1
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [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: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
2
|
GOTO S, MIKAMI O, NAGASAWA Y, WATANABE A. Bovine neutrophils stimulated with Streptococcus uberis induce neutrophil extracellular traps, and cause cytotoxicity and transcriptional upregulation of inflammatory cytokine genes in bovine mammary epithelial cells. J Vet Med Sci 2024; 86:141-149. [PMID: 38104974 PMCID: PMC10898994 DOI: 10.1292/jvms.23-0302] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
This study aimed to understand the response of neutrophils stimulated by Streptococcus uberis, a major cause of mastitis. It was found that the production of neutrophil extracellular traps (NETs) was induced in milk clots from mastitic milk produced by S. uberis-infected bovine udders. The release of NETs from neutrophils stimulated by S. uberis was investigated. Bovine neutrophils cocultured with S. uberis in vitro released the components of NETs, which contained extracellular DNA and elastase. Bovine mammary epithelial cells (BMECs) incubated in coculture supernatants containing components of NETs, caused cytotoxicity and transcriptional upregulation of inflammatory cytokines, including of interleukin (IL) -1β, tumor necrosis factor (TNF)-α, IL-6, and IL-8, in BMECs. These findings suggest that bovine neutrophils stimulated by S. uberis induce responses that cause exacerbated inflammation, such as NET formation, cytotoxicity against BMECs, and increased production of inflammatory cytokines. Bovine neutrophil responses stimulated by S. uberis could be involved in the progression of S. uberis-induced mastitis.
Collapse
Affiliation(s)
- Shinya GOTO
- Pathology and Production Disease Group, Division of Hygiene
Management, National Institute of Animal Health, National Agriculture and Food Research
Organization, Hokkaido, Japan
| | - Osamu MIKAMI
- Pathology and Production Disease Group, Division of Hygiene
Management, National Institute of Animal Health, National Agriculture and Food Research
Organization, Hokkaido, Japan
| | - Yuya NAGASAWA
- Pathology and Production Disease Group, Division of Hygiene
Management, National Institute of Animal Health, National Agriculture and Food Research
Organization, Hokkaido, Japan
| | - Atsushi WATANABE
- Pathology and Production Disease Group, Division of Hygiene
Management, National Institute of Animal Health, National Agriculture and Food Research
Organization, Hokkaido, Japan
| |
Collapse
|
3
|
Xiong Z, Xu X, Zhang Y, Ma C, Hou C, You Z, Shu L, Ke Y, Liu Y. IFITM3 promotes glioblastoma stem cell-mediated angiogenesis via regulating JAK/STAT3/bFGF signaling pathway. Cell Death Dis 2024; 15:45. [PMID: 38218875 PMCID: PMC10787840 DOI: 10.1038/s41419-023-06416-5] [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: 06/05/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/15/2024]
Abstract
Interferon-induced transmembrane protein 3 (IFITM3) has been previously verified to be an endosomal protein that prevents viral infection. Recent findings suggested IFITM3 as a key factor in tumor invasion and progression. To clarify the role and molecular mechanism of IFITM3 in Glioblastoma multiforme (GBM) progression, we investigated the expression of IFITM3 in glioma datasets culled from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA). Primary GBM stem cells (GSCs) were cultured and identified in vitro. Loss-of-function and gain-of-function experiments were established by using shRNAs and lentiviral vectors targeting IFITM3. Co-culture system of GSCs and vascular endothelial cells was constructed in a Transwell chamber. Tube formation and spheroid-based angiogenesis assays were performed to determine the angiogenic capacity of endothelial cells. Results revealed that IFITM3 is elevated in GBM samples and predictive of adverse outcome. Mechanistically, GSCs-derived IFITM3 causes activation of Jak2/STAT3 signaling and leads to robust secretion of bFGF into tumor environment, which eventually results in enhanced angiogenesis. Taken together, these evidence indicated IFITM3 as an essential factor in GBM angiogenesis. Our findings provide a new insight into mechanism by which IFITM3 modulates GBM angiogenesis.
Collapse
Affiliation(s)
- Zhangsheng Xiong
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Xiangdong Xu
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Yuxuan Zhang
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Chengcheng Ma
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Chongxian Hou
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Zhongsheng You
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
- Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Guangzhou, PR China.
| | - Yiquan Ke
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China.
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China.
| | - Yang Liu
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China.
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China.
| |
Collapse
|
4
|
Lin J, Moradi E, Salenius K, Lehtipuro S, Häkkinen T, Laiho JE, Oikarinen S, Randelin S, Parikh HM, Krischer JP, Toppari J, Lernmark Å, Petrosino JF, Ajami NJ, She JX, Hagopian WA, Rewers MJ, Lloyd RE, Rautajoki KJ, Hyöty H, Nykter M. Distinct transcriptomic profiles in children prior to the appearance of type 1 diabetes-linked islet autoantibodies and following enterovirus infection. Nat Commun 2023; 14:7630. [PMID: 37993433 PMCID: PMC10665402 DOI: 10.1038/s41467-023-42763-9] [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: 02/01/2023] [Accepted: 10/17/2023] [Indexed: 11/24/2023] Open
Abstract
Although the genetic basis and pathogenesis of type 1 diabetes have been studied extensively, how host responses to environmental factors might contribute to autoantibody development remains largely unknown. Here, we use longitudinal blood transcriptome sequencing data to characterize host responses in children within 12 months prior to the appearance of type 1 diabetes-linked islet autoantibodies, as well as matched control children. We report that children who present with insulin-specific autoantibodies first have distinct transcriptional profiles from those who develop GADA autoantibodies first. In particular, gene dosage-driven expression of GSTM1 is associated with GADA autoantibody positivity. Moreover, compared with controls, we observe increased monocyte and decreased B cell proportions 9-12 months prior to autoantibody positivity, especially in children who developed antibodies against insulin first. Lastly, we show that control children present transcriptional signatures consistent with robust immune responses to enterovirus infection, whereas children who later developed islet autoimmunity do not. These findings highlight distinct immune-related transcriptomic differences between case and control children prior to case progression to islet autoimmunity and uncover deficient antiviral response in children who later develop islet autoimmunity.
Collapse
Grants
- U01 DK063821 NIDDK NIH HHS
- UC4 DK063863 NIDDK NIH HHS
- UL1 TR002535 NCATS NIH HHS
- U01 DK128847 NIDDK NIH HHS
- U01 DK063790 NIDDK NIH HHS
- UL1 TR000064 NCATS NIH HHS
- HHSN267200700014C NLM NIH HHS
- U01 DK063836 NIDDK NIH HHS
- U01 DK063829 NIDDK NIH HHS
- U01 DK063865 NIDDK NIH HHS
- UC4 DK095300 NIDDK NIH HHS
- UC4 DK063861 NIDDK NIH HHS
- UC4 DK063829 NIDDK NIH HHS
- UC4 DK063821 NIDDK NIH HHS
- UC4 DK117483 NIDDK NIH HHS
- UC4 DK063836 NIDDK NIH HHS
- UC4 DK112243 NIDDK NIH HHS
- U01 DK124166 NIDDK NIH HHS
- U01 DK063861 NIDDK NIH HHS
- UC4 DK063865 NIDDK NIH HHS
- U01 DK063863 NIDDK NIH HHS
- UC4 DK106955 NIDDK NIH HHS
- UC4 DK100238 NIDDK NIH HHS
- Academy of Finland (Suomen Akatemia)
- Sigrid Juséliuksen Säätiö (Sigrid Jusélius Foundation)
- U.S. Department of Health & Human Services | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (National Institute of Diabetes & Digestive & Kidney Diseases)
- EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- The TEDDY Study is funded by U01 DK63829, U01 DK63861, U01 DK63821, U01 DK63865, U01 DK63863, U01 DK63836, U01 DK63790, UC4 DK63829, UC4 DK63861, UC4 DK63821, UC4 DK63865, UC4 DK63863, UC4 DK63836, UC4 DK95300, UC4 DK100238, UC4 DK106955, UC4 DK112243, UC4 DK117483, U01 DK124166, U01 DK128847, and Contract No. HHSN267200700014C from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institute of Allergy and Infectious Diseases (NIAID), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institute of Environmental Health Sciences (NIEHS), Centers for Disease Control and Prevention (CDC), and JDRF. This work is supported in part by the NIH/NCATS Clinical and Translational Science Awards to the University of Florida (UL1 TR000064) and the University of Colorado (UL1 TR002535).
- Päivikki and Sakari Sohlberg's Foundation
Collapse
Affiliation(s)
- Jake Lin
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
- Biostatistics, Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
- Finnish Institute of Molecular Medicine, FIMM, University of Helsinki, 00290, Helsinki, Finland
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Elaheh Moradi
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, 70150, Finland
| | - Karoliina Salenius
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Suvi Lehtipuro
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Tomi Häkkinen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Jutta E Laiho
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sami Oikarinen
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sofia Randelin
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Hemang M Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, and Centre for Population Health Research, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Platform for Innovative Microbiome & Translational Research (PRIME-TR), Moon Shots™ Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jin-Xiong She
- Jinfiniti Precision Medicine, Inc., Augusta, GA, USA
| | - William A Hagopian
- Pacific Northwest Research Institute, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Richard E Lloyd
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kirsi J Rautajoki
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland.
| | - Heikki Hyöty
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland.
- Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| |
Collapse
|
5
|
Amino Acid Substitutions at P1 Position Change the Inhibitory Activity and Specificity of Protease Inhibitors BmSPI38 and BmSPI39 from Bombyx mori. Molecules 2023; 28:molecules28052073. [PMID: 36903318 PMCID: PMC10004685 DOI: 10.3390/molecules28052073] [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: 01/31/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
It was found that silkworm serine protease inhibitors BmSPI38 and BmSPI39 were very different from typical TIL-type protease inhibitors in sequence, structure, and activity. BmSPI38 and BmSPI39 with unique structure and activity may be good models for studying the relationship between the structure and function of small-molecule TIL-type protease inhibitors. In this study, site-directed saturation mutagenesis at the P1 position was conducted to investigate the effect of P1 sites on the inhibitory activity and specificity of BmSPI38 and BmSPI39. In-gel activity staining and protease inhibition experiments confirmed that BmSPI38 and BmSPI39 could strongly inhibit elastase activity. Almost all mutant proteins of BmSPI38 and BmSPI39 retained the inhibitory activities against subtilisin and elastase, but the replacement of P1 residues greatly affected their intrinsic inhibitory activities. Overall, the substitution of Gly54 in BmSPI38 and Ala56 in BmSPI39 with Gln, Ser, or Thr was able to significantly enhance their inhibitory activities against subtilisin and elastase. However, replacing P1 residues in BmSPI38 and BmSPI39 with Ile, Trp, Pro, or Val could seriously weaken their inhibitory activity against subtilisin and elastase. The replacement of P1 residues with Arg or Lys not only reduced the intrinsic activities of BmSPI38 and BmSPI39, but also resulted in the acquisition of stronger trypsin inhibitory activities and weaker chymotrypsin inhibitory activities. The activity staining results showed that BmSPI38(G54K), BmSPI39(A56R), and BmSPI39(A56K) had extremely high acid-base and thermal stability. In conclusion, this study not only confirmed that BmSPI38 and BmSPI39 had strong elastase inhibitory activity, but also confirmed that P1 residue replacement could change their activity and inhibitory specificity. This not only provides a new perspective and idea for the exploitation and utilization of BmSPI38 and BmSPI39 in biomedicine and pest control, but also provides a basis or reference for the activity and specificity modification of TIL-type protease inhibitors.
Collapse
|
6
|
Petrelli A, Popp SK, Fukuda R, Parish CR, Bosi E, Simeonovic CJ. The Contribution of Neutrophils and NETs to the Development of Type 1 Diabetes. Front Immunol 2022; 13:930553. [PMID: 35874740 PMCID: PMC9299437 DOI: 10.3389/fimmu.2022.930553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease resulting from the destruction of insulin-producing beta cells in pancreatic islets. T lymphocytes are the claimed pathogenic effectors but abnormalities of other immune cell types, including neutrophils, also characterize T1D development. During human T1D natural history, neutrophils are reduced in the circulation, while accumulate in the pancreas where release of neutrophil extracellular traps (NETs), or NETosis, is manifest. Recent-onset T1D patients also demonstrate activated circulating neutrophils, associated with a unique neutrophil gene signature. Neutrophils can bind to platelets, leading to the formation of platelet-neutrophil aggregates (PNAs). PNAs increase in the circulation during the development of human T1D and provide a mechanism for neutrophil activation and mobilization/recruitment to the pancreas. In non-obese diabetic or NOD mice, T1D autoimmunity is accompanied by dynamic changes in neutrophil numbers, activation state, PNAs and/or NETosis/NET proteins in the circulation, pancreas and/or islets. Such properties differ between stages of T1D disease and underpin potentially indirect and direct impacts of the innate immune system in T1D pathogenesis. Supporting the potential for a pathogenic role in T1D, NETs and extracellular histones can directly damage isolated islets in vitro, a toxicity that can be prevented by small polyanions. In human T1D, NET-related damage can target the whole pancreas, including both the endocrine and exocrine components, and contribute to beta cell destruction, providing evidence for a neutrophil-associated T1D endotype. Future intervention in T1D could therefore benefit from combined strategies targeting T cells and accessory destructive elements of activated neutrophils.
Collapse
Affiliation(s)
- Alessandra Petrelli
- San Raffaele Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Sarah K Popp
- Immunology and Infectious Disease Division, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riho Fukuda
- Immunology and Infectious Disease Division, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,Department of Medicine, Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Christopher R Parish
- Genome Sciences and Cancer Division, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy.,Department of Medicine, San Raffaele Vita Salute University, Milan, Italy
| | - Charmaine J Simeonovic
- Immunology and Infectious Disease Division, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
7
|
Uribe-Querol E, Rosales C. Neutrophils Actively Contribute to Obesity-Associated Inflammation and Pathological Complications. Cells 2022; 11:1883. [PMID: 35741012 PMCID: PMC9221045 DOI: 10.3390/cells11121883] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is characterized by an increase in body weight associated with an exaggerated enlargement of the adipose tissue. Obesity has serious negative effects because it is associated with multiple pathological complications such as type 2 diabetes mellitus, cardiovascular diseases, cancer, and COVID-19. Nowadays, 39% of the world population is obese or overweight, making obesity the 21st century epidemic. Obesity is also characterized by a mild, chronic, systemic inflammation. Accumulation of fat in adipose tissue causes stress and malfunction of adipocytes, which then initiate inflammation. Next, adipose tissue is infiltrated by cells of the innate immune system. Recently, it has become evident that neutrophils, the most abundant leukocytes in blood, are the first immune cells infiltrating the adipose tissue. Neutrophils then get activated and release inflammatory factors that recruit macrophages and other immune cells. These immune cells, in turn, perpetuate the inflammation state by producing cytokines and chemokines that can reach other parts of the body, creating a systemic inflammatory condition. In this review, we described the recent findings on the role of neutrophils during obesity and the initiation of inflammation. In addition, we discuss the involvement of neutrophils in the generation of obesity-related complications using diabetes as a prime example.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- Laboratorio de Biología del Desarrollo, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
8
|
Shen Y, You Q, Wu Y, Wu J. Inhibition of PAD4-mediated NET formation by cl-amidine prevents diabetes development in nonobese diabetic mice. Eur J Pharmacol 2022; 916:174623. [PMID: 34767782 DOI: 10.1016/j.ejphar.2021.174623] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022]
Abstract
Many evidences indicated that neutrophil extracellular traps (NETs) play pathogenic roles in type 1 diabetes (T1D). Peptidylarginine deiminases 4 (PAD4) has been proved to be indispensable for generation of NETs. In the current study, we investigated whether oral administration of cl-amidine, an effective inhibitor of PAD4, protects non-obese diabetic (NOD) mice from T1D development. Female NOD mice were orally administrated with cl-amidine (5 μg/g body weight) from the age of 8 weeks up to 16 weeks. It showed that cl-amidine inhibit NET formation in vitro and in vivo. The onset of T1D was delayed nearly 8 weeks and the incidence of disease was significantly decreased in cl-amidine treated mice compared with the control group. Moreover, cl-amidine decreased the serum levels of anti-citrullinated peptide antibody (ACPA) and anti-neutrophil cytoplasmic antibodies (ANCA) in NOD mice. Also, it decreased generation of T1D autoantibodies such as glutamic acid decarboxylase antibody (GADA), tyrosine phosphatase-related islet antigen-2 antibody (IA2A) and zinc transporter 8 antibody (ZnT8A), which were strongly correlated with the reduced serum PAD4 and MPO-DNA levels. Furthermore, cl-amidine administration inhibited pancreatic inflammation and increased frequency of regulatory T cells in pancreatic lymph nodes (PLNs). In addition, cl-amidine improved gut barrier dysfunction and decreased the serum level of lipopolysaccharide (LPS), which was positively correlated with the NETs markers (PAD4 and MPO-DNA) and T1D autoantibody IA2A. In conclusion, our data showed that orally delivery of cl-amidine effectively prevent T1D development and suggested inhibition of PAD4-dependent NET formation as a potential way of clinical treatment in T1D.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Autoantibodies/blood
- Autoimmune Diseases/prevention & control
- Blood Glucose/drug effects
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Extracellular Traps/drug effects
- Female
- Inflammation/prevention & control
- Intestines/drug effects
- Mice, Inbred NOD
- Ornithine/administration & dosage
- Ornithine/analogs & derivatives
- Ornithine/pharmacology
- Protective Agents/administration & dosage
- Protective Agents/pharmacology
- Protein-Arginine Deiminase Type 4/antagonists & inhibitors
- Protein-Arginine Deiminase Type 4/blood
- Protein-Arginine Deiminase Type 4/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
- Tight Junctions/drug effects
- Mice
Collapse
Affiliation(s)
- Yiming Shen
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qi You
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yiling Wu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jie Wu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
9
|
Herrero-Cervera A, Soehnlein O, Kenne E. Neutrophils in chronic inflammatory diseases. Cell Mol Immunol 2022; 19:177-191. [PMID: 35039631 PMCID: PMC8803838 DOI: 10.1038/s41423-021-00832-3] [Citation(s) in RCA: 264] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is a component of many disease conditions that affect a large group of individuals worldwide. Chronic inflammation is characterized by persistent, low-grade inflammation and is increased in the aging population. Neutrophils are normally the first responders to acute inflammation and contribute to the resolution of inflammation. However, in chronic inflammation, the role of neutrophils is less well understood and has been described as either beneficial or detrimental, causing tissue damage and enhancing the immune response. Emerging evidence suggests that neutrophils are important players in several chronic diseases, such as atherosclerosis, diabetes mellitus, nonalcoholic fatty liver disease and autoimmune disorders. This review will highlight the interaction of neutrophils with other cells in the context of chronic inflammation, the contribution of neutrophils to selected chronic inflammatory diseases, and possible future therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Herrero-Cervera
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Popp SK, Vecchio F, Brown DJ, Fukuda R, Suzuki Y, Takeda Y, Wakamatsu R, Sarma MA, Garrett J, Giovenzana A, Bosi E, Lafferty AR, Brown KJ, Gardiner EE, Coupland LA, Thomas HE, Chong BH, Parish CR, Battaglia M, Petrelli A, Simeonovic CJ. Circulating platelet-neutrophil aggregates characterize the development of type 1 diabetes in humans and NOD mice. JCI Insight 2022; 7:153993. [PMID: 35076023 PMCID: PMC8855805 DOI: 10.1172/jci.insight.153993] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/01/2021] [Indexed: 12/19/2022] Open
Abstract
Platelet-neutrophil aggregates (PNAs) facilitate neutrophil activation and migration and could underpin the recruitment of neutrophils to the pancreas during type 1 diabetes (T1D) pathogenesis. PNAs, measured by flow cytometry, were significantly elevated in the circulation of autoantibody-positive (Aab+) children and new-onset T1D children, as well as in pre-T1D (at 4 weeks and 10–12 weeks) and T1D-onset NOD mice, compared with relevant controls, and PNAs were characterized by activated P-selectin+ platelets. PNAs were similarly increased in pre-T1D and T1D-onset NOD isolated islets/insulitis, and immunofluorescence staining revealed increased islet-associated neutrophil extracellular trap (NET) products (myeloperoxidase [MPO] and citrullinated histones [CitH3]) in NOD pancreata. In vitro, cell-free histones and NETs induced islet cell damage, which was prevented by the small polyanionic drug methyl cellobiose sulfate (mCBS) that binds to histones and neutralizes their pathological effects. Elevated circulating PNAs could, therefore, act as an innate immune and pathogenic biomarker of T1D autoimmunity. Platelet hyperreactivity within PNAs appears to represent a previously unrecognized hematological abnormality that precedes T1D onset. In summary, PNAs could contribute to the pathogenesis of T1D and potentially function as a pre-T1D diagnostic.
Collapse
Affiliation(s)
- Sarah K. Popp
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| | - Federica Vecchio
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Debra J. Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| | - Riho Fukuda
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Yuri Suzuki
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Yuma Takeda
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Rikako Wakamatsu
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Mahalakshmi A. Sarma
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| | - Jessica Garrett
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Anna Giovenzana
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- San Raffaele Vita Salute University, Milan, Italy
| | - Antony R.A. Lafferty
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Department of Pediatrics, The Canberra Hospital, Canberra, Australia
| | - Karen J. Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Department of Pediatrics, The Canberra Hospital, Canberra, Australia
| | - Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Lucy A. Coupland
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Helen E. Thomas
- St. Vincent’s Institute of Medical Research, Melbourne, Australia
| | - Beng H. Chong
- Hematology Research Unit, St. George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Christopher R. Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Manuela Battaglia
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Charmaine J. Simeonovic
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| |
Collapse
|
11
|
Hu J, Zhang R, Zou H, Xie L, Zhou Z, Xiao Y. Latent Autoimmune Diabetes in Adults (LADA): From Immunopathogenesis to Immunotherapy. Front Endocrinol (Lausanne) 2022; 13:917169. [PMID: 35937817 PMCID: PMC9350734 DOI: 10.3389/fendo.2022.917169] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/26/2022] [Indexed: 11/26/2022] Open
Abstract
Latent autoimmune diabetes in adults (LADA) is a type of diabetes characterized by slow autoimmune damage of pancreatic β cells without insulin treatment in the early clinical stage. There are differences between LADA and classical type 1 diabetes (T1D) and type 2 diabetes (T2D) in genetic background, autoimmune response, rate of islet function decline, clinical metabolic characteristics, and so on. The disease progression and drug response of patients with LADA are closely related to the level of islet autoimmunity, thus exploring the pathogenesis of LADA is of great significance for its prevention and treatment. Previous studies reported that adaptive immunity and innate immunity play a critical role in the etiology of LADA. Recent studies have shown that the intestinal microbiota which impacts host immunity hugely, participates in the pathogenesis of LADA. In addition, the progression of autoimmune pancreatic β cell destruction in LADA is slower than in classical T1D, providing a wider window of opportunities for intervention. Therefore, therapies including antidiabetic drugs with immune-regulation effects and immunomodulators could contribute to promising interventions for LADA. We also shed light on potential interventions targeting the gut microbiota and gut-associated immunity, which may be envisaged to halt or delay the process of autoimmunity in LADA.
Collapse
|
12
|
Giovenzana A, Carnovale D, Phillips B, Petrelli A, Giannoukakis N. Neutrophils and their role in the aetiopathogenesis of type 1 and type 2 diabetes. Diabetes Metab Res Rev 2022; 38:e3483. [PMID: 34245096 DOI: 10.1002/dmrr.3483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 05/12/2021] [Accepted: 06/19/2021] [Indexed: 12/25/2022]
Abstract
Multiple and complex aetiological processes underlie diabetes mellitus, which invariably result in the development of hyperglycaemia. Although there are two prevalent distinct forms of the disease, that is, type 1 and type 2 diabetes, accumulating evidence indicates that these syndromes share more aetiopathological mechanisms than originally thought. This compels a rethinking of the approaches to prevent and treat the different manifestations of what eventually becomes a hyperglycaemic state. This review aims to address the involvement of neutrophils, the most abundant type of granulocytes involved in the initiation of the acute phase of inflammation, in the aetiopathogenesis of diabetes mellitus, with a focus on type 1 and type 2 diabetes. We review the evidence that neutrophils are the first leucocytes to react to and accumulate inside target tissues of diabetes, such as the pancreas and insulin-sensitive tissues. We then review available data on the role of neutrophils and their functional alteration, with a focus on NETosis, in the progression towards clinical disease. Finally, we review potential approaches as secondary and adjunctive treatments to limit neutrophil-mediated damage in the prevention of the progression of subclinical disease to clinical hyperglycaemia.
Collapse
Affiliation(s)
- Anna Giovenzana
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
| | - Debora Carnovale
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
| | - Brett Phillips
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
| | - Nick Giannoukakis
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Nichols BE, Hook JS, Weng K, Ahn C, Moreland JG. Novel neutrophil phenotypic signature in pediatric patients with type 1 diabetes and diabetic ketoacidosis. J Leukoc Biol 2021; 111:849-856. [PMID: 34342036 DOI: 10.1002/jlb.3a1220-826r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic inflammatory condition sometimes complicated by acute diabetic ketoacidosis (DKA). A subset of patients with T1D develop DKA independent of known risk factors. This study tested the hypothesis that circulating polymorphonuclear leukocytes (PMN) from children with T1D and DKA would exhibit a primed phenotype and that the signature would be unique in patients predisposed to have DKA. Using a prospective cohort study design, neutrophil phenotype was assessed in 30 patients with T1D seen in endocrinology clinic for routine care, 30 patients with acute DKA, and 36 healthy donors. Circulating PMN from patients with DKA display a primed phenotype with increased basal cell-surface CD11b, L-selectin shedding, and enhanced fMLF-elicited reactive oxygen species (ROS) production. Moreover, PMN from T1D patients both with and without DKA lack the capacity to be further primed by incubation with TNF-α, a classic priming stimulus. Primed PMN phenotypic signatures demonstrated are independent of hemoglobin A1c, the premier biological marker for DKA risk, and are consistent with a hyperinflammatory state. A single nucleotide polymorphism in TLR-1 (1805G>T), known to be associated with a hyperinflammatory PMN phenotype, correlated with DKA. This study elucidated a novel phenotypic signature in circulating PMN from children with T1D with and without DKA, and suggests the possibility of a previously unrecognized PMN phenotype with potential clinical implications. Immunophenotype and genotype may be applicable as biomarkers for DKA risk stratification in patients with T1D.
Collapse
Affiliation(s)
- Blake E Nichols
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica S Hook
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kayson Weng
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chul Ahn
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica G Moreland
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
14
|
Sodré FMC, Bissenova S, Bruggeman Y, Tilvawala R, Cook DP, Berthault C, Mondal S, Callebaut A, You S, Scharfmann R, Mallone R, Thompson PR, Mathieu C, Buitinga M, Overbergh L. Peptidylarginine Deiminase Inhibition Prevents Diabetes Development in NOD Mice. Diabetes 2021; 70:516-528. [PMID: 33203696 PMCID: PMC7881854 DOI: 10.2337/db20-0421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Protein citrullination plays a role in several autoimmune diseases. Its involvement in murine and human type 1 diabetes has recently been recognized through the discovery of antibodies and T-cell reactivity against citrullinated peptides. In the current study, we demonstrate that systemic inhibition of peptidylarginine deiminases (PADs), the enzymes mediating citrullination, through BB-Cl-amidine treatment, prevents diabetes development in NOD mice. This prevention was associated with reduced levels of citrullination in the pancreas, decreased circulating autoantibody titers against citrullinated glucose-regulated protein 78, and reduced spontaneous neutrophil extracellular trap formation of bone marrow-derived neutrophils. Moreover, BB-Cl-amidine treatment induced a shift from Th1 to Th2 cytokines in the serum and an increase in the frequency of regulatory T cells in the blood and spleen. In the pancreas, BB-Cl-amidine treatment preserved insulin production and was associated with a less destructive immune infiltrate characterized by reduced frequencies of effector memory CD4+ T cells and a modest reduction in the frequency of interferon-γ-producing CD4+ and CD8+ T cells. Our results point to a role of citrullination in the pathogenesis of autoimmune diabetes, with PAD inhibition leading to disease prevention through modulation of immune pathways. These findings provide insight in the potential of PAD inhibition for treating autoimmune diseases like type 1 diabetes.
Collapse
Affiliation(s)
- Fernanda M C Sodré
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Samal Bissenova
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Ylke Bruggeman
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Ronak Tilvawala
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS
| | - Dana P Cook
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Claire Berthault
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Santanu Mondal
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Aïsha Callebaut
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Sylvaine You
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | | | - Roberto Mallone
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires de Paris Centre-Université de Paris, Cochin Hospital, Service de Diabétologie et Immunologie Clinique, Paris, France
| | - Paul R Thompson
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Chantal Mathieu
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Mijke Buitinga
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
McNulty MJ, Silberstein DZ, Kuhn BT, Padgett HS, Nandi S, McDonald KA, Cross CE. Alpha-1 antitrypsin deficiency and recombinant protein sources with focus on plant sources: Updates, challenges and perspectives. Free Radic Biol Med 2021; 163:10-30. [PMID: 33279618 DOI: 10.1016/j.freeradbiomed.2020.11.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Alpha-1 antitrypsin deficiency (A1ATD) is an autosomal recessive disease characterized by low plasma levels of A1AT, a serine protease inhibitor representing the most abundant circulating antiprotease normally present at plasma levels of 1-2 g/L. The dominant clinical manifestations include predispositions to early onset emphysema due to protease/antiprotease imbalance in distal lung parenchyma and liver disease largely due to unsecreted polymerized accumulations of misfolded mutant A1AT within the endoplasmic reticulum of hepatocytes. Since 1987, the only FDA licensed specific therapy for the emphysema component has been infusions of A1AT purified from pooled human plasma at the 2020 cost of up to US $200,000/year with the risk of intermittent shortages. In the past three decades various, potentially less expensive, recombinant forms of human A1AT have reached early stages of development, one of which is just reaching the stage of human clinical trials. The focus of this review is to update strategies for the treatment of the pulmonary component of A1ATD with some focus on perspectives for therapeutic production and regulatory approval of a recombinant product from plants. We review other competitive technologies for treating the lung disease manifestations of A1ATD, highlight strategies for the generation of data potentially helpful for securing FDA Investigational New Drug (IND) approval and present challenges in the selection of clinical trial strategies required for FDA licensing of a New Drug Approval (NDA) for this disease.
Collapse
Affiliation(s)
- Matthew J McNulty
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - David Z Silberstein
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Brooks T Kuhn
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA
| | | | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Karen A McDonald
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Carroll E Cross
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA; Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.
| |
Collapse
|