effect of dexamethasone that are reproducible in publicly available datasets, including steroid-resistant interferon pathway expression in the respiratory tract, which may be additional therapeutic targets. In summary, we demonstrate compartment-specific effects of dexamethasone in critically ill COVID-19 patients, providing mechanistic insights with potential therapeutic relevance. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.
Collapse
Affiliation(s)
- Lucile P A Neyton
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Ravi K Patel
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Andrew Willmore
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Sidney C Haller
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | | | - Walter L Eckalbar
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - David J Erle
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Lung Biology Center, University of California, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Carolyn M Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, CA, USA
| | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA.
- Division of Rheumatology, University of California, San Francisco, CA, USA.
| |
Collapse
|
14
|
Zhou S, Li W, Lv R, Zhang M, Liu W. Neuroprotective effects and mechanisms of action of artemisinin in retinal ganglion cells in a mouse model of traumatic optic neuropathy. Heliyon 2024; 10:e31378. [PMID: 38828288 PMCID: PMC11140598 DOI: 10.1016/j.heliyon.2024.e31378] [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: 12/21/2023] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction Traumatic optic neuropathy is known to be a critical condition that can cause blindness; however, the specific mechanism underlying optic nerve injury is unclear. Recent studies have reported that artemisinin, considered vital in malaria treatment, can also be used to treat neurodegenerative diseases; however, its precise role and mechanism of action remain unknown. Therefore, in this study, we aimed to investigate the impact and probable mechanism of action of artemisinin in retinal ganglion cells (RGCs) in a mouse model of traumatic optic neuropathy induced by optic nerve crush (ONC). Methods ONC was induced in the left eye of mice by short-term clamping of the optic nerve; oral artemisinin was administered daily. The neuroprotective effect of the drug was assessed using Tuj-1 staining in RGCs. In addition, the inflammatory response and the expression levels of phosphorylated tau protein and tau oligomers were observed using RT-qPCR, TUNEL assay, and fluorescence staining to investigate the underlying mechanisms. Results Artemisinin increased the survival rate of RGCs 14 days after ONC. Artemisinin significantly reduced the levels of inflammatory factors such as CXCL10, CXCR3, and IL-1β in the retina and decreased the apoptosis of RGCs. Moreover, downregulation of the phosphorylation of tau proteins and the expression of tau oligomers were observed after artemisinin treatment. Conclusion Our results suggest that artemisinin can increase the survival rate of RGCs after ONC and reduce their apoptosis. This effect may be achieved by inhibiting the inflammatory response it triggers and downregulating tau protein phosphorylation and tau oligomer expression. These findings suggest the potential application of artemisinin as a therapeutic agent for neuropathy.
Collapse
Affiliation(s)
- Shirui Zhou
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wangzi Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruohan Lv
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - MingChang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Jones HN, Davenport BN, Wilson RL. Maternal-fetal interfaces transcriptome changes associated with placental insufficiency and a novel gene therapy intervention. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597595. [PMID: 38895421 PMCID: PMC11185673 DOI: 10.1101/2024.06.05.597595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The etiology of fetal growth restriction (FGR) is multifactorial, although many cases often involve placental insufficiency. Placental insufficiency is associated with inadequate trophoblast invasion resulting in high resistance to blood flow, decreased availability of nutrients, and increased hypoxia. We have developed a non-viral, polymer-based nanoparticle that facilitates delivery and transient gene expression of human insulin-like 1 growth factor ( hIGF1 ) in placental trophoblast for the treatment of placenta insufficiency and FGR. Using the established guinea pig maternal nutrient restriction (MNR) model of placental insufficiency and FGR, the aim of the study was to identify novel pathways in the sub-placenta/decidua that provide insight into the underlying mechanism driving placental insufficiency, and may be corrected with hIGF1 nanoparticle treatment. Pregnant guinea pigs underwent ultrasound-guided sham or hIGF1 nanoparticle treatment at mid-pregnancy, and sub-placenta/decidua tissue was collected 5 days later. Transcriptome analysis was performed using RNA Sequencing on the Illumina platform. The MNR sub-placenta/decidua demonstrated fewer maternal spiral arteries lined by trophoblast, shallower trophoblast invasion and downregulation of genelists involved in the regulation of cell migration. hIGF1 nanoparticle treatment resulted in marked changes to transporter activity in the MNR + hIGF1 sub-placenta/decidua when compared to sham MNR. Under normal growth conditions however, hIGF1 nanoparticle treatment decreased genelists enriched for kinase signaling pathways and increased genelists enriched for proteolysis indicative of homeostasis. Overall, this study identified changes to the sub-placenta/decidua transcriptome that likely result in inadequate trophoblast invasion and increases our understanding of pathways that hIGF1 nanoparticle treatment acts on in order to restore or maintain appropriate placenta function.
Collapse
|
16
|
Hailstock T, Dai C, Aquino J, Walker KE, Chick S, Manirarora JN, Suresh R, Patil V, Renukaradhya GJ, Sullivan YB, LaBresh J, Lunney JK. Production and characterization of anti-porcine CXCL10 monoclonal antibodies. Cytokine 2024; 174:156449. [PMID: 38141459 DOI: 10.1016/j.cyto.2023.156449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/25/2023]
Abstract
Research on C-X-C motif chemokine ligand 10 (CXCL10) has been widely reported for humans and select animal species, yet immune reagents are limited for pig chemokines. Our goal is to provide veterinary immunologists and the biomedical community with new commercial immune reagents and standardized assays. Recombinant porcine CXCL10 (rPoCXCL10) protein was produced by yeast expression and used to generate a panel of α CXCL10 monoclonal antibodies (mAbs). All mAbs were assessed for cross-inhibition and reactivity to orthologous yeast expressed CXCL10 proteins. Characterization of a panel of nine α PoCXCL10 mAbs identified six distinct antigenic determinants. A sensitive quantitative sandwich ELISA was developed with anti-PoCXCL10-1.6 and -1.9 mAb; reactivity was verified with both rPoCXCL10 and native PoCXCL10, detected in supernatants of peripheral blood mononuclear cells stimulated with rPoIFNγ or PMA/Ionomycin. Immunostaining of in vitro rPoIFNγ stimulated pig spleen and blood cells verified CXCL10 + cells as CD3-CD4-CD172+, with occasional CD3-CD4 + CD172 + subsets. Comparison studies determined that α PoCXCL10-1.4 mAb was the ideal mAb clone for intracellular staining, whereas with α PoCXCL10-1.1 and -1.2 mAbs were best for immunohistochemistry analyses. These techniques and tools will be useful for evaluating swine immune development, responses to infectious diseases and vaccines, as well as for improving utility of pigs as an important biomedical model.
Collapse
Affiliation(s)
- Taylor Hailstock
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Chaohui Dai
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA; Yangzhou University, Yangzhou, Jiangsu, China
| | - Jovan Aquino
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Kristen E Walker
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Shannon Chick
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Jean N Manirarora
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Raksha Suresh
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH, USA
| | - Veerupaxagouda Patil
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH, USA
| | - Gourapura J Renukaradhya
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH, USA
| | | | | | - Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA.
| |
Collapse
|
17
|
Manuel G, Coleman M, Orvis AS, Munson J, Li A, Kapur RP, Li M, Li E, Armistead B, Rajagopal L, Adams Waldorf KM. Spatial profiling of the placental chorioamniotic membranes reveals upregulation of immune checkpoint proteins during Group B Streptococcus infection in a nonhuman primate model. Front Cell Infect Microbiol 2024; 13:1299644. [PMID: 38239507 PMCID: PMC10794649 DOI: 10.3389/fcimb.2023.1299644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
Background Preterm birth is a leading cause of neonatal mortality, which is often complicated by intrauterine infection and inflammation. We have established a nonhuman primate model of Group B Streptococcus (GBS, Streptococcus agalactiae) infection-associated preterm birth. Immune checkpoints are modulators of the immune response by activating or suppressing leukocyte function and are understudied in preterm birth. The objective of this study was to spatially profile changes in immune protein expression at the maternal-fetal interface during a GBS infection with a focus on immune checkpoints. Methods Twelve nonhuman primates (pigtail macaques, Macaca nemestrina) received a choriodecidual inoculation of either: 1) 1-5 X 108 colony forming units (CFU) of hyperhemolytic/hypervirulent GBS (GBSΔcovR, N=4); 2) an isogenic/nonpigmented strain (GBS ΔcovRΔcylE, N=4); or, 3) saline (N=4). A Cesarean section was performed at preterm labor or 3 days after GBS infection or 7 days after saline inoculation. Nanostring GeoMx® Digital Spatial Profiling technology was used to segment protein expression within the amnion, chorion, and maternal decidua at the inoculation site using an immuno-oncology panel targeting 56 immunoproteins enriched in stimulatory and inhibitory immune checkpoint proteins or their protein ligands. Statistical analysis included R studio, Kruskal-Wallis, Pearson and Spearman tests. Results Both inhibitory and stimulatory immune checkpoint proteins were significantly upregulated within the chorioamniotic membranes and decidua (VISTA, LAG3, PD-1, CD40, GITR), as well as their ligands (PD-L1, PD-L2, CD40L; all p<0.05). Immunostaining for VISTA revealed positive (VISTA+) cells, predominantly in the chorion and decidua. There were strong correlations between VISTA and amniotic fluid concentrations of IL-1β, IL-6, IL-8, and TNF-α (all p<0.05), as well as maternal placental histopathology scores (p<0.05). Conclusion Differential regulation of multiple immune checkpoint proteins in the decidua at the site of a GBS infection indicates a major perturbation in immunologic homeostasis that could benefit the host by restricting immune-driven pathologies or the pathogen by limiting immune surveillance. Protein expression of VISTA, an inhibitory immune checkpoint, was upregulated in the chorion and decidua after GBS infection. Investigating the impact of innate immune cell expression of inhibitory immune checkpoints may reveal new insights into placental host-pathogen interactions at the maternal-fetal interface.
Collapse
Affiliation(s)
- Gygeria Manuel
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Morehouse School of Medicine, Atlanta, GA, United States
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Austyn S. Orvis
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Jeff Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Raj P. Kapur
- Department of Laboratory Medicine and Pathology, Seattle Children’s Hospital and University of Washington, Seattle, WA, United States
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- School of Medicine, University of Washington, Seattle, WA, United States
| | - Edmunda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
18
|
Meyhöfer S, Steffen A, Plötze-Martin K, Marquardt JU, Meyhöfer SM, Bruchhage KL, Pries R. Obesity-related Plasma CXCL10 Drives CX3CR1-dependent Monocytic Secretion of Macrophage Migration Inhibitory Factor. Immunohorizons 2024; 8:19-28. [PMID: 38175171 PMCID: PMC10835669 DOI: 10.4049/immunohorizons.2300114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Obesity is characterized by excessive body fat accumulation and comorbidities such as diabetes mellitus, cardiovascular disease, and obstructive sleep apnea syndrome (OSAS). Both obesity and OSAS are associated with immune disturbance, alterations of systemic inflammatory mediators, and immune cell recruitment to metabolic tissues. Chemokine CXCL10 is an important regulator of proinflammatory immune responses and is significantly increased in patients with severe obesity. This research project aims to investigate the impact of CXCL10 on human monocytes in patients with obesity. We studied the distribution of the CD14/CD16 monocyte subsets as well as their CX3CR1 expression patterns in whole-blood measurements from 92 patients with obesity and/or OSAS with regard to plasma CXCL10 values and individual clinical parameters. Furthermore, cytokine secretion by THP-1 monocytes in response to CXCL10 was analyzed. Data revealed significantly elevated plasma CXCL10 in patients with obesity with an additive effect of OSAS. CXCL10 was found to drive monocytic secretion of macrophage migration inhibitory factor via receptor protein CX3CR1, which significantly correlated with the individual body mass index. Our data show, for the first time, to our knowledge, that CX3CR1 is involved in alternative CXCL10 signaling in human monocytes in obesity-related inflammation. Obesity is a multifactorial disease, and further investigations regarding the complex interplay between obesity-related inflammatory mediators and systemic immune balances will help to better understand and improve the individual situation of our patients.
Collapse
Affiliation(s)
- Svenja Meyhöfer
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
- Institute for Endocrinology & Diabetes, Department of Internal Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Armin Steffen
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Jens-Uwe Marquardt
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Sebastian M. Meyhöfer
- Institute for Endocrinology & Diabetes, Department of Internal Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| |
Collapse
|
19
|
Nagura Y, Suzuki T, Matsuura K, Ogawa S, Kawamura H, Kuno K, Fujiwara K, Nojiri S, Nagaoka K, Iio E, Watanabe T, Kataoka H, Tanaka Y. Serum inducible protein 10 kDa/C-X-C motif chemokine 10 levels predict regression of M2BPGi-based liver fibrosis after hepatitis C virus eradication by direct-acting antiviral agents. Hepatol Res 2024; 54:32-42. [PMID: 37638483 DOI: 10.1111/hepr.13962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
AIM It is desirable to identify predictors of regression of liver fibrosis after achieving sustained virological response by anti-hepatitis C virus (anti-HCV) therapy. We retrospectively investigated the serum interferon-γ inducible protein 10 kDa (IP-10) level as a predictive indicator of regression of liver fibrosis after successful hepatitis C virus eradication by direct-acting antiviral agents (DAAs) therapy. METHODS The study participants were recruited from a historical cohort of 116 chronically hepatitis C virus-infected patients who had achieved sustained virological response by DAAs therapy and whose serum Mac-2 binding protein glycosylation isomer (M2BPGi) levels at baseline (before DAAs therapy) were ≥2.0 cut-off index. We defined patients with M2BPGi levels <1.76 and ≥1.76 cut-off index at 2 years after the end of treatment (EOT) as the regression (n = 71) and non-regression (n = 45) groups, respectively. RESULTS Multivariate analyses revealed that the albumin-bilirubin score at baseline, and albumin-bilirubin score, Fibrosis-4 index at 24 weeks after the EOT, and serum IP-10 change from baseline to 24 weeks after the EOT (IP-10 change) were significantly associated with regression of M2BPGi-based liver fibrosis. In addition, IP-10 change was significantly associated with regression of M2BPGi-based liver fibrosis by a multivariate analysis, even when the serum M2BPGi levels were aligned by propensity score matching and in patients with advanced M2BPGi-based liver fibrosis: M2BPGi levels ≥3.3 cut-off index at baseline. CONCLUSIONS Serum IP-10 change from baseline to 24 weeks after the EOT is a feasible predictor of regression of M2BPGi-based liver fibrosis after achieving sustained virological response with DAA therapy.
Collapse
Affiliation(s)
- Yoshihito Nagura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Gastroenterology, Kasugai Municipal Hospital, Kasugai, Japan
| | - Takanori Suzuki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kentaro Matsuura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shintaro Ogawa
- Department of Virology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hayato Kawamura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kayoko Kuno
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kei Fujiwara
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shunsuke Nojiri
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Katsuya Nagaoka
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Etsuko Iio
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takehisa Watanabe
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
20
|
Dufva O, Gandolfi S, Huuhtanen J, Dashevsky O, Duàn H, Saeed K, Klievink J, Nygren P, Bouhlal J, Lahtela J, Näätänen A, Ghimire BR, Hannunen T, Ellonen P, Lähteenmäki H, Rumm P, Theodoropoulos J, Laajala E, Härkönen J, Pölönen P, Heinäniemi M, Hollmén M, Yamano S, Shirasaki R, Barbie DA, Roth JA, Romee R, Sheffer M, Lähdesmäki H, Lee DA, De Matos Simoes R, Kankainen M, Mitsiades CS, Mustjoki S. Single-cell functional genomics reveals determinants of sensitivity and resistance to natural killer cells in blood cancers. Immunity 2023; 56:2816-2835.e13. [PMID: 38091953 DOI: 10.1016/j.immuni.2023.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/19/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Cancer cells can evade natural killer (NK) cell activity, thereby limiting anti-tumor immunity. To reveal genetic determinants of susceptibility to NK cell activity, we examined interacting NK cells and blood cancer cells using single-cell and genome-scale functional genomics screens. Interaction of NK and cancer cells induced distinct activation and type I interferon (IFN) states in both cell types depending on the cancer cell lineage and molecular phenotype, ranging from more sensitive myeloid to less sensitive B-lymphoid cancers. CRISPR screens in cancer cells uncovered genes regulating sensitivity and resistance to NK cell-mediated killing, including adhesion-related glycoproteins, protein fucosylation genes, and transcriptional regulators, in addition to confirming the importance of antigen presentation and death receptor signaling pathways. CRISPR screens with a single-cell transcriptomic readout provided insight into underlying mechanisms, including regulation of IFN-γ signaling in cancer cells and NK cell activation states. Our findings highlight the diversity of mechanisms influencing NK cell susceptibility across different cancers and provide a resource for NK cell-based therapies.
Collapse
Affiliation(s)
- Olli Dufva
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland
| | - Sara Gandolfi
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland; Department of Computer Science, Aalto University, 02150 Espoo, Finland
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hanna Duàn
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland
| | - Khalid Saeed
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland
| | - Petra Nygren
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland
| | - Jonas Bouhlal
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland
| | - Jenni Lahtela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Anna Näätänen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Bishwa R Ghimire
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Tiina Hannunen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Hanna Lähteenmäki
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland
| | - Pauliina Rumm
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland
| | - Jason Theodoropoulos
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland
| | - Essi Laajala
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland
| | - Jouni Härkönen
- Faculty of Health Sciences, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Petri Pölönen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Merja Heinäniemi
- Faculty of Health Sciences, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Maija Hollmén
- Medicity Research Laboratory, University of Turku, 20014 Turku, Finland
| | - Shizuka Yamano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ryosuke Shirasaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rizwan Romee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - Harri Lähdesmäki
- Department of Computer Science, Aalto University, 02150 Espoo, Finland
| | - Dean A Lee
- Hematology/Oncology/BMT, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Ricardo De Matos Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA
| | - Matti Kankainen
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland; Laboratory of Genetics, HUS Diagnostic Center, Hospital District of Helsinki and Uusima (HUS), 00290 Helsinki, Finland
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, 00290 Helsinki, Finland.
| |
Collapse
|
21
|
Singh A, Ghosh R, Guchhait P. CXCR3 antagonist rescues ER stress and reduces inflammation and JEV infection in mice brain. Cytokine 2023; 172:156380. [PMID: 37812996 DOI: 10.1016/j.cyto.2023.156380] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/29/2023] [Accepted: 09/22/2023] [Indexed: 10/11/2023]
Abstract
The endoplasmic reticulum (ER) is crucial for maintaining cellular homeostasis, and synthesis and folding of proteins and lipids. The ER is sensitive to stresses including viral infection that perturb the intracellular energy level and redox state, and accumulating unfolded/misfolded proteins. Viruses including Japanese encephalitis virus (JEV) activates unfolded protein response (UPR) causing ER stress in host immune cells and promotes inflammation and apoptotic cell death. The chemokine receptor CXCR3 has been reported to play important role in the accumulation of inflammatory immune cells and neuronal cell death in several disease conditions. Recently we described the involvement of CXCR3 in regulating inflammation and JEV infection in mice brain. Supplementation with a CXCR3 antagonist AMG487 significantly reduced JEV infection in the mice brain in conjunction with the downregulation of UPR pathway via PERK:eIF2α:CHOP, and decreased mitochondrial ROS generation, inflammation and apoptotic cell death. Alongside, AMG487 treatment improved interferon (IFN)-α/β synthesis in JEV-infected mice brain. Thus, suggesting a potential therapeutic role of CXCR3 antagonist against JEV infection.
Collapse
Affiliation(s)
- Anamika Singh
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Riya Ghosh
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
22
|
Liu Y, Si D, Bai P, Zhu L, Zhang L, Chen Q, Qi Y. CXCL10 May Be Responsible for Susceptibility to Pulmonary Embolism in COVID-19 Patients. J Inflamm Res 2023; 16:4913-4924. [PMID: 37927958 PMCID: PMC10625331 DOI: 10.2147/jir.s431212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
Background Although the potential of coronavirus disease 2019 (COVID-19) patients to develop pulmonary embolism (PE) is widely recognized, the underlying mechanism has not been completely elucidated. This study aimed to identify genes common to COVID-19 and PE to reveal the underlying pathogenesis of susceptibility to PE in COVID-19 patients. Methods COVID-19 genes were obtained from the GEO database and the OMIM, CTD, GeneCards, and DisGeNET databases; PE genes were obtained from the OMIM, CTD, GeneCards, and DisGeNET databases. We overlapped the genes of COVID-19 and PE to obtain common genes for additional analysis, including functional enrichment, protein-protein interaction, and immune infiltration analysis. Hub genes were identified using cytoHubba, a plugin of Cytoscape, and validated using the independent datasets GSE167000 and GSE13535. The genes validated by the above datasets were further validated in clinical samples. Results We obtained 36 genes shared by PE and COVID-19. Functional enrichment and immune infiltration analyses revealed the involvement of cytokines and immune activation. Five genes (CCL2, CXCL10, ALB, EGF, and MKI67) were identified as hub genes common to COVID-19 and PE. CXCL10 was validated in both independent datasets (GSE167000 and GSE13535). Serum levels of CXCL10 in the COVID-19 group and the COVID-19 combined with PE group were significantly higher than those in the healthy control group (P<0.001). In addition, there were significant differences between the COVID-19 group and the COVID-19 combined with PE group (P<0.01). Conclusion Our study reveals common genes shared by PE and COVID-19 and identifies CXCL10 as a possible cause of susceptibility to PE in COVID-19 patients.
Collapse
Affiliation(s)
- Yingli Liu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Dan Si
- Department of Pulmonary and Critical Care Medicine, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, People’s Republic of China
| | - Pingping Bai
- Department of Health Management, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Li Zhu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Lili Zhang
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Qi Chen
- Department of Pulmonary and Critical Care Medicine, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Yong Qi
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan, 450003, People’s Republic of China
| |
Collapse
|
23
|
Hoffmann JA, Gründler K, Richter DU, Stubert J. Prediction of spontaneous preterm birth using CCL2 and CXCL10 in maternal serum of symptomatic high-risk pregnant women: a prospective cohort study. BMC Pregnancy Childbirth 2023; 23:697. [PMID: 37770883 PMCID: PMC10537471 DOI: 10.1186/s12884-023-06016-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/21/2023] [Indexed: 09/30/2023] Open
Abstract
INTRODUCTION CCL2 and CXCL10 are putative biomarkers for the prediction of spontaneous preterm birth. This study evaluates these markers in a cohort of pregnant high-risk women. MATERIAL AND METHODS In our prospective study, we included 109 women with signs of preterm labor between 20 + 0 and 31 + 6 weeks of gestation. Inclusion criteria were regular (< 3/30 min) or painful contractions, cervical length < 25 mm or a history of previous preterm birth (PTB). Blood samples were obtained upon first admission to our clinic. Biomarker concentrations were measured using pre-coated sandwich immunoassays (ELISA). Primary study outcome was spontaneous preterm birth < 34 weeks, secondary outcome was delivery < 37 weeks or within seven days after study inclusion. RESULTS Sixteen women (14.7%) delivered < 34 weeks and twenty women between 34 + 0 and 36 + 6 weeks (18.4%). Six patients (5.5%) gave birth within seven days after study admission. CXCL10 showed higher medium serum levels in women with PTB < 34 weeks (115 pg/ml compared to 61 pg/ml ≥ 34 weeks; p < 0.001) and < 37 weeks (103 pg/ml vs. 53 pg/ml; p < 0.001). In contrary, lower CCL2 serum levels were associated with PTB < 34 weeks (46 pg/ml vs. 73 pg/ml; p = 0.032) and birth within 7 days (25 pg/ml vs. 73 pg/ml; p = 0.008). The CXCL10/CCL2-ratio further improved the predictive model with a ROC-AUC of 0.83 (95% CI 0.73-0.93, p < 0.001) for delivery < 34 weeks. These corresponds to a sensitivity, specificity and positive predictive value of 0.67, 0.86 and 0.43 at a cut-off of 2.2. CONCLUSION Low maternal serum CCL2 levels are associated with a higher risk of preterm delivery within seven days. High CXCL10 serum levels are more associated with a high risk for preterm birth < 34 weeks. Elevated CXCL10/CCL2-ratio is showing the best predictive performance. TRIAL REGISTRATION NUMBER (DRKS-ID) DRKS00010763, Registration date: September 02, 2016.
Collapse
Affiliation(s)
- Jessica Alana Hoffmann
- Department of Obstetrics and Gynecology, Rostock University Medical Centre, Rostock, Germany
| | - Kathleen Gründler
- Department of Obstetrics and Gynecology, HELIOS Hospital Schwerin, Schwerin, Germany
| | - Dagmar- Ulrike Richter
- Department of Obstetrics and Gynecology, Rostock University Medical Centre, Rostock, Germany
| | - Johannes Stubert
- Department of Obstetrics and Gynecology, Rostock University Medical Centre, Rostock, Germany.
| |
Collapse
|
24
|
Di Trani CA, Cirella A, Arrizabalaga L, Alvarez M, Bella Á, Fernandez-Sendin M, Russo-Cabrera JS, Gomar C, Ardaiz N, Teijeira A, Bolaños E, González-Gomariz J, Otano I, Aranda F, Palencia B, Segués A, Huang S, van Duijnhoven SM, van Elsas A, Melero I, Berraondo P. Intratumoral injection of IL-12-encoding mRNA targeted to CSFR1 and PD-L1 exerts potent anti-tumor effects without substantial systemic exposure. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:599-616. [PMID: 37637207 PMCID: PMC10450355 DOI: 10.1016/j.omtn.2023.07.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023]
Abstract
IL-12 is a potent cytokine for cancer immunotherapy. However, its systemic delivery as a recombinant protein has shown unacceptable toxicity in the clinic. Currently, the intratumoral injection of IL-12-encoding mRNA or DNA to avoid such side effects is being evaluated in clinical trials. In this study, we aimed to improve this strategy by further favoring IL-12 tethering to the tumor. We generated in vitro transcribed mRNAs encoding murine single-chain IL-12 fused to diabodies binding to CSF1R and/or PD-L1. These targeted molecules are expressed in the tumor microenvironment, especially on myeloid cells. The binding capacity of chimeric constructs and the bioactivity of IL-12 were demonstrated in vitro and in vivo. Doses as low as 0.5 μg IL-12-encoding mRNA achieved potent antitumor effects in subcutaneously injected B16-OVA and MC38 tumors. Treatment delivery was associated with increases in IL-12p70 and IFN-γ levels in circulation. Fusion of IL-12 to the diabodies exerted comparable efficacy against bilateral tumor models. However, it achieved tethering to myeloid cells infiltrating the tumor, resulting in nearly undetectable systemic levels of IL-12 and IFN-γ. Overall, tethering IL-12 to intratumoral myeloid cells in the mRNA-transferred tumors achieves similar efficacy while reducing the dangerous systemic bioavailability of IL-12.
Collapse
Affiliation(s)
- Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Leire Arrizabalaga
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Maite Alvarez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| | - Ángela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Joan Salvador Russo-Cabrera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Celia Gomar
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Nuria Ardaiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - José González-Gomariz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Itziar Otano
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Belén Palencia
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Aina Segués
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, EH9 3FL Edinburgh, UK
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Shuyu Huang
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, EH9 3FL Edinburgh, UK
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, 3584 CS Utrecht, The Netherlands
| | | | | | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid, Spain
| |
Collapse
|
25
|
Neyton LPA, Patel RK, Sarma A, Willmore A, Haller SC, Kangelaris KN, Eckalbar WL, Erle DJ, Krummel MF, Hendrickson CM, Woodruff PG, Langelier CR, Calfee CS, Fragiadakis GK. Distinct pulmonary and systemic effects of dexamethasone in severe COVID-19. RESEARCH SQUARE 2023:rs.3.rs-3168149. [PMID: 37577607 PMCID: PMC10418533 DOI: 10.21203/rs.3.rs-3168149/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. We performed bulk and single-cell RNA sequencing of the lower respiratory tract and blood, and plasma cytokine profiling to study the effect of dexamethasone on systemic and pulmonary immune cells. We find decreased signatures of antigen presentation, T cell recruitment, and viral injury in patients treated with dexamethasone. We identify compartment- and cell- specific differences in the effect of dexamethasone in patients with severe COVID-19 that are reproducible in publicly available datasets. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.
Collapse
Affiliation(s)
- Lucile P A Neyton
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Ravi K Patel
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Andrew Willmore
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Sidney C Haller
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | | | - Walter L Eckalbar
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - David J Erle
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Lung Biology Center, University of California, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Carolyn M Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, CA, USA
| | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Division of Rheumatology, University of California, San Francisco, CA, USA
| |
Collapse
|
26
|
Kim J, Rosenberger MG, Chen S, IP CKM, Bahmani A, Chen Q, Shen J, Tang Y, Wang A, Kenna E, Son M, Tay S, Ferguson AL, Esser-Kahn AP. Discovery of New States of Immunomodulation for Vaccine Adjuvants via High Throughput Screening: Expanding Innate Responses to PRRs. ACS CENTRAL SCIENCE 2023; 9:427-439. [PMID: 36968540 PMCID: PMC10037445 DOI: 10.1021/acscentsci.2c01351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Indexed: 06/18/2023]
Abstract
Stimulation of the innate immune system is crucial in both effective vaccinations and immunotherapies. This is often achieved through adjuvants, molecules that usually activate pattern recognition receptors (PRRs) and stimulate two innate immune signaling pathways: the nuclear factor kappa-light-chain-enhancer of activated B-cells pathway (NF-κB) and the interferon regulatory factors pathway (IRF). Here, we demonstrate the ability to alter and improve adjuvant activity via the addition of small molecule "immunomodulators". By modulating signaling activity instead of receptor binding, these molecules allow the customization of select innate responses. We demonstrate both inhibition and enhancement of the products of the NF-κB and IRF pathways by several orders of magnitude. Some modulators apply generally across many receptors, while others focus specifically on individual receptors. Modulators boost correlates of a protective immune responses in a commercial flu vaccine model and reduced correlates of reactogenicity in a typhoid vaccine model. These modulators have a range of applications: from adjuvanticity in prophylactics to enhancement of immunotherapy.
Collapse
Affiliation(s)
| | | | - Siquan Chen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Carman KM IP
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Azadeh Bahmani
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Qing Chen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Jinjing Shen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Yifeng Tang
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Andrew Wang
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Emma Kenna
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Minjun Son
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Savaş Tay
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Andrew L. Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| |
Collapse
|
27
|
Lawrence JM, Schardien K, Wigdahl B, Nonnemacher MR. Roles of neuropathology-associated reactive astrocytes: a systematic review. Acta Neuropathol Commun 2023; 11:42. [PMID: 36915214 PMCID: PMC10009953 DOI: 10.1186/s40478-023-01526-9] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/08/2023] [Indexed: 03/16/2023] Open
Abstract
In the contexts of aging, injury, or neuroinflammation, activated microglia signaling with TNF-α, IL-1α, and C1q induces a neurotoxic astrocytic phenotype, classified as A1, A1-like, or neuroinflammatory reactive astrocytes. In contrast to typical astrocytes, which promote neuronal survival, support synapses, and maintain blood-brain barrier integrity, these reactive astrocytes downregulate supportive functions and begin to secrete neurotoxic factors, complement components like C3, and chemokines like CXCL10, which may facilitate recruitment of immune cells across the BBB into the CNS. The proportion of pro-inflammatory reactive astrocytes increases with age through associated microglia activation, and these pro-inflammatory reactive astrocytes are particularly abundant in neurodegenerative disorders. As the identification of astrocyte phenotypes progress, their molecular and cellular effects are characterized in a growing array of neuropathologies.
Collapse
Affiliation(s)
- Jill M Lawrence
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kayla Schardien
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Loos P, Baiwir J, Maquet C, Javaux J, Sandor R, Lallemand F, Marichal T, Machiels B, Gillet L. Dampening type 2 properties of group 2 innate lymphoid cells by a gammaherpesvirus infection reprograms alveolar macrophages. Sci Immunol 2023; 8:eabl9041. [PMID: 36827420 DOI: 10.1126/sciimmunol.abl9041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Immunological dysregulation in asthma is associated with changes in exposure to microorganisms early in life. Gammaherpesviruses (γHVs), such as Epstein-Barr virus, are widespread human viruses that establish lifelong infection and profoundly shape host immunity. Using murid herpesvirus 4 (MuHV-4), a mouse γHV, we show that after infection, lung-resident and recruited group 2 innate lymphoid cells (ILC2s) exhibit a reduced ability to expand and produce type 2 cytokines in response to house dust mites, thereby contributing to protection against asthma. In contrast, MuHV-4 infection triggers GM-CSF production by those lung ILC2s, which orders the differentiation of monocytes (Mos) into alveolar macrophages (AMs) without promoting their type 2 functions. In the context of γHV infection, ILC2s are therefore essential cells within the pulmonary niche that imprint the tissue-specific identity of Mo-derived AMs and shape their function well beyond the initial acute infection.
Collapse
Affiliation(s)
- Pauline Loos
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Jérôme Baiwir
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Céline Maquet
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Justine Javaux
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Rémy Sandor
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - François Lallemand
- Centre Hospitalier Universitaire de Liège, Département de Physique Médicale, Service médical de radiothérapie, Liège 4000, Belgium
| | - Thomas Marichal
- Laboratory of Immunophysiology, GIGA-Research and Faculty of Veterinary Medicine, ULiège, Liège 4000, Belgium
| | - Bénédicte Machiels
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| | - Laurent Gillet
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège 4000, Belgium
| |
Collapse
|
29
|
Good PI, Li L, Hurst HA, Serrano Herrera I, Xu K, Rao M, Bateman DA, Al-Awqati Q, D’Agati VD, Costantini F, Lin F. Low nephron endowment increases susceptibility to renal stress and chronic kidney disease. JCI Insight 2023; 8:e161316. [PMID: 36626229 PMCID: PMC9977438 DOI: 10.1172/jci.insight.161316] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Preterm birth results in low nephron endowment and increased risk of acute kidney injury (AKI) and chronic kidney disease (CKD). To understand the pathogenesis of AKI and CKD in preterm humans, we generated potentially novel mouse models with a 30%-70% reduction in nephron number by inhibiting or deleting Ret tyrosine kinase in the developing ureteric bud. These mice developed glomerular and tubular hypertrophy, followed by the transition to CKD, recapitulating the renal pathological changes seen in humans born preterm. We injected neonatal mice with gentamicin, a ubiquitous nephrotoxic exposure in preterm infants, and detected more severe proximal tubular injury in mice with low nephron number compared with controls with normal nephron number. Mice with low nephron number had reduced proliferative repair with more rapid development of CKD. Furthermore, mice had more profound inflammation with highly elevated levels of MCP-1 and CXCL10, produced in part by damaged proximal tubules. Our study directly links low nephron endowment with postnatal renal hypertrophy, which in this model is maladaptive and results in CKD. Underdeveloped kidneys are more susceptible to gentamicin-induced AKI, suggesting that AKI in the setting of low nephron number is more severe and further increases the risk of CKD in this vulnerable population.
Collapse
Affiliation(s)
| | - Ling Li
- Department of Pediatrics and
| | | | | | - Katherine Xu
- Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons New York, New York, USA
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston Massachusetts, USA
| | | | - Qais Al-Awqati
- Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons New York, New York, USA
| | - Vivette D. D’Agati
- Department of Pathology and Cellular Biology at Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Frank Costantini
- Department of Genetics and Development at Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | |
Collapse
|
30
|
Walaas GA, Gopalakrishnan S, Bakke I, Skovdahl HK, Flatberg A, Østvik AE, Sandvik AK, Bruland T. Physiological hypoxia improves growth and functional differentiation of human intestinal epithelial organoids. Front Immunol 2023; 14:1095812. [PMID: 36793710 PMCID: PMC9922616 DOI: 10.3389/fimmu.2023.1095812] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Background The epithelium in the colonic mucosa is implicated in the pathophysiology of various diseases, including inflammatory bowel diseases and colorectal cancer. Intestinal epithelial organoids from the colon (colonoids) can be used for disease modeling and personalized drug screening. Colonoids are usually cultured at 18-21% oxygen without accounting for the physiological hypoxia in the colonic epithelium (3% to <1% oxygen). We hypothesize that recapitulating the in vivo physiological oxygen environment (i.e., physioxia) will enhance the translational value of colonoids as pre-clinical models. Here we evaluate whether human colonoids can be established and cultured in physioxia and compare growth, differentiation, and immunological responses at 2% and 20% oxygen. Methods Growth from single cells to differentiated colonoids was monitored by brightfield images and evaluated with a linear mixed model. Cell composition was identified by immunofluorescence staining of cell markers and single-cell RNA-sequencing (scRNA-seq). Enrichment analysis was used to identify transcriptomic differences within cell populations. Pro-inflammatory stimuli induced chemokines and Neutrophil gelatinase-associated lipocalin (NGAL) release were analyzed by Multiplex profiling and ELISA. Direct response to a lower oxygen level was analyzed by enrichment analysis of bulk RNA sequencing data. Results Colonoids established in a 2% oxygen environment acquired a significantly larger cell mass compared to a 20% oxygen environment. No differences in expression of cell markers for cells with proliferation potential (KI67 positive), goblet cells (MUC2 positive), absorptive cells (MUC2 negative, CK20 positive) and enteroendocrine cells (CGA positive) were found between colonoids cultured in 2% and 20% oxygen. However, the scRNA-seq analysis identified differences in the transcriptome within stem-, progenitor- and differentiated cell clusters. Both colonoids grown at 2% and 20% oxygen secreted CXCL2, CXCL5, CXCL10, CXCL12, CX3CL1 and CCL25, and NGAL upon TNF + poly(I:C) treatment, but there appeared to be a tendency towards lower pro-inflammatory response in 2% oxygen. Reducing the oxygen environment from 20% to 2% in differentiated colonoids altered the expression of genes related to differentiation, metabolism, mucus lining, and immune networks. Conclusions Our results suggest that colonoids studies can and should be performed in physioxia when the resemblance to in vivo conditions is important.
Collapse
Affiliation(s)
- Gunnar Andreas Walaas
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Shreya Gopalakrishnan
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Helene Kolstad Skovdahl
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research (CEMIR), NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Arnar Flatberg
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Central Administration, St. Olav's University Hospital, Trondheim, Norway
| | - Ann Elisabet Østvik
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Arne Kristian Sandvik
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research (CEMIR), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine (IKOM), NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
31
|
Upadhyay TK, Trivedi R, Khan F, Pandey P, Sharangi AB, Goel H, Saeed M, Park MN, Kim B. Potential Therapeutic Role of Mesenchymal-Derived Stem Cells as an Alternative Therapy to Combat COVID-19 through Cytokines Storm. Cells 2022; 11:2686. [PMID: 36078094 PMCID: PMC9455060 DOI: 10.3390/cells11172686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 01/08/2023] Open
Abstract
Medical health systems continue to be challenged due to newly emerging COVID-19, and there is an urgent need for alternative approaches for treatment. An increasing number of clinical observations indicate cytokine storms to be associated with COVID-19 severity and also to be a significant cause of death among COVID-19 patients. Cytokine storm involves the extensive proliferative and hyperactive activity of T and macrophage cells and the overproduction of pro-inflammatory cytokines. Stem cells are the type of cell having self-renewal properties and giving rise to differentiated cells. Currently, stem cell therapy is an exciting and promising therapeutic approach that can treat several diseases that were considered incurable in the past. It may be possible to develop novel methods to treat various diseases by identifying stem cells' growth and differentiation factors. Treatment with mesenchymal stem cells (MSCs) in medicine is anticipated to be highly effective. The present review article is organized to put forward the positive arguments and implications in support of mesenchymal stem cell therapy as an alternative therapy to cytokine storms, to combat COVID-19. Using the immunomodulatory potential of the MSCs, it is possible to fight against COVID-19 and counterbalance the cytokine storm.
Collapse
Affiliation(s)
- Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Rashmi Trivedi
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida 201306, India
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida 201306, India
| | - Amit Baran Sharangi
- Department of Plantation, Spices, Medicinal & Aromatic Crops, BCKV-Agricultural University, Mohanpur 741252, India
| | - Harsh Goel
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi 110023, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail 34464, Saudi Arabia
| | - Moon Nyeo Park
- Department of Korean Medicine, Kyung Hee University, Seoul 05254, Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
32
|
André S, Azarias da Silva M, Picard M, Alleaume-Buteau A, Kundura L, Cezar R, Soudaramourty C, André SC, Mendes-Frias A, Carvalho A, Capela C, Pedrosa J, Gil Castro A, Loubet P, Sotto A, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Tran TA, Zghidi-Abouzid O, Nioche P, Silvestre R, Corbeau P, Mammano F, Estaquier J. Low quantity and quality of anti-spike humoral response is linked to CD4 T-cell apoptosis in COVID-19 patients. Cell Death Dis 2022; 13:741. [PMID: 36030261 PMCID: PMC9419645 DOI: 10.1038/s41419-022-05190-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
In addition to an inflammatory reaction, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)-infected patients present lymphopenia, which we recently reported as being related to abnormal programmed cell death. As an efficient humoral response requires CD4 T-cell help, we hypothesized that the propensity of CD4 T cells to die may impact the quantity and quality of the humoral response in acutely infected individuals. In addition to specific immunoglobulins (Ig)A, IgM, and IgG against SARS-CoV-2 nucleocapsid (N), membrane (M), and spike (S1) proteins, we assessed the quality of IgG response by measuring the avidity index. Because the S protein represents the main target for neutralization and antibody-dependent cellular cytotoxicity responses, we also analyzed anti-S-specific IgG using S-transfected cells (S-Flow). Our results demonstrated that most COVID-19 patients have a predominant IgA anti-N humoral response during the early phase of infection. This specific humoral response preceded the anti-S1 in time and magnitude. The avidity index of anti-S1 IgG was low in acutely infected individuals compared to convalescent patients. We showed that the percentage of apoptotic CD4 T cells is inversely correlated with the levels of specific IgG antibodies. These lower levels were also correlated positively with plasma levels of CXCL10, a marker of disease severity, and soluble Fas ligand that contributes to T-cell death. Finally, we found lower S-Flow responses in patients with higher CD4 T-cell apoptosis. Altogether, these results demonstrate that individuals with high levels of CD4 T-cell apoptosis and CXCL10 have a poor ability to build an efficient anti-S response. Consequently, preventing CD4 T-cell death might be a strategy for improving humoral response during the acute phase, thereby reducing COVID-19 pathogenicity.
Collapse
Affiliation(s)
- Sonia André
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Marne Azarias da Silva
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Morgane Picard
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Aurélie Alleaume-Buteau
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,grid.508487.60000 0004 7885 7602Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Lucy Kundura
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | - Renaud Cezar
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | | | - Santa Cruz André
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Ana Mendes-Frias
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Alexandre Carvalho
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Carlos Capela
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Jorge Pedrosa
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Gil Castro
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paul Loubet
- grid.411165.60000 0004 0593 8241Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Albert Sotto
- grid.411165.60000 0004 0593 8241Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Laurent Muller
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Jean-Yves Lefrant
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Claire Roger
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Pierre-Géraud Claret
- grid.411165.60000 0004 0593 8241Urgences Médico-Chirugicales Hospitalisation, CHU de Nîmes, Nîmes, France
| | - Sandra Duvnjak
- grid.411165.60000 0004 0593 8241Service de Gérontologie et Prévention du Vieillissement, CHU de Nîmes, Nîmes, France
| | - Tu-Anh Tran
- grid.411165.60000 0004 0593 8241Service de Pédiatrie, CHU de Nîmes, Nîmes, France
| | | | - Pierre Nioche
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,grid.508487.60000 0004 7885 7602Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Ricardo Silvestre
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pierre Corbeau
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France ,grid.121334.60000 0001 2097 0141Institut de Génétique Humaine UMR9002 CNRS-Université de Montpellier, Montpellier, France
| | - Fabrizio Mammano
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,INSERM U1259 MAVIVH, Université de Tours, Tours, France
| | - Jérôme Estaquier
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,CHU de Québec—Université Laval Research Center, Québec City, QC Canada
| |
Collapse
|
33
|
Szpakowski P, Ksiazek-Winiarek D, Turniak-Kusy M, Pacan I, Glabinski A. Human Primary Astrocytes Differently Respond to Pro- and Anti-Inflammatory Stimuli. Biomedicines 2022; 10:biomedicines10081769. [PMID: 35892669 PMCID: PMC9331936 DOI: 10.3390/biomedicines10081769] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022] Open
Abstract
For a long time, astrocytes were considered a passive brain cell population. However, recently, many studies have shown that their role in the central nervous system (CNS) is more active. Previously, it was stated that there are two main functional phenotypes of astrocytes. However, nowadays, it is clear that there is rather a broad spectrum of these phenotypes. The major goal of this study was to evaluate the production of some inflammatory chemokines and neurotrophic factors by primary human astrocytes after pro- or anti-inflammatory stimulation. We observed that only astrocytes induced by inflammatory mediators TNFα/IL-1a/C1q produced CXCL10, CCL1, and CXCL13 chemokines. Unstimulated astrocytes and those cultured with anti-inflammatory cytokines (IL-4, IL-10, or TGF-β1) did not produce these chemokines. Interestingly, astrocytes cultured in proinflammatory conditions significantly decreased the release of neurotrophic factor PDGF-A, as compared to unstimulated astrocytes. However, in response to anti-inflammatory cytokine TGF-β1, astrocytes significantly increased PDGF-A production compared to the medium alone. The production of another studied neurotrophic factor BDNF was not influenced by pro- or anti-inflammatory stimulation. The secretory response was accompanied by changes in HLA-DR, CD83, and GFAP expression. Our study confirms that astrocytes differentially respond to pro- and anti-inflammatory stimuli, especially to inflammatory cytokines TNF-α, IL-1a, and C1q, suggesting their role in leukocyte recruitment.
Collapse
|
34
|
Smith R, Wafa EI, Geary SM, Ebeid K, Alhaj-Suliman SO, Salem AK. Cationic nanoparticles enhance T cell tumor infiltration and antitumor immune responses to a melanoma vaccine. SCIENCE ADVANCES 2022; 8:eabk3150. [PMID: 35857851 PMCID: PMC9299550 DOI: 10.1126/sciadv.abk3150] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
In clinical settings, cancer vaccines as monotherapies have displayed limited success compared to other cancer immunotherapeutic treatments. Nanoscale formulations have the ability to increase the efficacy of cancer vaccines by combatting the immunosuppressive nature of the tumor microenvironment. Here, we have synthesized a previously unexplored cationic polymeric nanoparticle formulation using polyamidoamine dendrimers and poly(d,l-lactic-co-glycolic acid) that demonstrate adjuvant properties in vivo. Tumor-challenged mice vaccinated with an adenovirus-based cancer vaccine [encoding tumor-associated antigen (TAA)] and subsequently treated with this nanoparticulate formulation showed significant increases in TAA-specific T cells in the peripheral blood, reduced tumor burden, protection against tumor rechallenge, and a significant increase in median survival. An investigation into cell-based pathways suggests that administration of the nanoformulation at the site of the developing tumor may have created an inflammatory environment that attracted activated TAA-specific CD8+ T cells to the vicinity of the tumor, thus enhancing the efficacy of the vaccine.
Collapse
Affiliation(s)
| | | | | | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Suhaila O. Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
35
|
Liang R, Chen S, Jin Y, Tao L, Ji W, Zhu P, Li D, Zhang Y, Zhang W, Duan G. The CXCL10/CXCR3 Axis Promotes Disease Pathogenesis in Mice upon CVA2 Infection. Microbiol Spectr 2022; 10:e0230721. [PMID: 35604176 PMCID: PMC9241849 DOI: 10.1128/spectrum.02307-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/16/2022] [Indexed: 11/20/2022] Open
Abstract
Coxsackievirus A2 (CVA2) is an emerging pathogen that results in hand-foot-and-mouth disease (HFMD) outbreaks. Systemic inflammatory response and central nervous system inflammation are the main pathological features of fatal HFMD. However, the immunopathogenesis of CVA2 infection is poorly understood. We first detected the transcriptional levels of 81 inflammation-related genes in neonatal mice with CVA2 infection. Remarkably, CVA2 induced higher expression of chemokine (C-X-C motif) ligand 10 (CXCL10) in multiple organs and tissues. CXCL10 acts through its cognate receptor chemokine (C-X-C motif) receptor 3 (CXCR3) and regulates immune responses. CXCL10/CXCR3 activation contributes to the pathogenesis of many inflammatory diseases. Next, we found CXCL10 and CXCR3 expression to be significantly elevated in the organs and tissues from CVA2-infected mice at 5 days postinfection (dpi) using immunohistochemistry (IHC). To further explore the role of CXCL10/CXCR3 in CVA2 pathogenesis, an anti-CXCR3 neutralizing antibody (αCXCR3) or IgG isotype control antibody was used to treat CVA2-infected mice on the same day as infection and every 24 h until 5 dpi. Our results showed that αCXCR3 therapy relieved the clinical manifestations and pathological damage and improved the survival rate of CVA2-infected mice. Additionally, αCXCR3 treatment reduced viral loads and reversed the proinflammatory cytokine (interleukin 6 [IL-6], tumor necrosis factor alpha [TNF-α], and IL-1β) expression, apoptosis, and inflammatory cell infiltration induced by CVA2. Collectively, our study presents evidence for the involvement of the CXCL10/CXCR3 axis in CVA2 pathogenesis. The activation of CXCL10/CXCR3 contributes to CVA2 pathogenesis by inducing apoptosis, proinflammatory cytokine expression, and inflammatory cell infiltration, which can be reversed by αCXCR3 therapy. This study provides new insight into the pathogenesis of HFMD, which has an important guiding significance for the treatment of HFMD. IMPORTANCE Systemic inflammatory response and central nervous system inflammation are the main pathological features of fatal HFMD cases. We detected the expression of 81 inflammation-related genes and found higher expression of CXCL10 in CVA2-infected mice. Next, we confirmed CXCL10/CXCR3 activation using immunohistochemistry and found that anti-CXCR3 neutralizing antibody (αCXCR3) therapy could relieve the clinical manifestations and pathological damage and improve the survival rate of CVA2-infected mice. Additionally, αCXCR3 treatment reduced viral loads and reversed the proinflammatory cytokine (IL-6, TNF-α, and IL-1β) expression, apoptosis, and inflammatory cell infiltration induced by CVA2. Collectively, our study presents the first evidence for the involvement of the CXCL10/CXCR3 axis in CVA2 pathogenesis. The activation of CXCL10/CXCR3 contributes to CVA2 pathogenesis via inducing apoptosis, proinflammatory cytokine expression, and inflammatory cell infiltration, which can be reversed by αCXCR3 therapy. This study provides new insight into the pathogenesis of HFMD, which has an important guiding significance for the treatment of HFMD.
Collapse
Affiliation(s)
- Ruonan Liang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shuaiyin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuefei Jin
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ling Tao
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Wangquan Ji
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peiyu Zhu
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Dong Li
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yu Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Weiguo Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Guangcai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Molecular Medicine, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Inflammatory gene silencing in activated monocytes by a cholesterol tagged-miRNA/siRNA: a novel approach to ameliorate diabetes induced inflammation. Cell Tissue Res 2022; 389:219-240. [PMID: 35604451 DOI: 10.1007/s00441-022-03637-6] [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: 08/13/2021] [Accepted: 05/09/2022] [Indexed: 11/02/2022]
Abstract
There is a major unmet need for the development of effective therapies for diabetes induced inflammation. Increased adenosine-uridine rich elements (AREs) containing mRNAs of inflammatory molecules are reported in inflamed monocytes. Destabilizing these inflammatory mRNAs by the miR-16 could reduce inflammation. DNA microarrays and in vitro cell studies showed that exogenous miR16 and its mimic treatment, in LPS/PMA induced monocytes, significantly downregulated several ARE containing inflammatory cytokine mRNAs similar to those seen in the normal monocytes. Ingenuity pathway analyses showed exogenous miR-16 or its synthetic mimic treatment alleviates inflammatory responses. To selectively target uptake, especially to inflamed cells, one of the CD36 substrate cholesterol was tagged to miR16/siRNA. Cholesterol tagged miR-16/ARE-siRNA showed enhanced uptake in CD36 expressing inflamed cells. In LPS or PMA, treated monocytes, candidate genes expressions levels such as IL-6, IL-8, IL-12β, IP-10, and TNF-α mRNA were increased, as measured by RT-qPCR as seen in primary monocytes of diabetes patients. Exogenous miR16 or ARE-siRNA transfection reduced mRNAs of pro-inflammatory cytokines levels in monocyte, and its adhesion. Increased uptake of cholesterol tagged miR-16 through the CD36 receptor was observed. This destabilizes numerous inflammatory ARE containing mRNAs and alleviates inflammatory responses. Cholesterol-tagged miR-16 and its mimic are novel anti-inflammatory molecules that can be specifically targeted to, via through CD36 expressing, "inflamed" cells and thus serve as therapeutic candidates to alleviate inflammatory diseases.
Collapse
|
37
|
Hong C, Lu J, Chen R, Liu H, Chen H, Wu X, Guo W, Huang Z, Liao H. CXCL10 levels in diagnosis and improved hemodynamics in patients with chronic thromboembolic pulmonary hypertension undergoing balloon pulmonary angioplasty. Pulm Circ 2022; 12:e12091. [PMID: 35685949 PMCID: PMC9171940 DOI: 10.1002/pul2.12091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/09/2022] [Accepted: 04/30/2022] [Indexed: 11/06/2022] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a progressive pulmonary vascular disease characterized by pulmonary artery stenosis or obstructions resulting from insufficient thrombus resolution. Chemokine (C‐X‐C motif) ligand 10 (CXCL10) is a chemokine that contributes to the pathogenesis of many autoimmune diseases and cancers. The present study aims to investigate the levels of CXCL10 in patients with CTEPH throughout balloon pulmonary angioplasty (BPA) and its correlation with the improvement of pulmonary hemodynamics. Plasma CXCL10 levels were measured in 38 CTEPH patients with 100 BPA sessions and in 28 healthy controls. Correlations between CXCL10 and pulmonary hemodynamics were investigated. Receiver operating characteristic (ROC) curves were plotted to display the diagnostic value and the predictive ability for perioperative complications of CXCL10 and CXCL10‐related models. Nomograms were plotted to visualize the diagnostic value and the predictive ability for perioperative complications of CXCL10 and CXCL10‐related models. CXCL10 levels are higher in CTEPH patients compared with healthy controls (36.5 [95% confidence interval {CI}: 25.0–51.1] vs. 14.8 [95% CI: 11.1–30.9], p < 0.0001) and decreased significantly after BPA treatment (36.5 [95% CI: 25.0–51.1] vs. 24.7 [95% CI: 17.2–36.6], p < 0.0005). Preoperative CXCL10 levels positively correlated with mean right atrial pressure (r = 0.25), systolic pulmonary artery pressure (PAP; r = 0.28), diastolic PAP (r = 0.33), mean PAP (r = 0.36), pulmonary vascular resistance (r = 0.31), and N‐terminal pro‐B‐type natriuretic peptide (NT‐proBNP; r = 0.46). Furthermore, plasma CXCL10 levels adjusting for age and sex displayed a sensitivity of 86.0% and a specificity of 67.9% for discriminating CTEPH patients from healthy controls. Preoperative CXCL10 levels, in combination with NT‐proBNP, predicted perioperative complications with a sensitivity of 100.0% and a specificity of 46.9% as displayed in ROC analysis. In conclusion, circulating CXCL10 might contribute to the evaluation of disease severity in CTEPH patients and be useful to evaluate the treatment effect of BPA. Future studies are warranted to further study the relationship between pulmonary hemodynamics and circulating CXCL10.
Collapse
Affiliation(s)
- Cheng Hong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Jianmin Lu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Riken Chen
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Haimin Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Haiming Chen
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Xiaofeng Wu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Wenliang Guo
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Zijie Huang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Huizhao Liao
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| |
Collapse
|
38
|
De Almeida SM, Rotta I, Tang B, Umlauf A, Vaida F, Cherner M, Franklin D, Letendre S, Ellis RJ. Higher Cerebrospinal Fluid Soluble Urokinase-type Plasminogen Activator Receptor, But Not Interferon γ-inducible Protein 10, Correlate With Higher Working Memory Deficits. J Acquir Immune Defic Syndr 2022; 90:106-114. [PMID: 35090158 PMCID: PMC8986587 DOI: 10.1097/qai.0000000000002924] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND We hypothesized that the induction of monocyte activation biomarkers, especially soluble urokinase-type plasminogen activator receptor (suPAR) and interferon γ-inducible protein 10 (IP-10), is lower in HIV-1C than HIV-1B, owing to a defective Tat cysteine dimotif (C30S). METHODS A total of 68 paired cerebrospinal fluid (CSF) and blood samples from people with HIV (PWH), free of CNS opportunistic infections, from a Southern Brazil outpatient HIV clinic were evaluated such as HIV-1B subtype (n = 27), HIV-1C (n = 26), other (n = 15), and 19 HIV-negative controls. The levels of suPAR, IP-10, neopterin, and β2 microglobulin (β2m) in the CSF and serum were quantified using different immunoassays. RESULTS Overall, in PWH, increases in CSF suPAR, CSF/serum suPAR, and CSF/serum β2m correlated with worse working memory deficits (r = 0.303, 0.353, and 0.289, respectively, all P < 0.05). The medians of IP-10, suPAR, neopterin, and β2m in CSF and serum and the CSF/serum ratio and suPAR index were comparable between the HIV-1B and HIV-1C subtypes. CSF IP-10 and neopterin and serum IP-10 and suPAR levels were higher in PWH than the HIV-negative controls (P = 0.015, P = 0.001, P < 0.0001, and P < 0.001, respectively). The serum β2m level was higher in HIV-associated dementia than neuropsychologically normal or asymptomatic (P = 0.024). DISCUSSION We observed that higher levels of CSF suPAR and the suPAR quotient correlated with worse working memory deficit. Elevated levels of monocyte activation were similar in both HIV-1 B and C subtypes, providing no evidence of reduced neuropathogenicity of HIV-1 subtype C Tat compared with subtype B.
Collapse
Affiliation(s)
- Sergio M De Almeida
- Neuroinfection Unity and Virology Laboratory, Universidade Federal Do Paraná, Curitiba, Paraná, Brazil
| | - Indianara Rotta
- Neuroinfection Unity and Virology Laboratory, Universidade Federal Do Paraná, Curitiba, Paraná, Brazil
| | - Bin Tang
- Department of Psychiatry, University of California, San Diego, CA
| | - Anya Umlauf
- Department of Psychiatry, University of California, San Diego, CA
| | - Florin Vaida
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California, San Diego, CA
| | - Mariana Cherner
- Department of Psychiatry, University of California, San Diego, CA
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
| | - Donald Franklin
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
| | - Scott Letendre
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, CA; and
| | - Ronald J Ellis
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
- Department of Neurosciences, University of California, San Diego, CA
| |
Collapse
|
39
|
Weissman R, Diamond EL, Haroche J, Durham BH, Cohen F, Buthorn J, Amoura Z, Emile JF, Mazor RD, Shomron N, Abdel-Wahab OI, Shpilberg O, Hershkovitz-Rokah O. MicroRNA-15a-5p acts as a tumor suppressor in histiocytosis by mediating CXCL10-ERK-LIN28a-let-7 axis. Leukemia 2022; 36:1139-1149. [PMID: 34785791 PMCID: PMC8979810 DOI: 10.1038/s41375-021-01472-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 01/18/2023]
Abstract
Erdheim-Chester disease (ECD) is characterized by excessive production and accumulation of histiocytes within multiple tissues and organs. ECD patients harbor recurrent mutations of genes associated with the RAS/RAF/MEK/ERK signaling pathway, particularly, the BRAFV600E mutation. Following our previous finding that miR-15a-5p is the most prominently downregulated microRNA in ECD patients compared to healthy individuals, we elucidated its role in ECD pathogenesis. Bioinformatics analysis followed by a luciferase assay showed that chemokine ligand 10 (CXCL10) is a target gene regulated by miRNA-15a-5p. This was confirmed in 24/34 ECD patients that had low expression of miR-15a-5p concurrent with upregulated CXCL10. Overexpression of miR-15a-5p in cell lines harboring BRAF or RAS mutations (Ba/F3, KG-1a and OCI-AML3) resulted in CXCL10 downregulation, followed by LIN28a and p-ERK signaling downregulation and let-7 family upregulation. Overexpression of miR-15a-5p inhibited cell growth and induced apoptosis by decreasing Bcl-2 and Bcl-xl levels. Analysis of sequential samples from 7 ECD patients treated with MAPK inhibitors (vemurafenib/cobimetinib) for 4 months showed miR-15a-5p upregulation and CXCL10 downregulation. Our findings suggest that miR-15a-5p is a tumor suppressor in ECD through the CXCL10-ERK-LIN28a-let7 axis, highlighting another layer of post-transcriptional regulation in this disease. Upregulation of miR-15a-5p in ECD patients may have a potential therapeutic role.
Collapse
Affiliation(s)
- Ran Weissman
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
- Translational Research Lab, Assuta Medical Centers, Tel-Aviv, Israel
| | - Eli L Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julien Haroche
- Service de Médecine Interne, Hôpital Universitaire Pitié Salpêtrière - Charles Foix, Sorbonne Université, Faculté de Médecine, Paris, France
| | - Benjamin H Durham
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fleur Cohen
- Service de Médecine Interne, Hôpital Universitaire Pitié Salpêtrière - Charles Foix, Sorbonne Université, Faculté de Médecine, Paris, France
| | - Justin Buthorn
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zahir Amoura
- Service de Médecine Interne, Hôpital Universitaire Pitié Salpêtrière - Charles Foix, Sorbonne Université, Faculté de Médecine, Paris, France
| | - Jean-François Emile
- Research Unit EA4340, Versailles University, Paris-Saclay University, Boulogne, France
- Pathology Department, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Boulogne, France
| | - Roei D Mazor
- Institute of Hematology/Clinic of Histiocytic Neoplasms, Assuta Medical Centers, Tel-Aviv, Israel
| | - Noam Shomron
- Faculty of Medicine and Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Omar I Abdel-Wahab
- Research Unit EA4340, Versailles University, Paris-Saclay University, Boulogne, France
| | - Ofer Shpilberg
- Translational Research Lab, Assuta Medical Centers, Tel-Aviv, Israel
- Institute of Hematology/Clinic of Histiocytic Neoplasms, Assuta Medical Centers, Tel-Aviv, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Oshrat Hershkovitz-Rokah
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel.
- Translational Research Lab, Assuta Medical Centers, Tel-Aviv, Israel.
| |
Collapse
|
40
|
Panda A, Betigeri A, Ganesan S. A Gene Panel for Early Identification of Future Responders to Immune Checkpoint Blockade. Front Genet 2022; 13:706468. [PMID: 35309122 PMCID: PMC8928072 DOI: 10.3389/fgene.2022.706468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 02/09/2022] [Indexed: 11/28/2022] Open
Abstract
Immune checkpoint blockade (ICB), therapies that target the PD-1 pathway, CTLA-4 pathway, and other checkpoint pathways, lead to durable responses in many cancer types. Since only a minority of patients respond to ICB, it may be useful to identify the future responders early in the course of treatment. In this study we evaluated a small (15 genes) biologically motivated panel, consisting of genes involved in immune activation and checkpoint pathways, for early identification of future responders to ICB. The panel passed consistency check, pathological and in-silico validations, and was an excellent predictor (area under ROC curve >0.95) of eventual response to ICB, both CTLA-4 and PD-1 blockade, when applied to metastatic melanoma patients undergoing ICB (i.e., “on-treatment”) in a publicly available dataset. These results suggest that this small biologically motivated panel may be useful for early identification of future responders to ICB.
Collapse
Affiliation(s)
- Anshuman Panda
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- *Correspondence: Anshuman Panda,
| | - Anil Betigeri
- Akash Institute of Medical Sciences, Bangalore, India
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
41
|
Qiao X, Zhang W, Zhao W. Role of CXCL10 in Spinal Cord Injury. Int J Med Sci 2022; 19:2058-2070. [PMID: 36483597 PMCID: PMC9724238 DOI: 10.7150/ijms.76694] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury (SCI) results in acute inflammatory responses and secondary damages, including neuronal and glial cell death, axonal damage and demyelination, and blood-brain barrier (BBB) damage, eventually leading to neuronal dysfunction and other complications. C-X-C motif Chemokine Ligand 10 (CXCL10) is expressed after the injury, playing multiple roles in the development and progression of SCI. Moreover, the CXCL10 antagonist can restrict inflammatory immune responses and promote neuronal regeneration and functional recovery. In this review, we summarize the structure and biological functions of CXCL10, and the roles of the CXCL10 / CXCR3 axis in acute inflammatory responses, secondary damages, and complications during SCI, thus providing a potential theoretical basis by highlighting CXCL10 as a new potential drug target for the treatment of SCI.
Collapse
Affiliation(s)
- Xinyu Qiao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, China
| | - Weijiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
42
|
Krebs R, Tikkanen JM, Raissadati A, Hollmén M, Dhaygude K, Lemström KB. Inhibition of Vascular Endothelial Growth Factor Receptors 1 and 2 Attenuates Natural Killer Cell and Innate Immune Responses in an Experimental Model for Obliterative Bronchiolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 192:254-269. [PMID: 34774518 DOI: 10.1016/j.ajpath.2021.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023]
Abstract
Obliterative bronchiolitis (OB) after lung transplantation is a nonreversible, life-threatening complication. We investigated the role of vascular endothelial growth factor receptor (VEGFR)-1 and -2 in the development of obliterative airway disease (OAD), an experimental model for OB. The nonimmunosuppressed recipients underwent transplantation with fully major histocompatibility complex mismatched heterotopic tracheal allografts and received VEGFR-1 and -2-specific monoclonal antibodies either alone or in combination or rat IgG as a control. The treatment with VEGFR-1- or -2-blocking antibody significantly decreased intragraft mRNA expression of natural killer cell activation markers early after transplantation. This was followed by reduced infiltration of CD11b+ cells and CD4+ T cells as well as down-regulated mRNA expression of proinflammatory chemokines and profibrotic growth factors. However, blocking of both VEGFR-1 and -2 was necessary to reduce luminal occlusion. Furthermore, concomitant inhibition of the calcineurin activation pathway almost totally abolished the development of OAD. This study proposes that blocking of VEGF receptors blunted natural killer cell and innate immune responses early after transplantation and attenuated the development of OAD. The results of this study suggest that further studies on the role of VEGFR-1 and -2 blocking in development of obliterative airway lesions might be rewarding.
Collapse
Affiliation(s)
- Rainer Krebs
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland.
| | - Jussi M Tikkanen
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland; Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Alireza Raissadati
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Maria Hollmén
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Kishor Dhaygude
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Karl B Lemström
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland; Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
43
|
The Involvement of CXC Motif Chemokine Ligand 10 (CXCL10) and Its Related Chemokines in the Pathogenesis of Coronary Artery Disease and in the COVID-19 Vaccination: A Narrative Review. Vaccines (Basel) 2021; 9:vaccines9111224. [PMID: 34835155 PMCID: PMC8623875 DOI: 10.3390/vaccines9111224] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) and coronary heart disease (CHD) constitute two of the leading causes of death in Europe, USA and the rest of the world. According to the latest reports of the Iranian National Health Ministry, CAD is the main cause of death in Iranian patients with an age over 35 years despite a significant reduction in mortality due to early interventional treatments in the context of an acute coronary syndrome (ACS). Inflammation plays a fundamental role in coronary atherogenesis, atherosclerotic plaque formation, acute coronary thrombosis and CAD establishment. Chemokines are well-recognized mediators of inflammation involved in several bio-functions such as leucocyte migration in response to inflammatory signals and oxidative vascular injury. Different chemokines serve as chemo-attractants for a wide variety of cell types including immune cells. CXC motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10/CXLC10), is a chemokine with inflammatory features whereas CXC chemokine receptor 3 (CXCR3) serves as a shared receptor for CXCL9, 10 and 11. These chemokines mediate immune responses through the activation and recruitment of leukocytes, eosinophils, monocytes and natural killer (NK) cells. CXCL10, interleukin (IL-15) and interferon (IFN-g) are increased after a COVID-19 vaccination with a BNT162b2 mRNA (Pfizer/BioNTech) vaccine and are enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the second vaccination. The aim of the present study is the presentation of the elucidation of the crucial role of CXCL10 in the patho-physiology and pathogenesis of CAD and in identifying markers associated with the vaccination resulting in antibody development.
Collapse
|
44
|
Cheng Z, Lin P, Cheng N. HBV/HIV Coinfection: Impact on the Development and Clinical Treatment of Liver Diseases. Front Med (Lausanne) 2021; 8:713981. [PMID: 34676223 PMCID: PMC8524435 DOI: 10.3389/fmed.2021.713981] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/23/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatitis B virus (HBV) infection is a common contributor to chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Approximately 10% of people with human immunodeficiency virus (HIV) also have chronic HBV co-infection, owing to shared transmission routes. HIV/HBV coinfection accelerates the progression of chronic HBV to cirrhosis, end-stage liver disease, or hepatocellular carcinoma compared to chronic HBV mono-infection. HBV/HIV coinfection alters the natural history of hepatitis B and renders the antiviral treatment more complex. In this report, we conducted a critical review on the epidemiology, natural history, and pathogenesis of liver diseases related to HBV/HIV coinfection. We summarized the novel therapeutic options for these coinfected patients.
Collapse
Affiliation(s)
- Zhimeng Cheng
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Panpan Lin
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Nansheng Cheng
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Wang Z, Yin X, Ma M, Ge H, Lang B, Sun H, He S, Fu Y, Sun Y, Yu X, Zhang Z, Cui H, Han X, Xu J, Ding H, Chu Z, Shang H, Wu Y, Jiang Y. IP-10 Promotes Latent HIV Infection in Resting Memory CD4 + T Cells via LIMK-Cofilin Pathway. Front Immunol 2021; 12:656663. [PMID: 34447368 PMCID: PMC8383741 DOI: 10.3389/fimmu.2021.656663] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/15/2021] [Indexed: 12/31/2022] Open
Abstract
A major barrier to HIV eradication is the persistence of viral reservoirs. Resting CD4+ T cells are thought to be one of the major viral reservoirs, However, the underlying mechanism regulating HIV infection and the establishment of viral reservoir in T cells remain poorly understood. We have investigated the role of IP-10 in the establishment of HIV reservoirs in CD4+ T cells, and found that in HIV-infected individuals, plasma IP-10 was elevated, and positively correlated with HIV viral load and viral reservoir size. In addition, we found that binding of IP-10 to CXCR3 enhanced HIV latent infection of resting CD4+ T cells in vitro. Mechanistically, IP-10 stimulation promoted cofilin activity and actin dynamics, facilitating HIV entry and DNA integration. Moreover, treatment of resting CD4+ T cells with a LIM kinase inhibitor R10015 blocked cofilin phosphorylation and abrogated IP-10-mediated enhancement of HIV latent infection. These results suggest that IP-10 is a critical factor involved in HIV latent infection, and that therapeutic targeting of IP-10 may be a potential strategy for inhibiting HIV latent infection.
Collapse
Affiliation(s)
- Zhuo Wang
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China.,Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaowan Yin
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Meichen Ma
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongchi Ge
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bin Lang
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hong Sun
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Sijia He
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China.,National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Yajing Fu
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu Sun
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaowen Yu
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zining Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hualu Cui
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaoxu Han
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Junjie Xu
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Haibo Ding
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhenxing Chu
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuntao Wu
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Yongjun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
46
|
Anastasiou M, Newton GA, Kaur K, Carrillo-Salinas FJ, Smolgovsky SA, Bayer AL, Ilyukha V, Sharma S, Poltorak A, Luscinskas FW, Alcaide P. Endothelial STING controls T cell transmigration in an IFNI-dependent manner. JCI Insight 2021; 6:e149346. [PMID: 34156982 PMCID: PMC8410041 DOI: 10.1172/jci.insight.149346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
The stimulator of IFN genes (STING) protein senses cyclic dinucleotides released in response to double-stranded DNA and functions as an adaptor molecule for type I IFN (IFNI) signaling by activating IFNI-stimulated genes (ISG). We found impaired T cell infiltration into the peritoneum in response to TNF-α in global and EC-specific STING-/- mice and discovered that T cell transendothelial migration (TEM) across mouse and human endothelial cells (EC) deficient in STING was strikingly reduced compared with control EC, whereas T cell adhesion was not impaired. STING-/- T cells showed no defect in TEM or adhesion to EC, or immobilized endothelial cell-expressed molecules ICAM1 and VCAM1, compared with WT T cells. Mechanistically, CXCL10, an ISG and a chemoattractant for T cells, was dramatically reduced in TNF-α-stimulated STING-/- EC, and genetic loss or pharmacologic antagonisms of IFNI receptor (IFNAR) pathway reduced T cell TEM. Our data demonstrate a central role for EC-STING during T cell TEM that is dependent on the ISG CXCL10 and on IFNI/IFNAR signaling.
Collapse
Affiliation(s)
- Marina Anastasiou
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Department of Internal Medicine, University of Crete Medical School, Crete, Greece
| | - Gail A. Newton
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Kuljeet Kaur
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Sasha A. Smolgovsky
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Abraham L. Bayer
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Vladimir Ilyukha
- Petrozavodsk State University, Petrozavodsk, Republic of Karelia, Russia
| | - Shruti Sharma
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Alexander Poltorak
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA.,Petrozavodsk State University, Petrozavodsk, Republic of Karelia, Russia
| | - Francis W. Luscinskas
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Pandey V, Fleming-Martinez A, Bastea L, Doeppler HR, Eisenhauer J, Le T, Edenfield B, Storz P. CXCL10/CXCR3 signaling contributes to an inflammatory microenvironment and its blockade enhances progression of murine pancreatic precancerous lesions. eLife 2021; 10:60646. [PMID: 34328416 PMCID: PMC8360647 DOI: 10.7554/elife.60646] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/29/2021] [Indexed: 01/18/2023] Open
Abstract
The development of pancreatic cancer requires recruitment and activation of different macrophage populations. However, little is known about how macrophages are attracted to the pancreas after injury or an oncogenic event, and how they crosstalk with lesion cells or other cells of the lesion microenvironment. Here, we delineate the importance of CXCL10/CXCR3 signaling during the early phase of murine pancreatic cancer. We show that CXCL10 is produced by pancreatic precancerous lesion cells in response to IFNγ signaling and that inflammatory macrophages are recipients for this chemokine. CXCL10/CXCR3 signaling in macrophages mediates their chemoattraction to the pancreas, enhances their proliferation, and maintains their inflammatory identity. Blocking of CXCL10/CXCR3 signaling in vivo shifts macrophage populations to a tumor-promoting (Ym1+, Fizz+, Arg1+) phenotype, increases fibrosis, and mediates progression of lesions, highlighting the importance of this pathway in PDA development. This is reversed when CXCL10 is overexpressed in PanIN cells.
Collapse
Affiliation(s)
- Veethika Pandey
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Alicia Fleming-Martinez
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Heike R Doeppler
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Jillian Eisenhauer
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Tam Le
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Brandy Edenfield
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| |
Collapse
|
48
|
Solis-Castro OO, Wong N, Boissonade FM. Chemokines and Pain in the Trigeminal System. FRONTIERS IN PAIN RESEARCH 2021; 2:689314. [PMID: 35295531 PMCID: PMC8915704 DOI: 10.3389/fpain.2021.689314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
Chemotactic cytokines or chemokines are a large family of secreted proteins able to induce chemotaxis. Chemokines are categorized according to their primary amino acid sequence, and in particular their cysteine residues that form disulphide bonds to maintain the structure: CC, CXC, CX3C, and XC, in which X represents variable amino acids. Among their many roles, chemokines are known to be key players in pain modulation in the peripheral and central nervous systems. Thus, they are promising candidates for novel therapeutics that could replace current, often ineffective treatments. The spinal and trigeminal systems are intrinsically different beyond their anatomical location, and it has been suggested that there are also differences in their sensory mechanisms. Hence, understanding the different mechanisms involved in pain modulation for each system could aid in developing appropriate pharmacological alternatives. Here, we aim to describe the current landscape of chemokines that have been studied specifically with regard to trigeminal pain. Searching PubMed and Google Scholar, we identified 30 reports describing chemokines in animal models of trigeminal pain, and 15 reports describing chemokines involved in human pain associated with the trigeminal system. This review highlights the chemokines studied to date at different levels of the trigeminal system, their cellular localization and, where available, their role in a variety of animal pain models.
Collapse
Affiliation(s)
- Oscar O. Solis-Castro
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- The Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Natalie Wong
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- The Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Fiona M. Boissonade
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- The Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
- *Correspondence: Fiona M. Boissonade
| |
Collapse
|
49
|
Chen M, Brackett CM, Burdelya LG, Punnanitinont A, Patnaik SK, Matsuzaki J, Odunsi AO, Gudkov AV, Singh AK, Repasky EA, Gurova KV. Stimulation of an anti-tumor immune response with "chromatin-damaging" therapy. Cancer Immunol Immunother 2021; 70:2073-2086. [PMID: 33439292 PMCID: PMC8726059 DOI: 10.1007/s00262-020-02846-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023]
Abstract
Curaxins are small molecules that bind genomic DNA and interfere with DNA-histone interactions leading to the loss of histones and decondensation of chromatin. We named this phenomenon 'chromatin damage'. Curaxins demonstrated anti-cancer activity in multiple pre-clinical tumor models. Here, we present data which reveals, for the first time, a role for the immune system in the anti-cancer effects of curaxins. Using the lead curaxin, CBL0137, we observed elevated expression of several group of genes in CBL0137-treated tumor cells including interferon sensitive genes, MHC molecules, some embryo-specific antigens suggesting that CBL0137 increases tumor cell immunogenicity and improves recognition of tumor cells by the immune system. In support of this, we found that the anti-tumor activity of CBL0137 was reduced in immune deficient SCID mice when compared to immune competent mice. Anti-tumor activity of CBL0137 was abrogated in CD8+ T cell depleted mice but only partially lost when natural killer or CD4+ T cells were depleted. Further support for a key role for the immune system in the anti-tumor activity of CBL0137 is evidenced by an increased antigen-specific effector CD8+ T cell and NK cell response, and an increased ratio of effector T cells to Tregs in the tumor and spleen. CBL0137 also elevated the number of CXCR3-expressing CTLs in the tumor and the level of interferon-γ-inducible protein 10 (IP-10) in serum, suggesting IP-10/CXCR3 controls CBL0137-elicited recruitment of effector CTLs to tumors. Our collective data underscores a previously unrecognized role for both innate and adaptive immunity in the anti-tumor activity of curaxins.
Collapse
Affiliation(s)
- Minhui Chen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Craig M Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Lyudmila G Burdelya
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Achamaporn Punnanitinont
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Santosh K Patnaik
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Junko Matsuzaki
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Adekunle O Odunsi
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Anurag K Singh
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| | - Katerina V Gurova
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| |
Collapse
|
50
|
Extranodal NK/T-Cell Lymphoma, Nasal Type: Genetic, Biologic, and Clinical Aspects with a Central Focus on Epstein-Barr Virus Relation. Microorganisms 2021; 9:microorganisms9071381. [PMID: 34202088 PMCID: PMC8304202 DOI: 10.3390/microorganisms9071381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
Extranodal NK/T-Cell Lymphoma, nasal type (ENKTL-NT) has some salient aspects. The lymphoma is commonly seen in Eastern Asia, has progressive necrotic lesions in the nasal cavity, makes midfacial destructive lesions, and shows poor prognosis. The lymphoma cell is originated from either NK- or γδ T-cells, which express CD56. Since the authors first demonstrated the existence of Epstein–Barr virus (EBV) DNA and EBV oncogenic proteins in lymphoma cells, ENKTL-NT has been recognized as an EBV-associated malignancy. Because the angiocentric and polymorphous lymphoma cells are mixed with inflammatory cells on a necrotic background, the diagnosis of ENKTL-NT requires CD56 immunostaining and EBER in situ hybridization. In addition, serum the EBV DNA level is useful for the diagnosis and monitoring of ENKTL-NT. Although ENKTL-NT is refractory lymphoma, the prognosis is improved by the development of therapies such as concomitant chemoradiotherapy. The basic research reveals that a wide variety of intracellular/cell surface molecules, cytokines, chemokines, and micro RNAs are involved in lymphomagenesis, and some of them are related to EBV. Understanding lymphoma behavior introduces new therapeutic strategies, such as the usage of immune checkpoint inhibitors, peptide vaccines, and molecular targeting therapy. This review addresses recent advances in basic and clinical aspects of ENKTL-NT, especially its relation to EBV features.
Collapse
|