1
|
Halpern D, Farber I, Anav Y, Tsitrina A, Lewis EC, Silberstein E. Alpha-1-Antitrypsin Enhances Fat Graft Survival in a Murine Model. Adv Wound Care (New Rochelle) 2025. [PMID: 40105893 DOI: 10.1089/wound.2024.0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Objective: Fat grafting is widely applied for various purposes, including volume restoration, improving tissue quality, and promoting wound healing, but it has poor long-term graft survival predictability. Alpha-1-antitrypsin (AAT) administration is hypothesized to improve fat graft outcomes by expediting inflammatory resolution and graft vascularity and reducing necrosis. Approach: Mice heterozygote to human AAT was grafted fat under the scalp alongside 400 µg/graft AAT or albumin (ALB) on days 0 and 3. Graft volume was determined by micro-magnetic resonance imaging, and explants were assessed for viability, histology, immunohistochemistry, and expression of selected genes. AAT expression was examined in hypoxia-exposed adipose-derived stem cells (ADSCs). Results: After 90 days, AAT-treated grafts maintained higher volumes (70.06% vs. 34.54%, n = 8, p = 0.02) and displayed improved tissue quality. On day 10 after grafting, grafts exhibited more blood vessels (mean 1.94/mm2 vs. 0.33/mm2) and 6.25-fold more adiponectin transcript levels (n = 12, p = 0.02). Although day-3 interleukin (IL)-1β expression was 5-fold greater in AAT-treated grafts (n = 6, p = 0.4), day-10 IL-1β expression was 2-fold lower compared to ALB-treated grafts (n = 22, p = 0.01). In the Methoxynitrosulfophenyl-tetrazolium carboxanilide (XTT) assay, day-3 AAT-treated grafts were 1.56-fold more metabolically functional (n = 6, p = 0.04) and exhibited greater perilipin-positive regions (18.5% versus 3.1%). Hypoxia-exposed ADSC expressed 9-fold higher AAT transcript levels (p = 0.04). Innovation: Fat grafting outcomes improved by early AAT treatment, probably by accelerating inflammatory resolution. Due to its marked safety profile, the study's findings are for adjunct clinical-grade AAT therapy. Conclusion: AAT has a promising potential to be utilized as a fat graft outcome enhancer in terms of volume retention predictability and tissue quality.
Collapse
Affiliation(s)
- Dor Halpern
- Plastic and Reconstructive Surgery Department, Soroka University Medical Centre, Beer Sheva, Israel
| | - Idan Farber
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Yuval Anav
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
- Joyce and Irving Goldman Medical School, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Alexandra Tsitrina
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev,Beer Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Eldad Silberstein
- Plastic and Reconstructive Surgery Department, Soroka University Medical Centre, Beer Sheva, Israel
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
2
|
Erion DM, Liu LY, Brown CR, Rennard S, Farah H. Editing Approaches to Treat Alpha-1 Antitrypsin Deficiency. Chest 2025; 167:444-452. [PMID: 39401571 DOI: 10.1016/j.chest.2024.09.038] [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: 05/10/2024] [Revised: 09/03/2024] [Accepted: 09/27/2024] [Indexed: 11/09/2024] Open
Abstract
TOPIC IMPORTANCE Alpha-1 antitrypsin (AAT) deficiency is a genetic disorder most commonly due to a single G to A point mutation (E342K), leading to debilitating lung and/or liver disorders and is associated with increased mortality. The E342K point mutation causes a conformational change of the AAT protein resulting in its retention in liver hepatocytes. This reduces AAT secretion into the serum resulting in higher protease activities due to the lack of inhibition from AAT, causing damage to healthy lung tissue. The current standard of care for lung manifestations involves weekly IV augmentation therapy and is considered suboptimal for these patients. Furthermore, there is currently no approved treatment for liver manifestations. The unmet medical need for patients with AAT deficiency remains high, and new treatment options are needed to treat the underlying disease etiology. REVIEW FINDINGS Advances in genomic medicines may enable treatment by editing the DNA or RNA sequence to produce wild-type AAT instead of the mutated AAT caused by the E342K mutation. One approach can be achieved by directing endogenous adenosine deaminases that act on RNA to the E342K RNA site, where they catalyze adenosine to inosine conversion through a process known as RNA editing. The A-I RNA change will be read as a G during protein translation, resulting in an altered amino acid and restoration of wild-type AAT secretion and function. SUMMARY In this review, we will discuss the pathophysiology of AAT deficiency and emerging treatment options with particular focus on RNA editing as a disease-modifying treatment for both liver and lung disease.
Collapse
Affiliation(s)
| | | | | | - Stephen Rennard
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
| | | |
Collapse
|
3
|
Annunziata A, Fiorentino G, Coppola A, Cauteruccio R, Ferrentino L, Fiorentino L, Calabrese C. Alpha-1 antitripsyn deficiency and augmentation therapy in pregnancy: two case reports. Front Med (Lausanne) 2024; 11:1479877. [PMID: 39736979 PMCID: PMC11682898 DOI: 10.3389/fmed.2024.1479877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is an inherited condition characterized by reduced plasma levels of alpha-1 antitrypsin (AAT), often leading to pulmonary diseases primarily emphysema and/or chronic obstructive pulmonary disease (COPD), but also bronchiectasis, bronchial asthma, or other less common disorders. Early diagnosis enables AAT augmentation therapy, which has proven to be effective in slowing down functional decline and improving survival rates. This article presents two cases of pregnant women with rare allelic variants of AATD who received AAT augmentation therapy, exploring the limited evidence on its safety during pregnancy and the potential role of decreased serum AAT levels in pregnancy-related complications.
Collapse
Affiliation(s)
- Anna Annunziata
- Department of Intensive Care, Unit of Pathophysiology and Respiratory Rehabilitation, AORN Ospedali dei Colli, Naples, Italy
| | - Giuseppe Fiorentino
- Department of Intensive Care, Unit of Pathophysiology and Respiratory Rehabilitation, AORN Ospedali dei Colli, Naples, Italy
| | - Antonietta Coppola
- Department of Intensive Care, Unit of Pathophysiology and Respiratory Rehabilitation, AORN Ospedali dei Colli, Naples, Italy
| | - Rosa Cauteruccio
- Department of Intensive Care, Unit of Pathophysiology and Respiratory Rehabilitation, AORN Ospedali dei Colli, Naples, Italy
| | - Laura Ferrentino
- Department of Translational Medical Sciences, University Federico II, Naples, Italy
| | - Luigi Fiorentino
- Department of Intensive Care, Unit of Pathophysiology and Respiratory Rehabilitation, AORN Ospedali dei Colli, Naples, Italy
| | - Cecilia Calabrese
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, AORN Ospedali dei Colli, Naples, Italy
| |
Collapse
|
4
|
Feitosa PHR, Castellano MVCDO, Costa CHD, Cardoso ADRO, Pereira LFF, Fernandes FLA, Costa FM, Felisbino MB, Oliveira AFFD, Jardim JR, Miravitlles M. Recommendations for the diagnosis and treatment of alpha-1 antitrypsin deficiency. J Bras Pneumol 2024; 50:e20240235. [PMID: 39661838 PMCID: PMC11601085 DOI: 10.36416/1806-3756/e20240235] [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: 07/15/2024] [Accepted: 08/02/2024] [Indexed: 12/13/2024] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a relatively rare genetic disorder, inherited in an autosomal codominant manner, that results in reduced serum AAT concentrations, with a consequent reduction in antielastase activity in the lungs, as well as an increased risk of diseases such as pulmonary emphysema, liver cirrhosis, and necrotizing panniculitis. It results from different mutations in the SERPINA1 gene, leading to changes in the AAT glycoprotein, which can alter its concentration, conformation, and function. Unfortunately, underdiagnosis is quite common; it is possible that only 10% of cases are diagnosed. The most common deficiency is in the Z variant, and it is estimated that more than 3 million people worldwide have combinations of alleles associated with severe AATD. Serum AAT concentrations should be determined, and allelic variants should be identified by phenotyping or genotyping. Monitoring lung function, especially through spirometry, is essential, because it provides information on the progression of the disease. Although pulmonary densitometry appears to be the most sensitive measure of emphysema progression, it should not be used in routine clinical practice to monitor patients. In general, the treatment is similar to that indicated for patients with COPD not caused by AATD. Exogenous administration of purified human serum-derived AAT is the only specific treatment approved for AATD in nonsmoking patients with severe deficiency (serum AAT concentration of < 57 mg/dL or < 11 µM), with evidence of functional loss above the physiological level.
Collapse
Affiliation(s)
| | | | | | | | | | - Frederico Leon Arrabal Fernandes
- . Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo ( SP) Brasil
| | - Fábio Marcelo Costa
- . Complexo Hospital das Clínicas, Universidade Federal do Paraná - CHC-UFPR - Curitiba (PR) Brasil
| | - Manuela Brisot Felisbino
- . Hospital Universitário, Universidade Federal de Santa Catarina - HU-UFSC - Florianópolis (SC) Brasil
| | | | - Jose R Jardim
- . Universidade Federal de São Paulo, São Paulo (SP) Brasil
| | - Marc Miravitlles
- . Vall d'Hebron Institut de Recerca - VHIR - Hospital Universitário Valld'Hebron, Barcelona, España
| |
Collapse
|
5
|
González-Carracedo MA, Herrera-Luis E, Marco-Simancas M, Escuela-Escobar A, Martín-González E, Sardón-Prado O, Corcuera P, Hernández-Pérez JM, Lorenzo-Díaz F, Pérez-Pérez JA. Haplotype-Aware Detection of SERPINA1 Variants by Nanopore Sequencing. J Mol Diagn 2024; 26:971-987. [PMID: 39276924 DOI: 10.1016/j.jmoldx.2024.08.002] [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: 05/04/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 09/17/2024] Open
Abstract
α-1 Antitrypsin (AAT) is an acute-phase reactant with immunomodulatory properties that mainly inhibits neutrophil elastase. Low serum levels cause AAT deficiency (AATD), an underdiagnosed condition that predisposes to pulmonary and hepatic diseases. The SERPINA1 gene, which encodes AAT, contains >500 variants. PI∗Z and PI∗S alleles are the most diagnosed causes of AATD, but the role of the SERPINA1 haplotypes in AAT function remains unknown. SERPINA1 gene was PCR amplified from 94 patients with asthma, using primers with tails for indexing. Sequencing libraries were loaded into a MinION-Mk1C, and MinKNOW was used for basecalling and demultiplexing. Nanofilt and Minimap2 were used for filtering and mapping/alignment. Variant calling/phasing were performed with PEPPER-Margin-DeepVariant. SERPINA1 gene was 100% covered for all samples, with a minimum sequencing depth of 500×. A total of 75 single-nucleotide variants (SNVs) and 4 insertions/deletions were detected, with 45 and 2 of them highly polymorphic (minor allele frequency >0.1), respectively. Nine of the SNVs showed differences in allele frequencies when compared with the overall Spanish population. More than 90% of heterozygous SNVs were phased, yielding 91 and 58 different haplotypes for each SERPINA1 amplified region. Haplotype-based linkage disequilibrium analysis suggests that a recombination hotspot could generate variation in the SERPINA1 gene. The proposed workflow enables haplotype-aware genotyping of the SERPINA1 gene by nanopore sequencing, which will allow the development of novel AATD diagnostic strategies.
Collapse
Affiliation(s)
- Mario A González-Carracedo
- Genetics Laboratory, Institute of Tropical Diseases and Public Health of the Canary Islands, Universidad de La Laguna, Tenerife, Spain; Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Tenerife, Spain
| | - Esther Herrera-Luis
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - María Marco-Simancas
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Tenerife, Spain
| | - Ainhoa Escuela-Escobar
- Genetics Laboratory, Institute of Tropical Diseases and Public Health of the Canary Islands, Universidad de La Laguna, Tenerife, Spain
| | - Elena Martín-González
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Tenerife, Spain
| | - Olaia Sardón-Prado
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain; Department of Pediatrics, University of the Basque Country, San Sebastián, Spain
| | - Paula Corcuera
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
| | - Jose M Hernández-Pérez
- Department of Respiratory Medicine, Hospital Universitario de N.S. de Candelaria, Tenerife, Spain
| | - Fabián Lorenzo-Díaz
- Genetics Laboratory, Institute of Tropical Diseases and Public Health of the Canary Islands, Universidad de La Laguna, Tenerife, Spain; Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Tenerife, Spain
| | - José A Pérez-Pérez
- Genetics Laboratory, Institute of Tropical Diseases and Public Health of the Canary Islands, Universidad de La Laguna, Tenerife, Spain; Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
6
|
Jochmans I, Lerut E, Monbaliu D, Pirenne J. Impact of a Single Dose of Alpha-1-Antitrypsin in a Rat Model of Bilateral Kidney Ischemia Reperfusion Injury. J Surg Res 2024; 299:179-187. [PMID: 38759334 DOI: 10.1016/j.jss.2024.04.023] [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: 08/20/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/19/2024]
Abstract
INTRODUCTION Renal ischemia reperfusion injury is a major cause of perioperative acute kidney injury. Alpha-1-antitrypsin (AAT), a protease inhibitor, might improve outcomes by reducing inflammation and apoptosis. We investigated the effects of a single intravenous dose of AAT immediately before ischemia in a rat bilateral renal clamping model. METHODS Both renal pedicles of male Sprague-Dawley rats were clamped (45 min). Plasma and renal tissue were collected at 3 h, 24 h, and 7 d. Intravenous AAT (60 mg/kg) was administered 5 min before clamping. Controls received saline. Shams underwent surgery without clamping or injection. Kidney function was assessed by plasma creatinine; injury by aspartate aminotransferase, heart-type-fatty-acid-binding-protein, and histopathology. Renal gene expression of tumor necrosis factor α, interleukin (IL)-6, heat shock protein 70, Chemokine (C-X-C motif) ligand 2, cyclo-oxygenase 2, endothelin-1, IL-10, heme oxygenase 1, B-cell lymphoma 2, and bcl-2-like protein 4 were determined by quantitative reverse transcriptase polymerase chain reaction. RESULTS None of the 3 h and 24 h end points were different between Control and AAT. In Sham, survival was 100% (6/6), 33% in Control (2/6), and 83% (5/6) in AAT (overall log-rank 0.03). At 7 d, plasma creatinine was lower with higher glomerular filtration rate in surviving AAT treated animals compared to Control (P < 0.001, P 0.03, respectively). These also had lower tumor necrosis factor α and IL-6 gene expression (P 0.001, P < 0.001, respectively). CONCLUSIONS These data suggest that a single intravenous dose of AAT immediately before ischemia might affect proinflammatory gene expression, glomerular filtration rate and animal survival at 1 wk after reperfusion despite an absence of improvement in early renal function and injury. These findings deserve further investigating in sufficiently powered studies including both sexes.
Collapse
Affiliation(s)
- Ina Jochmans
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| | - Evelyne Lerut
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Diethard Monbaliu
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Janciauskiene S, Lechowicz U, Pelc M, Olejnicka B, Chorostowska-Wynimko J. Diagnostic and therapeutic value of human serpin family proteins. Biomed Pharmacother 2024; 175:116618. [PMID: 38678961 DOI: 10.1016/j.biopha.2024.116618] [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: 01/11/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
SERPIN (serine proteinase inhibitors) is an acronym for the superfamily of structurally similar proteins found in animals, plants, bacteria, viruses, and archaea. Over 1500 SERPINs are known in nature, while only 37 SERPINs are found in humans, which participate in inflammation, coagulation, angiogenesis, cell viability, and other pathophysiological processes. Both qualitative or quantitative deficiencies or overexpression and/or abnormal accumulation of SERPIN can lead to diseases commonly referred to as "serpinopathies". Hence, strategies involving SERPIN supplementation, elimination, or correction are utilized and/or under consideration. In this review, we discuss relationships between certain SERPINs and diseases as well as putative strategies for the clinical explorations of SERPINs.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany; Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Magdalena Pelc
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Beata Olejnicka
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland.
| |
Collapse
|
8
|
Lotke R, Petersen M, Sauter D. Restriction of Viral Glycoprotein Maturation by Cellular Protease Inhibitors. Viruses 2024; 16:332. [PMID: 38543698 PMCID: PMC10975521 DOI: 10.3390/v16030332] [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: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 05/23/2024] Open
Abstract
The human genome is estimated to encode more than 500 proteases performing a wide range of important physiological functions. They digest proteins in our food, determine the activity of hormones, induce cell death and regulate blood clotting, for example. During viral infection, however, some proteases can switch sides and activate viral glycoproteins, allowing the entry of virions into new target cells and the spread of infection. To reduce unwanted effects, multiple protease inhibitors regulate the proteolytic processing of self and non-self proteins. This review summarizes our current knowledge of endogenous protease inhibitors, which are known to limit viral replication by interfering with the proteolytic activation of viral glycoproteins. We describe the underlying molecular mechanisms and highlight the diverse strategies by which protease inhibitors reduce virion infectivity. We also provide examples of how viruses evade the restriction imposed by protease inhibitors. Finally, we briefly outline how cellular protease inhibitors can be modified and exploited for therapeutic purposes. In summary, this review aims to summarize our current understanding of cellular protease inhibitors as components of our immune response to a variety of viral pathogens.
Collapse
Affiliation(s)
| | | | - Daniel Sauter
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
9
|
Abdel-Karim TR, Hodges JS, Herold KC, Pruett TL, Ramanathan KV, Hering BJ, Dunn TB, Kirchner VA, Beilman GJ, Bellin MD. Peri-Transplant Inflammation and Long-Term Diabetes Outcomes Were Not Impacted by Either Etanercept or Alpha-1-Antitrypsin Treatment in Islet Autotransplant Recipients. Transpl Int 2024; 37:12320. [PMID: 38357216 PMCID: PMC10864605 DOI: 10.3389/ti.2024.12320] [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: 10/27/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024]
Abstract
The instant blood-mediated inflammatory response (IBMIR) causes islet loss and compromises diabetes outcomes after total pancreatectomy with islet autotransplant (TPIAT). We previously reported a possible benefit of etanercept in maintaining insulin secretion 3 months post-TPIAT. Here, we report 2-year diabetes outcomes and peri-operative inflammatory profiles from a randomized trial of etanercept and alpha-1 antitrypsin (A1AT) in TPIAT. We randomized 43 TPIAT recipients to A1AT (90 mg/kg IV x6 doses, n = 13), etanercept (50 mg then 25 mg SQ x 5 doses, n = 14), or standard care (n = 16). Inflammatory cytokines, serum A1AT and unmethylated insulin DNA were drawn multiple times in the perioperative period. Islet function was assessed 2 years after TPIAT with mixed meal tolerance test, intravenous glucose tolerance test and glucose-potentiated arginine induced insulin secretion. Cytokines, especially IL-6, IL-8, IL-10, and MCP-1, were elevated during and after TPIAT. However, only TNFα differed significantly between groups, with highest levels in the etanercept group (p = 0.027). A1AT increased after IAT in all groups (p < 0.001), suggesting endogenous upregulation. Unmethylated insulin DNA ratios (a marker of islet loss) and 2 years islet function testing were similar in the three groups. To conclude, we found no sustained benefit from administering etanercept or A1AT in the perioperative period.
Collapse
Affiliation(s)
| | - James S. Hodges
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Kevan C. Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, United States
| | - Timothy L. Pruett
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | | | - Bernhard J. Hering
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Ty B. Dunn
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Varvara A. Kirchner
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
- Department of Surgery, Stanford University, Palo Alto, CA, United States
| | - Gregory J. Beilman
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Melena D. Bellin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
10
|
Brami I, Zuckerman T, Ram R, Avni B, Peretz G, Ostrovsky D, Lior Y, Faour C, McElvaney O, McElvaney NG, Lewis EC. Altered Serum Alpha1-Antitrypsin Protease Inhibition before and after Clinical Hematopoietic Stem Cell Transplantation: Association with Risk for Non-Relapse Mortality. Int J Mol Sci 2023; 25:422. [PMID: 38203593 PMCID: PMC10779144 DOI: 10.3390/ijms25010422] [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: 11/21/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
α1-Antitrypsin (AAT), an acute-phase reactant not unsimilar to C-reactive protein (CRP), is a serine protease inhibitor that harbors tissue-protective and immunomodulatory attributes. Its concentrations appropriately increase during conditions of extensive tissue injury, and it induces immune tolerance, in part, by inhibiting the enzymatic activity of the inflammatory serine protease, proteinase 3 (PR3). Typically administered to patients with genetic AAT deficiency, AAT treatment was recently shown to improve outcomes in patients with steroid-refractory graft-versus-host disease (GVHD). GVHD represents a grave outcome of allogeneic hematopoietic stem cell transplantation (HSCT), a potentially curative intervention for hematological diseases. The procedure requires radio/chemotherapy conditioning of the prospective marrow recipient, a cytotoxic process that causes vast tissue injury and, in some formats, interferes with liver production of AAT. To date, changes in the functional profile of AAT during allogeneic HSCT, and during the cytotoxic intervention that precedes HSCT, are unknown. The present study followed 53 patients scheduled for allogeneic HSCT (trial registration NCT03188601). Serum samples were tested before and after HSCT for AAT and CRP levels and for intrinsic anti-proteolytic activity. The ex vivo response to clinical-grade AAT was tested on circulating patient leukocytes and on a human epithelial cell line treated with patient sera in a gap closure assay. According to the ex vivo experiments, circulating leukocytes responded to AAT with a favorable immune-regulated profile, and epithelial gap closure was enhanced by AAT in sera from GVHD-free patients but not in sera from patients who developed GVHD. According to serum collected prior to HSCT, non-relapse mortality was reliably predicted by combining three components: AAT and CRP levels and serum anti-proteolytic activity. Taken together, HSCT outcomes are significantly affected by the anti-proteolytic function of circulating AAT, supporting early AAT augmentation therapy for allogeneic HSCT patients.
Collapse
Affiliation(s)
- Ido Brami
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Be’er-Sheva 8410501, Israel;
| | - Tsila Zuckerman
- Hematology Department and Bone Marrow Transplantation Unit, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Ron Ram
- Bone Marrow Transplantation Unit, The Division of Hematology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel;
| | - Batia Avni
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah-Hebrew University Medical Center, Ein Kerem, Jerusalem 9112001, Israel;
| | - Galit Peretz
- Department of Hematology, Soroka University Medical Center, Be’er-Sheva 8410101, Israel;
| | - Daniel Ostrovsky
- Clinical Research Center, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be’er-Sheva 8410101, Israel;
| | - Yotam Lior
- Division of Anesthesiology, Pain and Intensive Care, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel;
| | - Caroline Faour
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israeli Institute of Technology, Haifa 3109601, Israel;
| | - Oisin McElvaney
- The Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, D02 YN77 Dublin, Ireland; (O.M.); (N.G.M.)
| | - Noel G. McElvaney
- The Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, D02 YN77 Dublin, Ireland; (O.M.); (N.G.M.)
| | - Eli C. Lewis
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Be’er-Sheva 8410501, Israel;
| |
Collapse
|
11
|
He G, Yu W, Li H, Liu J, Tu Y, Kong D, Long Z, Liu R, Peng J, Wang Z, Liu P, Hai C, Yan W, Li W. Alpha-1 antitrypsin protects against phosgene-induced acute lung injury by activating the ID1-dependent anti-inflammatory response. Eur J Pharmacol 2023; 957:176017. [PMID: 37673367 DOI: 10.1016/j.ejphar.2023.176017] [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: 05/25/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023]
Abstract
Phosgene is widely used as an industrial chemical, and phosgene inhalation causes acute lung injury (ALI), which may further progress into pulmonary edema. Currently, an antidote for phosgene poisoning is not known. Alpha-1 antitrypsin (α1-AT) is a protease inhibitor used to treat patients with emphysema who are deficient in α1-AT. Recent studies have revealed that α1-AT has both anti-inflammatory and anti-SARS-CoV-2 effects. Herein, we aimed to investigate the role of α1-AT in phosgene-induced ALI. We observed a time-dependent increase in α1-AT expression and secretion in the lungs of rats exposed to phosgene. Notably, α1-AT was derived from neutrophils but not from macrophages or alveolar type II cells. Moreover, α1-AT knockdown aggravated phosgene- and lipopolysaccharide (LPS)-induced inflammation and cell death in human bronchial epithelial cells (BEAS-2B). Conversely, α1-AT administration suppressed the inflammatory response and prevented death in LPS- and phosgene-exposed BEAS-2B cells. Furthermore, α1-AT treatment increased the inhibitor of DNA binding 1 (ID1) gene expression, which suppressed NF-κB pathway activation, reduced inflammation, and inhibited cell death. These data demonstrate that neutrophil-derived α1-AT acts as a self-protective mechanism, which protects against phosgene-induced ALI by activating the ID1-dependent anti-inflammatory response. This study may provide novel strategies for the treatment of patients with phosgene-induced ALI.
Collapse
Affiliation(s)
- Gaihua He
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Weihua Yu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Hongwei Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiangzheng Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Yongmei Tu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Deqin Kong
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Zi Long
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Peng
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhao Wang
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Penghui Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Chunxu Hai
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an, 710032, China.
| | - Wenli Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
12
|
Bai X, Schountz T, Buckle AM, Talbert JL, Sandhaus RA, Chan ED. Alpha-1-antitrypsin antagonizes COVID-19: a review of the epidemiology, molecular mechanisms, and clinical evidence. Biochem Soc Trans 2023; 51:1361-1375. [PMID: 37294003 PMCID: PMC10317171 DOI: 10.1042/bst20230078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023]
Abstract
Alpha-1-antitrypsin (AAT), a serine protease inhibitor (serpin), is increasingly recognized to inhibit SARS-CoV-2 infection and counter many of the pathogenic mechanisms of COVID-19. Herein, we reviewed the epidemiologic evidence, the molecular mechanisms, and the clinical evidence that support this paradigm. As background to our discussion, we first examined the basic mechanism of SARS-CoV-2 infection and contend that despite the availability of vaccines and anti-viral agents, COVID-19 remains problematic due to viral evolution. We next underscored that measures to prevent severe COVID-19 currently exists but teeters on a balance and that current treatment for severe COVID-19 remains grossly suboptimal. We then reviewed the epidemiologic and clinical evidence that AAT deficiency increases risk of COVID-19 infection and of more severe disease, and the experimental evidence that AAT inhibits cell surface transmembrane protease 2 (TMPRSS2) - a host serine protease required for SARS-CoV-2 entry into cells - and that this inhibition may be augmented by heparin. We also elaborated on the panoply of other activities of AAT (and heparin) that could mitigate severity of COVID-19. Finally, we evaluated the available clinical evidence for AAT treatment of COVID-19.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, U.S.A
- Department of Academic Affairs, National Jewish Health, Denver, CO, U.S.A
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, U.S.A
| | - Tony Schountz
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, U.S.A
| | - Ashley M. Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- PTNG Bio, Melbourne, Australia
| | - Janet L. Talbert
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, U.S.A
| | | | - Edward D. Chan
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, U.S.A
- Department of Academic Affairs, National Jewish Health, Denver, CO, U.S.A
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, U.S.A
| |
Collapse
|
13
|
Jager S, Cramer DAT, Heck AJR. Normal Alpha-1-Antitrypsin Variants Display in Serum Allele-Specific Protein Levels. J Proteome Res 2023; 22:1331-1338. [PMID: 36946534 PMCID: PMC10088046 DOI: 10.1021/acs.jproteome.2c00833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Indexed: 03/23/2023]
Abstract
Alpha-1-antitrypsin (A1AT or SERPINA1) has been proposed as a putative biomarker distinguishing healthy from diseased donors throughout several proteomics studies. However, the SERPINA1 gene displays high variability of frequent occurring genotypes among the general population. These different genotypes may affect A1AT expression and serum protein concentrations, and this is often not known, ignored, and/or not reported in serum proteomics studies. Here, we address allele-specific protein serum levels of A1AT in donors carrying the normal M variants of A1AT by measuring the proteoform profiles of purified A1AT from 81 serum samples, originating from 52 donors. When focusing on heterozygous donors, our data clearly reveal a statistically relevant difference in allele-specific protein serum levels of A1AT. In donors with genotype PI*M1VM1A, the experimentally observed ratio was approximately 1:1 (M1V/M1A, 1.00:0.96 ± 0.07, n = 17). For individuals with genotype PI*M1VM2, this ratio was 1:1.28 (M1V/M2, 1.00:1.31, ±0.19, n = 7). For genotypes PI*M1VM3 and PI*M1AM3, a significant higher amount of M3 was observed compared to the M1-subtypes (M1V/M3, 1.00:1.84 ± 0.35, n = 8; M1A/M3, 1.00:1.61 ± 0.33, n = 5). We argue that these observations are important and should be considered when analyzing serum A1AT levels before proposing A1AT as a putative serum biomarker.
Collapse
Affiliation(s)
- Shelley Jager
- Biomolecular Mass Spectrometry
and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| | - Dario A. T. Cramer
- Biomolecular Mass Spectrometry
and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry
and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| |
Collapse
|
14
|
Zozulya SA, Sizov SV, Oleichik IV, Klyushnik TP. Clinical-Immunological Correlates in Post-COVID-19 Endogenous Psychoses. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2023; 53:174-179. [PMID: 37020644 PMCID: PMC10063428 DOI: 10.1007/s11055-023-01405-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 04/03/2023]
Abstract
Objectives. To carry out a clinical and immunological study of the potential impact of coronavirus infection on the course of endogenous psychoses. Materials and methods. A total of 33 female patients aged 16-48 years with depressive-delusional states (F20.01, F21, F31) developing after coronavirus infections took part; group 1 consisted of 15 people who developed depressive-delusional states 1-2 months after COVID-19; group 2 consisted of 18 people with similar psychoses developing at later time points (2-6 months). The severity of psychopathological symptoms was assessed using the PANSS and HDRS-21 scales. The activity of inflammatory markers leukocyte elastase (LE) and α1-proteinase inhibitor (α1-PI) was determined in patients' blood. Absolute neutrophil and lymphocyte contents and their ratio (the neutrophil:lymphocyte index) were also evaluated. Standard values for indicators from healthy donors corresponding to patients in terms of age and sex were used as control values. Results. Endogenous psychosis developing at longer intervals after coronavirus infection (group 2) was found to be associated with "typical" inflammatory reactions, with increases in the activity of acute-phase proteins (α1-PI: 43.0 (35.6-49.7) IU/ml, p = 0.001) and neutrophil degranulation activity (LE - 254.8 (238.0-271.0) nmol/min·ml, p < 0.001), which was associated with the development of depressive-delusional states with dominance of manifestations of positive affectivity (anxiety, melancholy) and the extended nature of delusional disorders, which were mostly incongruent to affect. Conversely, development of endogenous psychosis during the first two months after COVID-19 (group 1) was characterized by a spectrum of inflammatory biomarkers with a decrease in neutrophil count ((2.6 ± 0.9)·109/liter, p < 0.05) and low LE activity (196 (172-209.4) nmol/min·ml, p < 0.001). This immunological profile was associated with predominance of manifestations of negative affectivity (apathy, asthenia, adynamia) in the structure of depressive-delusional states and the relatively undeveloped nature of delusional disorders, which were predominantly congruent to affect. Conclusions. The clinical and biological correlates found here presumptively indicate that experience of COVID-19 infection has a modulatory effect on neuroinflammation and the structure of endogenous psychosis.
Collapse
Affiliation(s)
| | - S. V. Sizov
- Mental Health Research Center, Moscow, Russia
| | | | | |
Collapse
|
15
|
Park DJ, Duggan E, Ho K, Dorschner RA, Dobke M, Nolan JP, Eliceiri BP. Serpin-loaded extracellular vesicles promote tissue repair in a mouse model of impaired wound healing. J Nanobiotechnology 2022; 20:474. [PMID: 36335351 PMCID: PMC9636779 DOI: 10.1186/s12951-022-01656-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/29/2022] [Indexed: 11/08/2022] Open
Abstract
Chronic metabolic diseases such as diabetes are characterized by delayed wound healing and a dysregulation of the inflammatory phase of wound repair. Our study focuses on changes in the payload of extracellular vesicles (EVs) communicating between immune cells and stromal cells in the wound bed, which regulate the rate of wound closure. Adoptive transfer of EVs from genetically defined mouse models are used here to demonstrate a functional and molecular basis for differences in the pro-reparative biological activity of diabetic (db/db) vs. wildtype EVs in wound healing. We identify several members of the Serpin family of serine protease inhibitors that are absent in db/db EVs, then we overexpress Serpin A1, F2 and G1 in EVs to evaluate their effect on wound healing in db/db mice. Serpins have an important role in regulating levels of elastase, plasmin and complement factors that coordinate immune cell signaling in full thickness wounds in a diabetic model. Here, we establish a novel therapeutic approach by engineering the payload of EVs based on proteomic analysis. Serpin-loaded EVs were used to rescue the Serpin deficiency identified by proteomics and promote wound healing in db/db mice, as well as evaluated how EVs affected extracellular matrix remodeling and the resolution of tissue injury. Therefore, we propose that the identification of EV payloads that are downregulated in diabetic wounds can be systematically analyzed for their functional activity and potential as a therapeutic, based on whether their re-expression in engineered EVs restores normal kinetics of tissue repair in chronic wounds.
Collapse
Affiliation(s)
- Dong Jun Park
- Departments of Surgery, University of California San Diego, 9500 Gilman Drive, MC 8236, La Jolla, CA, 92093-8236, USA
| | - Erika Duggan
- Scintillon Institute, 6868 Nancy Ridge, San Diego, CA, 92121, USA
| | - Kayla Ho
- Departments of Surgery, University of California San Diego, 9500 Gilman Drive, MC 8236, La Jolla, CA, 92093-8236, USA
| | - Robert A Dorschner
- Dermatology, University of California San Diego, 9500 Gilman Drive, MC 8236, La Jolla, CA, 92093-8236, USA
| | - Marek Dobke
- Departments of Surgery, University of California San Diego, 9500 Gilman Drive, MC 8236, La Jolla, CA, 92093-8236, USA
| | - John P Nolan
- Scintillon Institute, 6868 Nancy Ridge, San Diego, CA, 92121, USA
| | - Brian P Eliceiri
- Departments of Surgery, University of California San Diego, 9500 Gilman Drive, MC 8236, La Jolla, CA, 92093-8236, USA.
| |
Collapse
|
16
|
Kunder M, Lakshmaiah V, Kutty AM. Selective decrease in alpha1-antitrypsin levels in diabetic retinopathy: Could the levels of it be playing a role in the pathophysiology of diabetic retinopathy? Indian J Med Res 2022; 156:104-110. [PMID: 36124492 PMCID: PMC9903389 DOI: 10.4103/ijmr.ijmr_1293_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background & objectives Type 2 diabetes mellitus (T2DM) is known to induce inflammation and activation of neutrophils causing the release of neutrophil elastase (NE), a pro-inflammatory proteinase. The activity of NE is regulated by endogenous inhibitors alpha1-antitrypsin (α1-AT) and alpha2-macroglobulin (α2-MG). Disrupted proteolytic homeostasis in T2DM patients is one of the causes for vascular complications. This study was carried out for evaluating the levels of plasma NE, α1-AT, α2-MG and NE-α1-AT complex to understand their roles in the pathophysiology of diabetic nephropathy (DN) and diabetic retinopathy (DR). Methods A total of 240 participants (Control, n=60; T2DM, n=60; DN, n=60; and DR, n=60) were recruited after recording history, clinical examination and laboratory investigations. Retinopathy was confirmed by fundoscopy and nephropathy by urinary albumin excretion and serum creatinine levels. NE was measured using STANA. α1-AT, α2-MG and NE-α1-AT complex were estimated by ELISA. Results Baseline clinical and laboratory findings were confirmatory to the study groups. The mean elastase activity was higher (P<0.0005) in diabetes groups (T2DM=0.73±0.31, DN=0.87±0.35, DR=0.76±0.41) than controls (0.35±0.20). The levels of α1-AT were lower in DR (8.77±2.85) than DN (26.26±6.16) and T2DM (41.13±14.06) when juxtaposed with controls (122.95±25.71). The approximate fold decrease of α1-AT levels was 15 for DR and four for DN compared to controls. The levels of α2-MG were lowered in T2DM (167.29±30.45), DN (144.66±13.72), and DR (104.67±11.47) than controls (208.87±31.16). The NE-α1-AT complex levels were: controls (215.83±13.61), T2DM (98.85±23.85), DN (129.26±20.40) and DR (153.25±17.11). Interpretation & conclusions Homeostasis of NE, α1-AT and α2-MG is disrupted in T2DM, DN and DR. Strikingly reduced levels of α1-AT observed in DR are indicative of its possible role in the pathophysiology of retinopathy and emphasizes α1-AT as a plausible therapeutic target.
Collapse
Affiliation(s)
- Mamatha Kunder
- Department of Biochemistry, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education & Research, Kolar, Karnataka, India
| | - V. Lakshmaiah
- Department of Medicine, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education & Research, Kolar, Karnataka, India
| | - A.V. Moideen Kutty
- Department of Biochemistry, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education & Research, Kolar, Karnataka, India,For correspondence: Dr A.V. Moideen Kutty, Department of Biochemistry, Sri Devaraj Urs Medical College, Tamaka, Kolar 563 103, Karnataka, India e-mail:
| |
Collapse
|
17
|
10,12-conjugated linoleic acid supplementation improves HDL composition and function in mice. J Lipid Res 2022; 63:100241. [PMID: 35714730 PMCID: PMC9283942 DOI: 10.1016/j.jlr.2022.100241] [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: 02/18/2022] [Revised: 05/12/2022] [Accepted: 06/06/2022] [Indexed: 12/31/2022] Open
Abstract
Obesity is associated with inflammation, insulin resistance, and type 2 diabetes, which are major risk factors for CVD. One dietary component of ruminant animal foods, 10,12-conjugated linoleic acid (10,12 CLA), has been shown to promote weight loss in humans. Previous work has shown that 10,12 CLA is atheroprotective in mice by a mechanism that may be distinct from its weight loss effects, but this exact mechanism is unclear. To investigate this, we evaluated HDL composition and function in obese LDL receptor (Ldlr−/−) mice that were losing weight because of 10,12 CLA supplementation or caloric restriction (CR; weight-matched control group) and in an obese control group consuming a high-fat high-sucrose diet. We show that 10,12 CLA-HDL exerted a stronger anti-inflammatory effect than CR- or high-fat high-sucrose-HDL in cultured adipocytes. Furthermore, the 10,12 CLA-HDL particle (HDL-P) concentration was higher, attributed to more medium- and large-sized HDL-Ps. Passive cholesterol efflux capacity of 10,12 CLA-HDL was elevated, as was expression of HDL receptor scavenger receptor class B type 1 in the aortic arch. Murine macrophages treated with 10,12 CLA in vitro exhibited increased expression of cholesterol transporters Abca1 and Abcg1, suggesting increased cholesterol efflux potential of these cells. Finally, proteomics analysis revealed elevated Apoa1 content in 10,12 CLA-HDL-Ps, consistent with a higher particle concentration, and particles were also enriched with alpha-1-antitrypsin, an emerging anti-inflammatory and antiatherosclerotic HDL-associated protein. We conclude that 10,12 CLA may therefore exert its atheroprotective effects by increasing HDL-P concentration, HDL anti-inflammatory potential, and promoting beneficial effects on cholesterol efflux.
Collapse
|
18
|
Ochoa A, Hassinger ATB, Holding ML, Gibbs HL. Genetic characterization of potential venom resistance proteins in California ground squirrels (
Otospermophilus beecheyi
) using transcriptome analyses. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B: MOLECULAR AND DEVELOPMENTAL EVOLUTION 2022; 340:259-269. [PMID: 35611404 DOI: 10.1002/jez.b.23145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/16/2022] [Accepted: 05/09/2022] [Indexed: 11/11/2022]
Abstract
Understanding the molecular basis of adaptations in coevolving species requires identifying the genes that underlie reciprocally selected phenotypes, such as those involved in venom in snakes and resistance to the venom in their prey. In this regard, California ground squirrels (CGS; Otospermophilus beecheyi) are eaten by northern Pacific rattlesnakes (Crotalus oreganus oreganus), but individual squirrels may still show substantial resistance to venom and survive bites. A recent study using proteomics identified venom interactive proteins (VIPs) in the blood serum of CGS. These VIPs represent possible resistance proteins, but the sequences of genes encoding them are unknown despite the value of such data to molecular studies of coevolution. To address this issue, we analyzed a de novo assembled transcriptome from CGS liver tissue-where many plasma proteins are synthesized-and other tissues from this species. We then examined VIP sequences in terms of three characteristics that identify them as possible resistance proteins: evidence for positive selection, high liver expression, and nonsynonymous variation across CGS populations. Based on these characteristics, we identified five VIPs (i.e., α-2-macroglobulin, α-1-antitrypsin-like protein GS55-LT, apolipoprotein A-II, hibernation-associated plasma protein HP-20, and hibernation-associated plasma protein HP-27) as the most likely candidates for resistance proteins among VIPs identified to date. Four of these proteins have been previously implicated in conferring resistance to the venom in mammals, validating our approach. When combined with the detailed information available for rattlesnake venom proteins, these results set the stage for future work focused on understanding coevolutionary interactions at the molecular level between these species.
Collapse
Affiliation(s)
- Alexander Ochoa
- Department of Evolution, Ecology, and Organismal Biology and Ohio Biodiversity Conservation Partnership Ohio State University Columbus Ohio USA
- Department of Biology University of Central Florida Orlando Florida USA
| | - Alyssa T. B. Hassinger
- Department of Evolution, Ecology, and Organismal Biology and Ohio Biodiversity Conservation Partnership Ohio State University Columbus Ohio USA
| | | | - H. Lisle Gibbs
- Department of Evolution, Ecology, and Organismal Biology and Ohio Biodiversity Conservation Partnership Ohio State University Columbus Ohio USA
| |
Collapse
|
19
|
LMAN1-MCFD2 complex is a cargo receptor for the ER-Golgi transport of α1-antitrypsin. Biochem J 2022; 479:839-855. [PMID: 35322856 PMCID: PMC9022998 DOI: 10.1042/bcj20220055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022]
Abstract
α1-antitrypsin (AAT) is a serine protease inhibitor synthesized in hepatocytes and protects the lung from damage by neutrophil elastase. AAT gene mutations result in AAT deficiency (AATD), which leads to lung and liver diseases. The AAT Z variant forms polymer within the endoplasmic reticulum (ER) of hepatocytes and results in reduction of AAT secretion and severe disease. Previous studies demonstrated a secretion defect of AAT in LMAN1 deficient cells, and mild decreases in AAT levels in male LMAN1 and MCFD2 deficient mice. LMAN1 is a transmembrane lectin that forms a complex with a small soluble protein MCFD2. The LMAN1-MCFD2 protein complex cycles between the ER and the Golgi. Here we report that LMAN1 and MCFD2 knockout (KO) HepG2 and HEK293T cells display reduced AAT secretion and elevated intracellular AAT levels due to a delayed ER-to-Golgi transport of AAT. Secretion defects in KO cells were rescued by wild-type LMAN1 or MCFD2, but not by mutant proteins. Elimination of the second glycosylation site of AAT abolished LMAN1 dependent secretion. Co-immunoprecipitation experiment in MCFD2 KO cells suggested that AAT interaction with LMAN1 is independent of MCFD2. Furthermore, our results suggest that secretion of the Z variant, both monomers and polymers, is also LMAN1-dependent. Results provide direct evidence supporting that the LMAN1-MCFD2 complex is a cargo receptor for the ER-to-Golgi transport of AAT and that interactions of LMAN1 with an N-glycan of AAT is critical for this process. These results have implications in production of recombinant AAT and in developing treatments for AATD patients.
Collapse
|
20
|
Jager S, Cramer DAT, Hoek M, Mokiem NJ, van Keulen BJ, van Goudoever JB, Dingess KA, Heck AJR. Proteoform Profiles Reveal That Alpha-1-Antitrypsin in Human Serum and Milk Is Derived From a Common Source. Front Mol Biosci 2022; 9:858856. [PMID: 35274008 PMCID: PMC8902301 DOI: 10.3389/fmolb.2022.858856] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The Alpha-1-Antitrypsin (A1AT) protein is an important protease inhibitor highly abundant in human serum and other body fluids. Additional to functioning as a protease inhibitor, A1AT is an important acute phase protein. Here, we set out to compare the proteoform profiles of A1AT purified from the human serum and milk of eight healthy donors to determine the origin of human milk A1AT. Following affinity purification, size-exclusion chromatography coupled to native mass spectrometry was used to monitor individual proteoform profiles comparing inter- and intra-donor profiles. The A1AT intra-donor proteoform profiles were found to be highly identical between serum and milk, while they were highly distinct between donors, even when comparing only serum or milk samples. The observed inter-donor proteoform variability was due to differences in the abundances of different N-glycoforms, mainly due to branching, fucosylation, and the relative abundance of N-terminally processed A1AT fragments. From our data we conclude that nearly all A1AT in serum and milk is synthesized by a common source, i.e. the liver, and then secreted into the circulation and enters the mammary gland via diffusion or transport. Thereby, proteoform profile changes, as seen upon infection and/or inflammation in the blood will be reflected in the milk, which may then be transferred to the breastfed infant.
Collapse
Affiliation(s)
- Shelley Jager
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Dario A. T. Cramer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Nadia J. Mokiem
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Britt J. van Keulen
- Department of Pediatrics, Vrije Universiteit, University of Amsterdam Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Johannes B. van Goudoever
- Department of Pediatrics, Vrije Universiteit, University of Amsterdam Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Kelly A. Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- Department of Pediatrics, Vrije Universiteit, University of Amsterdam Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- *Correspondence: Albert J. R. Heck,
| |
Collapse
|
21
|
Zozulya S, Sizov S, Oleichik I, Klyushnik T. Clinical and immunological correlates in endogenous psychoses developed after COVID-19. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:71-77. [DOI: 10.17116/jnevro202212206271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
22
|
Fazal Y, Zohaib M, Syed B, Ansari SH, Hashim Z, Ahmed A, Zarina S. Prenatal diagnosis of maternal serum from mothers carrying β-thalassemic fetus. Pediatr Int 2022; 64:e14999. [PMID: 34559910 DOI: 10.1111/ped.14999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND This study focuses on the discovery of protein biomarkers from the maternal serum of β-thalassemic trait mothers carrying the normal fetus and β-thalassemic major fetus. METHODS Serum samples from β-thalassemic trait mothers carrying major (N = 5) and normal fetuses (N = 5) were studied. The IVS1-5 thalassemia mutation was common among β-thalassemic trait mothers who were carrying a homozygous β-thalassemic fetus (IVS1-5/ IVS1-5 mutation) or a normal fetus (no mutation). We employed two-dimensional gel electrophoresis and mass spectrometry analysis to explore differentially expressed maternal serum proteins from thalassemia carrier couples with the same β-thalassemia mutation. Western blotting was performed for one of the identified proteins to validate our data. RESULTS Ten proteins were identified in the maternal serum of β-thalassemic trait mothers carrying the β-thalassemic major fetus and normal fetus. Among these, serotransferrin, haptoglobin, α-1 anti-trypsin, apo-lipoprotein A1, and the fibrinogen-β chain were found to be upregulated in mothers carrying major fetuses and are known to be associated with pregnancy-related disorders. The expression of α-1 anti-trypsin was validated through western blotting. CONCLUSIONS Proteins identified in the current study from maternal serum are reported to contribute to hereditary disorders. We suggest that these can serve as putative screening markers for non-invasive prenatal diagnosis in β-thalassemic pregnancies.
Collapse
Affiliation(s)
- Yumna Fazal
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, Pakistan
| | | | - Basir Syed
- Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Saqib H Ansari
- Omair Sana Foundation, Gulshan-e-Iqbal, Karachi, Pakistan
| | - Zehra Hashim
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, Pakistan
| | - Aftab Ahmed
- Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Shamshad Zarina
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, Pakistan
| |
Collapse
|
23
|
Lior Y, Shtriker E, Kahremany S, Lewis EC, Gruzman A. Development of anti-inflammatory peptidomimetics based on the structure of human alpha1-antitrypsin. Eur J Med Chem 2021; 228:113969. [PMID: 34763945 DOI: 10.1016/j.ejmech.2021.113969] [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] [Received: 08/04/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/10/2023]
Abstract
Human α1-antitrypsin (hAAT) has two distinguishing functions: anti-protease activity and regulation of the immune system. In the present study we hypothesized that those two protein functions are mediated by different structural domains on the hAAT surface. Indeed, such biologically active immunoregulatory sites (not associated with canonical anti-protease activity) on the surface of hAAT were identified by in silico methods. Several peptides were derived from those immunoregulatory sites. Four peptides exhibited impressive biological effects in pharmacological concentration ranges. Peptidomimetic (14) was developed, based on the structure of the most druggable and active peptide. The compound exhibited a potent anti-inflammatory activity in vitro and in vivo. Such a compound could be used as a basis for developing novel anti-inflammatory drug candidates and as a research tool for better understanding hAAT functions.
Collapse
Affiliation(s)
- Yotam Lior
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Efrat Shtriker
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Shirin Kahremany
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Skin Research Institute, The Dead Sea and Arava Science Center, 86910, Masada, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Arie Gruzman
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| |
Collapse
|
24
|
Alpha-1 Antitrypsin and Hepatocellular Carcinoma in Liver Cirrhosis: SERPINA1 MZ or MS Genotype Carriage Decreases the Risk. Int J Mol Sci 2021; 22:ijms221910560. [PMID: 34638908 PMCID: PMC8509047 DOI: 10.3390/ijms221910560] [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: 07/31/2021] [Revised: 09/12/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Heterozygotes for Z or S alleles of alpha-1-antrypsin (AAT) have low serum AAT levels. Our aim was to compare the risk of hepatocellular carcinoma (HCC) in patients with liver cirrhosis carrying the SERPINA1 MM, MZ and MS genotypes. The study groups consisted of 1119 patients with liver cirrhosis of various aetiologies, and 3240 healthy individuals served as population controls. The MZ genotype was significantly more frequent in the study group (55/1119 vs. 87/3240, p < 0.0001). The MS genotype frequency was comparable in controls (32/119 vs. 101/3240, p = 0.84). MZ and MS heterozygotes had lower serum AAT level than MM homozygotes (medians: 0.90 g/L; 1.40 g/L and 1.67 g/L; p < 0.001 for both). There were significantly fewer patients with HCC in the cirrhosis group among MZ and MS heterozygotes than in MM homozygotes (5/55 and 1/32 respectively, vs. 243/1022, p < 0.01 for both). The risk of HCC was lower in MZ and MS heterozygotes than in MM homozygotes (OR 0.3202; 95% CI 0.1361–0.7719 and OR 0.1522; 95% CI 0.02941–0.7882, respectively). Multivariate analysis of HCC risk factors identified MZ or MS genotype carriage as a protective factor, whereas age, male sex, BMI and viral aetiology of cirrhosis increased HCC risk.
Collapse
|
25
|
Ehlting C, Wolf SD, Bode JG. Acute-phase protein synthesis: a key feature of innate immune functions of the liver. Biol Chem 2021; 402:1129-1145. [PMID: 34323429 DOI: 10.1515/hsz-2021-0209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023]
Abstract
The expression of acute-phase proteins (APP's) maintains homeostasis and tissue repair, but also represents a central component of the organism's defense strategy, especially in the context of innate immunity. Accordingly, an inflammatory response is accompanied by significant changes in the serum protein composition, an aspect that is also used diagnostically. As the main site of APP synthesis the liver is constantly exposed to antigens or pathogens via blood flow, but also to systemic inflammatory signals originating either from the splanchnic area or from the circulation. Under both homeostatic and acute-phase response (APR) conditions the composition of APP's is determined by the pattern of regulatory mediators derived from the systemic circulation or from local cell populations, especially liver macrophages. The key regulators mentioned here most frequently are IL-1β, IL-6 and TNF-α. In addition to a variety of molecular mediators described mainly on the basis of in vitro studies, recent data emphasize the in vivo relevance of cellular key effectors as well as molecular key mediators and protein modifications for the regulation and function of APP's. These are aspects, on which the present review is primarily focused.
Collapse
Affiliation(s)
- Christian Ehlting
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Hospital of the Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Stephanie D Wolf
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Hospital of the Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Johannes G Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Hospital of the Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
26
|
Edinger F, Schmitt C, Koch C, McIntosh JM, Janciauskiene S, Markmann M, Sander M, Padberg W, Grau V. Application of alpha1-antitrypsin in a rat model of veno-arterial extracorporeal membrane oxygenation. Sci Rep 2021; 11:15849. [PMID: 34349162 PMCID: PMC8339069 DOI: 10.1038/s41598-021-95119-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 07/19/2021] [Indexed: 11/08/2022] Open
Abstract
Extracorporeal membrane oxygenation (ECMO) is a life-saving intervention for patients suffering from respiratory or cardiac failure. The ECMO-associated morbidity and mortality depends to a large extent on the underlying disease and is often related to systemic inflammation, consecutive immune paralysis and sepsis. Here we tested the hypothesis that human α1-antitrypsin (SERPINA1) due to its anti-protease and anti-inflammatory functions may attenuate ECMO-induced inflammation. We specifically aimed to test whether intravenous treatment with α1-antitrypsin reduces the release of cytokines in response to 2 h of experimental ECMO. Adult rats were intravenously infused with α1-antitrypsin immediately before starting veno-arterial ECMO. We measured selected pro- and anti-inflammatory cytokines and found, that systemic levels of tumor necrosis factor-α, interleukin-6 and interleukin-10 increase during experimental ECMO. As tachycardia and hypertension developed in response to α1-antitrypsin, a single additional bolus of fentanyl and midazolam was given. Treatment with α1-antitrypsin and higher sedative doses reduced all cytokine levels investigated. We suggest that α1-antitrypsin might have the potential to protect against both ECMO-induced systemic inflammation and immune paralysis. More studies are needed to corroborate our findings, to clarify the mechanisms by which α1-antitrypsin inhibits cytokine release in vivo and to explore the potential application of α1-antitrypsin in clinical ECMO.
Collapse
Affiliation(s)
- Fabian Edinger
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Justus-Liebig University of Giessen, Giessen, Germany.
| | - Christoph Schmitt
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Justus-Liebig University of Giessen, Giessen, Germany
| | - Christian Koch
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Justus-Liebig University of Giessen, Giessen, Germany
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- Department of Biology, University of Utah, Salt Lake City, UT, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, German Centre for Lung Research (DZL), Hannover, Germany
| | - Melanie Markmann
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Justus-Liebig University of Giessen, Giessen, Germany
| | - Michael Sander
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Justus-Liebig University of Giessen, Giessen, Germany
| | - Winfried Padberg
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, German Centre for Lung Research (DZL), Justus-Liebig-University of Giessen, Giessen, Germany
| | - Veronika Grau
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, German Centre for Lung Research (DZL), Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW To review recent evidence on the capacity of vitamin D to prevent atopic disease, focussing on food allergy and asthma, and potential underlying mechanisms. RECENT FINDINGS The incidence of allergic disease continues to increase worldwide. Vitamin D status is influenced by sun exposure and dietary intake. Vitamin D deficiency is linked to an increased incidence of allergic disease and asthma. These associations are generally strongest in early life. The capacity of vitamin D to enhance antimicrobial pathways, promote peripheral immunological tolerance and maintain mucosal barrier integrity may underlie these associations. Interventional studies have addressed the capacity of vitamin D supplementation in utero and early life to reduce the incidence of disease. Ancillary studies have provided insights into potential biological mechanisms linked to these effects. SUMMARY Observational studies show an inverse association between vitamin D levels and development of food allergy and asthma. Secondary analyses of two recent interventional studies suggest that achieving vitamin D sufficiency throughout pregnancy reduces the incidence of asthma/recurrent wheeze at 3 years. Longitudinal studies of vitamin D requirements in utero and postnatally, better understanding of factors that influence bioavailability of vitamin D and mechanistic insights into vitamin D effects on neonatal-specific immune pathways are awaited.
Collapse
|
28
|
Does Genetic Predisposition Contribute to the Exacerbation of COVID-19 Symptoms in Individuals with Comorbidities and Explain the Huge Mortality Disparity between the East and the West? Int J Mol Sci 2021; 22:ijms22095000. [PMID: 34066804 PMCID: PMC8125927 DOI: 10.3390/ijms22095000] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022] Open
Abstract
The elderly and patients with several comorbidities experience more severe cases of coronavirus disease 2019 (COVID-19) than healthy patients without underlying medical conditions. However, it is unclear why these people are prone to developing alveolar pneumonia, rapid exacerbations, and death. Therefore, we hypothesized that people with comorbidities may have a genetic predisposition that makes them more vulnerable to various factors; for example, they are likely to become more severely ill when infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To test this hypothesis, we searched the literature extensively. Polymorphisms of genes, such as those that encode angiotensin-converting enzyme 1 (ACE1), have been associated with numerous comorbidities, such as cardiovascular disease, hypertension, diabetes, chronic kidney disease, and obesity, and there are potential mechanisms to explain these associations (e.g., DD-type carriers have greater ACE1 activity, and patients with a genetic alpha-1 anti-trypsin (AAT) deficiency lack control over inflammatory mediators). Since comorbidities are associated with chronic inflammation and are closely related to the renin–angiotensin–aldosterone system (RAAS), these individuals may already have a mild ACE1/ACE2 imbalance before viral infection, which increases their risk for developing severe cases of COVID-19. However, there is still much debate about the association between ACE1 D/I polymorphism and comorbidities. The best explanation for this discrepancy could be that the D allele and DD subtypes are associated with comorbidities, but the DD genotype alone does not have an exceptionally large effect. This is also expected since the ACE1 D/I polymorphism is only an intron marker. We also discuss how polymorphisms of AAT and other genes are involved in comorbidities and the severity of SARS-CoV-2 infection. Presumably, a combination of multiple genes and non-genetic factors is involved in the establishment of comorbidities and aggravation of COVID-19.
Collapse
|
29
|
McCormack M, Talbot A, Dillon E, O’Connor I, MacCarthy E. Host Response of Atlantic Salmon ( Salmo salar) Re-Inoculated with Paramoeba perurans. Microorganisms 2021; 9:microorganisms9050993. [PMID: 34062978 PMCID: PMC8147987 DOI: 10.3390/microorganisms9050993] [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: 03/24/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 11/28/2022] Open
Abstract
In aquaculture, recurrence rates of amoebic gill disease (AGD) caused by the ectoparasite Paramoeba perurans are high and no prophylactic strategies exist for disease prevention. In this study, Atlantic salmon (Salmo salar) were initially inoculated with P. perurans and following the development of amoebic gill disease were treated with freshwater immersion on day 21 and day 35 post inoculation. Fish were re-inoculated following a negative qPCR analysis for the presence of P. perurans. The gill host immune response was investigated at 7, 14, and 18 days post re-inoculation. Differential proteome expression of immune related proteins was assessed by comparison of each time point against naïve controls. In the gill, some proteins of the innate immune system were expressed in response to gill re-colonization by P. perurans, while no features of adaptive immunity were found to be differentially expressed. Many of the proteins identified are novel in the context of AGD and their expression profiles suggest that their roles in the response to disease development and progression in single or multiple infections warrant further investigation.
Collapse
Affiliation(s)
- Michelle McCormack
- Marine and Freshwater Research Centre, Galway Mayo Institute of Technology, Dublin Road, H91 TRNW Galway, Ireland; (A.T.); (I.O.); (E.M.)
- Correspondence:
| | - Anita Talbot
- Marine and Freshwater Research Centre, Galway Mayo Institute of Technology, Dublin Road, H91 TRNW Galway, Ireland; (A.T.); (I.O.); (E.M.)
| | - Eugene Dillon
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland;
| | - Ian O’Connor
- Marine and Freshwater Research Centre, Galway Mayo Institute of Technology, Dublin Road, H91 TRNW Galway, Ireland; (A.T.); (I.O.); (E.M.)
| | - Eugene MacCarthy
- Marine and Freshwater Research Centre, Galway Mayo Institute of Technology, Dublin Road, H91 TRNW Galway, Ireland; (A.T.); (I.O.); (E.M.)
| |
Collapse
|
30
|
Stanke F, Janciauskiene S, Tamm S, Wrenger S, Raddatz EL, Jonigk D, Braubach P. Effect of Alpha-1 Antitrypsin on CFTR Levels in Primary Human Airway Epithelial Cells Grown at the Air-Liquid-Interface. Molecules 2021; 26:molecules26092639. [PMID: 33946490 PMCID: PMC8125203 DOI: 10.3390/molecules26092639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) gene is influenced by the fundamental cellular processes like epithelial differentiation/polarization, regeneration and epithelial–mesenchymal transition. Defects in CFTR protein levels and/or function lead to decreased airway surface liquid layer facilitating microbial colonization and inflammation. The SERPINA1 gene, encoding alpha1-antitrypsin (AAT) protein, is one of the genes implicated in CF, however it remains unknown whether AAT has any influence on CFTR levels. In this study we assessed CFTR protein levels in primary human lung epithelial cells grown at the air-liquid-interface (ALI) alone or pre-incubated with AAT by Western blots and immunohistochemistry. Histological analysis of ALI inserts revealed CFTR- and AAT-positive cells but no AAT-CFTR co-localization. When 0.5 mg/mL of AAT was added to apical or basolateral compartments of pro-inflammatory activated ALI cultures, CFTR levels increased relative to activated ALIs. This finding suggests that AAT is CFTR-modulating protein, albeit its effects may depend on the concentration and the route of administration. Human lung epithelial ALI cultures provide a useful tool for studies in detail how AAT or other pharmaceuticals affect the levels and activity of CFTR.
Collapse
Affiliation(s)
- Frauke Stanke
- Department of Pediatric Pneumology, Neonatology and Allergology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.T.); (E.L.R.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Correspondence: ; Tel.: +49-511-5326722
| | - Sabina Janciauskiene
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Department of Respiratory Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stephanie Tamm
- Department of Pediatric Pneumology, Neonatology and Allergology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.T.); (E.L.R.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
| | - Sabine Wrenger
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Department of Respiratory Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Ellen Luise Raddatz
- Department of Pediatric Pneumology, Neonatology and Allergology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.T.); (E.L.R.)
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Peter Braubach
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
31
|
Correlation of Low Levels of α-1 Antitrypsin and Elevation of Neutrophil to Lymphocyte Ratio with Higher Mortality in Severe COVID-19 Patients. Mediators Inflamm 2021; 2021:5555619. [PMID: 34007243 PMCID: PMC8080869 DOI: 10.1155/2021/5555619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/29/2021] [Accepted: 04/17/2021] [Indexed: 12/15/2022] Open
Abstract
Background Variations in COVID-19 prevalence, severity, and mortality rate remain ambiguous. Genetic or individual differences in immune response may be an explanation. Moreover, hyperinflammation and dysregulated immune response are involved in the etiology of severe forms of COVID-19. Therefore, the aim of the present study was to analyze serum alpha-1 antitrypsin (AAT) levels, as an acute-phase plasma protein with immunomodulatory effect and neutrophil to lymphocyte ratio (NLR) as a marker of inflammation response in severe COVID-19 illness. Methods In this retrospective observational cohort study, 64 polymerase chain reaction (PCR) positive COVID-19 hospitalized patients were studied for AAT, C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), troponin, complete blood count (CBC), random blood sugar, serum glutamate oxaloacetate transaminase (SGOT), serum glutamate pyruvate transaminase (SGPT), and arterial oxygen saturation (O2sat) at admission and during hospitalization. Results The results showed that hospitalized patients with COVID-19 had low serum levels of AAT and high CRP levels at the first days of hospitalization. In particular, the percentages of individuals with low, normal, and high AAT levels were 7.80%, 82.80%, and 9.40%, respectively, while high and low values of CRP accounted for 86.70% and 13.30% of patients. Most of the patients had an upward neutrophil to lymphocyte ratio (NLR) trend, with a higher mortality rate (p < 0.05) and troponin levels (p < 0.05). However, comorbidities, CRP alterations, ESR alterations, nonfasting blood sugar, SGOT, SGPT, O2sat, RBC, and PLT values were not significantly different between the NLR downward and upward trend groups. Conclusions The current study revealed that severe COVID-19 patients had low serum AAT levels related to CRP values. Therefore, AAT response may be considered as a new mechanism by which some COVID-19 patients show immune dysregulation and more severe symptoms.
Collapse
|
32
|
Azouz NP, Klingler AM, Callahan V, Akhrymuk IV, Elez K, Raich L, Henry BM, Benoit JL, Benoit SW, Noé F, Kehn-Hall K, Rothenberg ME. Alpha 1 Antitrypsin is an Inhibitor of the SARS-CoV-2-Priming Protease TMPRSS2. Pathog Immun 2021; 6:55-74. [PMID: 33969249 PMCID: PMC8097828 DOI: 10.20411/pai.v6i1.408] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Host proteases have been suggested to be crucial for dissemination of MERS, SARS-CoV, and SARS-CoV-2 coronaviruses, but the relative contribution of membrane versus intracellular proteases remains controversial. Transmembrane serine protease 2 (TMPRSS2) is regarded as one of the main proteases implicated in the coronavirus S protein priming, an important step for binding of the S protein to the angiotensin-converting enzyme 2 (ACE2) receptor before cell entry. METHODS We developed a cell-based assay to identify TMPRSS2 inhibitors. Inhibitory activity was established in SARS-CoV-2 viral load systems. RESULTS We identified the human extracellular serine protease inhibitor (serpin) alpha 1 anti-trypsin (A1AT) as a novel TMPRSS2 inhibitor. Structural modeling revealed that A1AT docked to an extracellular domain of TMPRSS2 in a conformation that is suitable for catalysis, resembling similar serine protease inhibitor complexes. Inhibitory activity of A1AT was established in a SARS-CoV-2 viral load system. Notably, plasma A1AT levels were associated with COVID-19 disease severity. CONCLUSIONS Our data support the key role of extracellular serine proteases in SARS CoV-2 infections and indicate that treatment with serpins, particularly the FDA-approved drug A1AT, may be effective in limiting SARS-CoV-2 dissemination by affecting the surface of the host cells.
Collapse
Affiliation(s)
- Nurit P. Azouz
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Andrea M. Klingler
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Victoria Callahan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA
| | - Ivan V. Akhrymuk
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Katarina Elez
- Freie Universität Berlin, Department of Mathematics and Computer Science, Berlin, Germany
| | - Lluís Raich
- Freie Universität Berlin, Department of Mathematics and Computer Science, Berlin, Germany
| | - Brandon M. Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Justin L. Benoit
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, OH
| | - Stefanie W. Benoit
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Frank Noé
- Freie Universität Berlin, Department of Mathematics and Computer Science, Berlin, Germany
- Freie Universität Berlin, Department of Physics, Berlin, Germany
- Rice University, Department of Chemistry, Houston, TX
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
33
|
de Loyola MB, dos Reis TTA, de Oliveira GXLM, da Fonseca Palmeira J, Argañaraz GA, Argañaraz ER. Alpha-1-antitrypsin: A possible host protective factor against Covid-19. Rev Med Virol 2021; 31:e2157. [PMID: 32844538 PMCID: PMC7461031 DOI: 10.1002/rmv.2157] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022]
Abstract
Understanding Covid-19 pathophysiology is crucial for a better understanding of the disease and development of more effective treatments. Alpha-1-antitrypsin (A1AT) is a constitutive tissue protector with antiviral and anti-inflammatory properties. A1AT inhibits SARS-CoV-2 infection and two of the most important proteases in the pathophysiology of Covid-19: the transmembrane serine protease 2 (TMPRSS2) and the disintegrin and metalloproteinase 17 (ADAM17). It also inhibits the activity of inflammatory molecules, such as IL-8, TNF-α, and neutrophil elastase (NE). TMPRSS2 is essential for SARS-CoV-2-S protein priming and viral infection. ADAM17 mediates ACE2, IL-6R, and TNF-α shedding. ACE2 is the SARS-CoV-2 entry receptor and a key component for the balance of the renin-angiotensin system, inflammation, vascular permeability, and pulmonary homeostasis. In addition, clinical findings indicate that A1AT levels might be important in defining Covid-19 outcomes, potentially partially explaining associations with air pollution and with diabetes. In this review, we focused on the interplay between A1AT with TMPRSS2, ADAM17 and immune molecules, and the role of A1AT in the pathophysiology of Covid-19, opening new avenues for investigating effective treatments.
Collapse
Affiliation(s)
| | | | | | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Faculty of Health ScienceUniversity of BrasíliaBrasiliaBrazil
| | - Gustavo A. Argañaraz
- Laboratory of Molecular Neurovirology, Faculty of Health ScienceUniversity of BrasíliaBrasiliaBrazil
| | - Enrique R. Argañaraz
- Laboratory of Molecular Neurovirology, Faculty of Health ScienceUniversity of BrasíliaBrasiliaBrazil
| |
Collapse
|
34
|
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
|
35
|
Golizeh M, Winter K, Roussel L, Landekic M, Langelier M, Loo VG, Ndao M, Vinh DC. Fecal host biomarkers predicting severity of Clostridioides difficile infection. JCI Insight 2021; 6:142976. [PMID: 33232301 PMCID: PMC7821589 DOI: 10.1172/jci.insight.142976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Clostridioides difficile is a major cause of health care-associated diarrhea. Severity ranges from mild to life-threatening, but this variability remains poorly understood. Microbiologic diagnosis of C. difficile infection (CDI) is straightforward but offers little insight into the patient's prognosis or into pathophysiologic determinants of clinical trajectory. The aim of this study was to discover host-derived, CDI-specific fecal biomarkers involved in disease severity. Subjects without and with CDI diarrhea were recruited. CDI severity was based on Infectious Diseases Society of America/Society for Healthcare Epidemiology of America criteria. We developed a liquid chromatography tandem mass spectrometry approach to identify host-derived protein biomarkers from stool and applied it to diagnostic samples for cohort-wise comparison (CDI-negative vs. nonsevere CDI vs. severe CDI). Selected biomarkers were orthogonally confirmed and subsequently verified in a CDI mouse model. We identified a protein signature from stool, consisting of alpha-2-macroglobulin (A2MG), matrix metalloproteinase-7 (MMP-7), and alpha-1-antitrypsin (A1AT), that not only discriminates CDI-positive samples from non-CDI ones but also is potentially associated with disease severity. In the mouse model, this signature with the murine homologs of the corresponding proteins was also identified. A2MG, MMP-7, and A1AT serve as biomarkers in patients with CDI and define novel components of the host response that may determine disease severity.
Collapse
Affiliation(s)
- Makan Golizeh
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
| | - Kaitlin Winter
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Department of Microbiology & Immunology and
| | - Lucie Roussel
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Host-directed Immunotherapy to Fight Infectious disease (HI-FI) Program, Montréal, Québec, Canada
| | - Marija Landekic
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Department of Microbiology & Immunology and.,Host-directed Immunotherapy to Fight Infectious disease (HI-FI) Program, Montréal, Québec, Canada
| | - Mélanie Langelier
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Host-directed Immunotherapy to Fight Infectious disease (HI-FI) Program, Montréal, Québec, Canada
| | - Vivian G Loo
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Department of Microbiology & Immunology and.,Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, Quebéc, Canada.,Host-directed Immunotherapy to Fight Infectious disease (HI-FI) Program, Montréal, Québec, Canada.,Division of Medical Microbiology, Department of Laboratory Medicine, MUHC, Montréal, Québec, Canada
| | - Momar Ndao
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Department of Microbiology & Immunology and.,Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, Quebéc, Canada
| | - Donald C Vinh
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada.,Department of Microbiology & Immunology and.,Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, Quebéc, Canada.,Host-directed Immunotherapy to Fight Infectious disease (HI-FI) Program, Montréal, Québec, Canada.,Division of Medical Microbiology, Department of Laboratory Medicine, MUHC, Montréal, Québec, Canada
| |
Collapse
|
36
|
Gioia M, Ciaccio C, Calligari P, De Simone G, Sbardella D, Tundo G, Fasciglione GF, Di Masi A, Di Pierro D, Bocedi A, Ascenzi P, Coletta M. Role of proteolytic enzymes in the COVID-19 infection and promising therapeutic approaches. Biochem Pharmacol 2020; 182:114225. [PMID: 32956643 PMCID: PMC7501082 DOI: 10.1016/j.bcp.2020.114225] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
In the Fall of 2019 a sudden and dramatic outbreak of a pulmonary disease (Coronavirus Disease COVID-19), due to a new Coronavirus strain (i.e., SARS-CoV-2), emerged in the continental Chinese area of Wuhan and quickly diffused throughout the world, causing up to now several hundreds of thousand deaths. As for common viral infections, the crucial event for the viral life cycle is the entry of genetic material inside the host cell, realized by the spike protein of the virus through its binding to host receptors and its activation by host proteases; this is followed by translation of the viral RNA into a polyprotein, exploiting the host cell machinery. The production of individual mature viral proteins is pivotal for replication and release of new virions. Several proteolytic enzymes either of the host and of the virus act in a concerted fashion to regulate and coordinate specific steps of the viral replication and assembly, such as (i) the entry of the virus, (ii) the maturation of the polyprotein and (iii) the assembly of the secreted virions for further diffusion. Therefore, proteases involved in these three steps are important targets, envisaging that molecules which interfere with their activity are promising therapeutic compounds. In this review, we will survey what is known up to now on the role of specific proteolytic enzymes in these three steps and of most promising compounds designed to impair this vicious cycle.
Collapse
Key Words
- covid-19, coronavirus disease – 19
- sars-cov, severe acute respiratory syndrome coronavirus
- sars-cov-2, severe acute respiratory syndrome – 2
- mers-cov, middle east respiratory syndrome coronavirus
- orf, open reading frame
- plpro, papain-like protease
- mpro, main protease
- pp, polyprotein
- nsp, non structural protein
- rdrp, rna dependent rna polymerase
- hel, helicase
- s protein, spike protein
- tmprss2, trans-membrane protease serine protease-2
- tmprss4, trans-membrane protease serine protease-4
- hat, human airway trypsin-like protease
- tgn, trans-golgi network
- ace2, angiotensin-converting enzyme receptor-2
- rbd, receptor binding domain
- pc, pro-protein convertase
- hcov-oc43, human coronavirus-oc43
- mhv-a59, murine hepatitis virus – a59
- hiv, human immunodeficiency virus
- cmk, chloro-methyl-ketone
- dec, decanoyl
- phac, phenyl-acetyl
- ttsp, type ii transmembrane serine proteases family
- hpv, human papillomavirus
- hbv, hepatitis b virus
- evd, ebola virus disease
- zikv, zika virus
- jev, japanese encephalitis virus
- fpv, feline panleukopenia virus
- hpaiv, highly pathogenic avian influenza virus
- cdv, canine distemper virus
- rsv, respiratory syncytial virus (rsv)
- a1at, alpha-1-anti trypsin
- aebsf, 4-(2-aminomethyl)-benzene sulphonyl fluoride
- bhh, bromhexine hydrochloride
- pcsk, pro-protein convertase subtilisin/kexin
- ampk, adenosine monophosphate-activated protein kinase
- hcov-nl63, human coronavirus – nl63
- hcov-229e, human coronavirus – 229e
- hcov-hku1, human coronavirus – hku1
- 3cpro, 3chymotrypsin protease of rhinoviruses
- 3d-qsar, three-dimensional quantitative structure-activity relationships
- fda, food and drug agency
Collapse
Affiliation(s)
- Magda Gioia
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Roma, Italy.
| | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Roma, Italy.
| | - Paolo Calligari
- Department of Chemical and Technological Sciences, University of Roma Tor Vergata, Roma, Italy
| | | | | | | | | | | | - Donato Di Pierro
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Roma, Italy
| | - Alessio Bocedi
- Department of Chemical and Technological Sciences, University of Roma Tor Vergata, Roma, Italy
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, Roma, Italy,Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, Roma, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Roma, Italy.
| |
Collapse
|
37
|
Martini F, De Mattei M, Contini C, Tognon MG. Potential Use of Alpha-1 Anti-trypsin in the Covid-19 Treatment. Front Cell Dev Biol 2020; 8:577528. [PMID: 33195215 PMCID: PMC7644540 DOI: 10.3389/fcell.2020.577528] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Fernanda Martini
- Laboraotories of Cell Biology and Molecular Genetics, University of Ferrara, Ferrara, Italy
| | - Monica De Mattei
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Carlo Contini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro G. Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
38
|
Azouz NP, Klingler AM, Callahan V, Akhrymuk IV, Elez K, Raich L, Henry BM, Benoit JL, Benoit SW, Noé F, Kehn-Hall K, Rothenberg ME. Alpha 1 Antitrypsin is an Inhibitor of the SARS-CoV-2-Priming Protease TMPRSS2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.05.04.077826. [PMID: 33052338 PMCID: PMC7553163 DOI: 10.1101/2020.05.04.077826] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Host proteases have been suggested to be crucial for dissemination of MERS, SARS-CoV, and SARS-CoV-2 coronaviruses, but the relative contribution of membrane versus intracellular proteases remains controversial. Transmembrane serine protease 2 (TMPRSS2) is regarded as one of the main proteases implicated in the coronavirus S protein priming, an important step for binding of the S protein to the angiotensin-converting enzyme 2 (ACE2) receptor before cell entry. The main cellular location where the SARS-CoV-2 S protein priming occurs remains debatable, therefore hampering the development of targeted treatments. Herein, we identified the human extracellular serine protease inhibitor (serpin) alpha 1 antitrypsin (A1AT) as a novel TMPRSS2 inhibitor. Structural modeling revealed that A1AT docked to an extracellular domain of TMPRSS2 in a conformation that is suitable for catalysis, resembling similar serine protease-inhibitor complexes. Inhibitory activity of A1AT was established in a SARS-CoV-2 viral load system. Notably, plasma A1AT levels were associated with COVID-19 disease severity. Our data support the key role of extracellular serine proteases in SARS-CoV-2 infections and indicate that treatment with serpins, particularly the FDA-approved drug A1AT, may be effective in limiting SARS-CoV-2 dissemination by affecting the surface of the host cells. SUMMARY Delivery of extracellular serine protease inhibitors (serpins) such as A1AT has the capacity to reduce SARS-CoV-2 dissemination by binding and inhibiting extracellular proteases on the host cells, thus, inhibiting the first step in SARS-CoV-2 cell cycle (i.e. cell entry).
Collapse
Affiliation(s)
- Nurit P. Azouz
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229-3026, USA
| | - Andrea M. Klingler
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229-3026, USA
| | - Victoria Callahan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Ivan V. Akhrymuk
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Katarina Elez
- Freie Universität Berlin, Department of Mathematics and Computer Science, Berlin, Germany
| | - Lluís Raich
- Freie Universität Berlin, Department of Mathematics and Computer Science, Berlin, Germany
| | - Brandon M. Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Justin L. Benoit
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Stefanie W. Benoit
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, OH, USA
| | - Frank Noé
- Freie Universität Berlin, Department of Mathematics and Computer Science, Berlin, Germany
- Freie Universität Berlin, Department of Physics, Berlin, Germany
- Rice University, Department of Chemistry, Houston, TX
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229-3026, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, OH, USA
| |
Collapse
|
39
|
Gibbs HL, Sanz L, Pérez A, Ochoa A, Hassinger ATB, Holding ML, Calvete JJ. The molecular basis of venom resistance in a rattlesnake-squirrel predator-prey system. Mol Ecol 2020; 29:2871-2888. [PMID: 32593182 DOI: 10.1111/mec.15529] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 06/11/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
Understanding how interspecific interactions mould the molecular basis of adaptations in coevolving species is a long-sought goal of evolutionary biology. Venom in predators and venom resistance proteins in prey are coevolving molecular phenotypes, and while venoms are highly complex mixtures it is unclear if prey respond with equally complex resistance traits. Here, we use a novel molecular methodology based on protein affinity columns to capture and identify candidate blood serum resistance proteins ("venom interactive proteins" [VIPs]) in California Ground Squirrels (Otospermophilus beecheyi) that interact with venom proteins from their main predator, Northern Pacific Rattlesnakes (Crotalus o. oreganus). This assay showed that serum-based resistance is both population- and species-specific, with serum proteins from ground squirrels showing higher binding affinities for venom proteins of local snakes compared to allopatric individuals. Venom protein specificity assays identified numerous and diverse candidate prey resistance VIPs but also potential targets of venom in prey tissues. Many specific VIPs bind to multiple snake venom proteins and, conversely, single venom proteins bind multiple VIPs, demonstrating that a portion of the squirrel blood serum "resistome" involves broad-based inhibition of nonself proteins and suggests that resistance involves a toxin scavenging mechanism. Analyses of rates of evolution of VIP protein homologues in related mammals show that most of these proteins evolve under purifying selection possibly due to molecular constraints that limit the evolutionary responses of prey to rapidly evolving snake venom proteins. Our method represents a general approach to identify specific proteins involved in co-evolutionary interactions between species at the molecular level.
Collapse
Affiliation(s)
- H Lisle Gibbs
- Department of Evolution, Ecology, and Organismal Biology, Ohio State University, Columbus, OH, USA
| | - Libia Sanz
- Evolutionary and Translational Venomics Laboratory, CSIC, Valencia, Spain
| | - Alicia Pérez
- Evolutionary and Translational Venomics Laboratory, CSIC, Valencia, Spain
| | - Alexander Ochoa
- Department of Evolution, Ecology, and Organismal Biology, Ohio State University, Columbus, OH, USA
| | - Alyssa T B Hassinger
- Department of Evolution, Ecology, and Organismal Biology, Ohio State University, Columbus, OH, USA
| | - Matthew L Holding
- Department of Evolution, Ecology, and Organismal Biology, Ohio State University, Columbus, OH, USA.,Department of Biological Sciences, Florida State University, Tallahassee, FL, USA
| | - Juan J Calvete
- Evolutionary and Translational Venomics Laboratory, CSIC, Valencia, Spain
| |
Collapse
|
40
|
Plasma-Based Proteomics Profiling of Patients with Hyperthyroidism after Antithyroid Treatment. Molecules 2020; 25:molecules25122831. [PMID: 32575434 PMCID: PMC7356574 DOI: 10.3390/molecules25122831] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022] Open
Abstract
Thyroid hormones critically modulate body homeostasis and haemostasis by regulating energy and metabolism. Previous studies have focused on individual pathways or proteins that are affected by increases in thyroid hormone levels, while an overall plasma proteomic signature of this increased level is lacking. Herein, an integrated untargeted proteomic approach with network analysis was used to identify changes in circulating proteins in the plasma proteome between hyperthyroid and euthyroid states. Plasma from 10 age-matched subjects at baseline (hyperthyroid) and post treatment with carbimazole (euthyroid) was compared by difference gel electrophoresis (DIGE) and matrix-assisted laser desorption/ionization time of flight (MALDI TOF) mass spectrometry (MS). A total of 20 proteins were identified with significant difference in abundance (analysis of variance (ANOVA) test, p ≤ 0.05; fold-change ≥ 1.5) between the two states (12 increased and 8 decreased in abundance in the hyperthyroid state). Twelve protein spots corresponding to ten unique proteins were significantly more abundant in the hyperthyroid state compared with the euthyroid state. These increased proteins were haptoglobin (HP), hemopexin (HPX), clusterin (CLU), apolipoprotein L1 (APOL1), alpha-1-B glycoprotein (A1BG), fibrinogen gamma chain (FGG), Ig alpha-1 chain C region (IGHA1), complement C6 (C6), leucine rich alpha 2 glycoprotein (LRG1), and carboxypeptidase N catalytic chain (CPN1). Eight protein spots corresponding to six unique proteins were significantly decreased in abundance in the hyperthyroid samples compared with euthyroid samples. These decreased proteins were apolipoprotein A1 (APOA1), inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4), plasminogen (PLG), alpha-1 antitrypsin (SERPINA1), fibrinogen beta chain (FGB), and complement C1r subcomponent (C1R). The differentially abundant proteins were investigated by ingenuity pathway analysis (IPA). The network pathway identified related to infectious disease, inflammatory disease, organismal injury and abnormalities, and the connectivity map focused around two central nodes, namely the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and p38 mitogen-activated protein kinase (MAPK) pathways. The plasma proteome of patients with hyperthyroidism revealed differences in the abundance of proteins involved in acute phase response signaling, and development of a hypercoagulable and hypofibrinolytic state. Our findings enhance our existing knowledge of the altered proteins and associated biochemical pathways in hyperthyroidism.
Collapse
|
41
|
Lior Y, Jasevitch M, Ochayon DE, Zaretsky M, Lewis EC, Aharoni A. Application of directed evolution and back-to-consensus algorithms to human alpha1-antitrypsin leads to diminished anti-protease activity and augmented anti-inflammatory activities. Cell Immunol 2020; 355:104135. [PMID: 32703529 DOI: 10.1016/j.cellimm.2020.104135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022]
Abstract
Primarily known as an elastase inhibitor, human alpha1-antitrypsin also exerts anti-inflammatory and immunomodulatory effects, both in vitro and in vivo. While the anti-protease mechanism of alpha1-antitrypsin is attributed to a particular protein domain coined the reactive center loop, anti-inflammatory and immunomodulatory loci within the molecule remain to be identified. In the present study, directed evolution and back-to-consensus algorithms were applied to human alpha1-antitrypsin. Six unique functional candidate sites were identified on the surface of the molecule; in manipulating these sites by point mutations, a recombinant mutant form of alpha1-antitrypsin was produced, depicting a requirement for sites outside the reactive center loop as essential for protease inhibition, and displaying enhanced anti-inflammatory activities. Taken together, outcomes of the present study establish a potential use for directed evolution in advancing our understanding of site-specific protein functions, offering a platform for development of context- and disease-specific alpha1-antitrypsin-based therapeutics.
Collapse
Affiliation(s)
- Yotam Lior
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Maria Jasevitch
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - David E Ochayon
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Mariana Zaretsky
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Amir Aharoni
- Department of Life Sciences, Ben-Gurion University of the Negev and National Institute for Biotechnology, Be'er Sheva, Israel
| |
Collapse
|
42
|
Abstract
Alpha-1 Antitrypsin Deficiency (A1AD) is a hereditary condition characterized by low levels of circulating alpha-antitrypsin (AAT) in plasma. It is the best understood genetic risk factor for the development of chronic obstructive pulmonary disease (COPD). The diagnosis of A1AD is under-recognized. While there is a significant heterogeneity in disease presentation in relation to the severity of symptoms and prognosis, it is not uncommon for young individuals, including pregnant women to already have moderate to advanced lung disease at the time of diagnosis. Reductions in AAT levels may have unique implications for a gravid patient beyond that of lung disease. Care of the pregnant A1AD patient with chronic lung disease follows the principles of care for the management of airways disease in general with control of symptoms and reduction in exacerbation risk the main tenets of treatment. The effect of A1AD and augmentation in pregnancy has not been studied and thus care is reliant on expert opinion and clinical experience. Providers caring for pregnant patients with A1AD should consider referral to health care systems and providers with specific expertise in A1AD. Ultimately the decision is left to the individual patient and their physician to weigh the risk benefit of cessation or continuation of therapies. In this review, we present the perinatal course of a woman with A1AD and review the available literature pertaining to AAT and pregnancy and discuss the clinical implications.
Collapse
Affiliation(s)
- Nate T Gaeckle
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laurel Stephenson
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ronald A Reilkoff
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
43
|
Phillips RA, Kraev I, Lange S. Protein Deimination and Extracellular Vesicle Profiles in Antarctic Seabirds. BIOLOGY 2020; 9:E15. [PMID: 31936359 PMCID: PMC7168935 DOI: 10.3390/biology9010015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/19/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Pelagic seabirds are amongst the most threatened of all avian groups. They face a range of immunological challenges which seem destined to increase due to environmental changes in their breeding and foraging habitats, affecting prey resources and exposure to pollution and pathogens. Therefore, the identification of biomarkers for the assessment of their health status is of considerable importance. Peptidylarginine deiminases (PADs) post-translationally convert arginine into citrulline in target proteins in an irreversible manner. PAD-mediated deimination can cause structural and functional changes in target proteins, allowing for protein moonlighting in physiological and pathophysiological processes. PADs furthermore contribute to the release of extracellular vesicles (EVs), which play important roles in cellular communication. In the present study, post-translationally deiminated protein and EV profiles of plasma were assessed in eight seabird species from the Antarctic, representing two avian orders: Procellariiformes (albatrosses and petrels) and Charadriiformes (waders, auks, gulls and skuas). We report some differences between the species assessed, with the narrowest EV profiles of 50-200 nm in the northern giant petrel Macronectes halli, and the highest abundance of larger 250-500 nm EVs in the brown skua Stercorarius antarcticus. The seabird EVs were positive for phylogenetically conserved EV markers and showed characteristic EV morphology. Post-translational deimination was identified in a range of key plasma proteins critical for immune response and metabolic pathways in three of the bird species under study; the wandering albatross Diomedea exulans, south polar skua Stercorarius maccormicki and northern giant petrel. Some differences in Gene Ontology (GO) biological and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways for deiminated proteins were observed between these three species. This indicates that target proteins for deimination may differ, potentially contributing to a range of physiological functions relating to metabolism and immune response, as well as to key defence mechanisms. PAD protein homologues were identified in the seabird plasma by Western blotting via cross-reaction with human PAD antibodies, at an expected 75 kDa size. This is the first study to profile EVs and to identify deiminated proteins as putative novel plasma biomarkers in Antarctic seabirds. These biomarkers may be further refined to become useful indicators of physiological and immunological status in seabirds-many of which are globally threatened.
Collapse
Affiliation(s)
- Richard A. Phillips
- British Antarctic Survey, Natural Environment Research Council, Cambridge CB3 0ET, UK;
| | - Igor Kraev
- Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes MK7 6AA, UK;
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK
| |
Collapse
|
44
|
Brami I, Ini D, Sassonker N, Zaknoun M, Zuckerman T, Lewis EC. Immunosuppressive Drugs Alter α1-Antitrypsin Production in Hepatocytes: Implications for Epithelial Gap Repair. Biol Blood Marrow Transplant 2019; 26:625-633. [PMID: 31899361 DOI: 10.1016/j.bbmt.2019.12.764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/18/2019] [Accepted: 12/25/2019] [Indexed: 01/04/2023]
Abstract
Immunosuppressive drugs are an inherent component of hematopoietic stem cell transplantation (HSCT) for the prevention of acute graft-versus-host disease (GVHD). Circulating α1-antitrypsin (AAT), a serine-protease inhibitor produced predominantly by hepatocytes that rises during acute phase responses, is lost in patient's stool due to gastrointestinal GVHD, and its augmentation has been found to attenuate GVHD. Here we explored the effect of immunosuppressive drugs on hepatocyte production of AAT and intestinal epithelial gap repair. The effect of commonly used immunosuppressants on AAT production was examined in vitro using HepG2 cells and primary mouse hepatocytes, and their impact on human intestinal epithelial cell line gap repair was evaluated. Sera from 12 allogeneic HSCT recipients, obtained at 14 days post-transplantation, predating the diagnosis of GVHD (n = 6), were examined for reepithelialization, with added clinical-grade AAT. Rapamycin compromised AAT production under inflammatory conditions. Mycophenolate mofetil and cyclosporine A (CSA) inhibited reepithelialization; AAT minimized the effect of CSA. Patient sera displayed superior gap repair with exogenous AAT. Functional insufficiency in circulating AAT may be the result of drug toxicities leading to ineffective gut reepithelization and compromised gut lining. Taken together, our data strengthen the rationale for incorporating AAT augmentation therapy into immunosuppressive treatment protocols.
Collapse
Affiliation(s)
- Ido Brami
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel.
| | - Dor Ini
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| | - Nofit Sassonker
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| | - Melodie Zaknoun
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| | - Tsila Zuckerman
- Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Be'er Sheva, Israel
| |
Collapse
|
45
|
A Novel Small Molecule Inhibits Intrahepatocellular Accumulation of Z-Variant Alpha 1-Antitrypsin In Vitro and In Vivo. Cells 2019; 8:cells8121586. [PMID: 31817705 PMCID: PMC6953066 DOI: 10.3390/cells8121586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/01/2019] [Accepted: 12/03/2019] [Indexed: 11/30/2022] Open
Abstract
Alpha 1-antitrypsin deficiency (AATD) is the most common genetic cause of liver disease in children and is associated with early-onset chronic liver disease in adults. AATD associated liver injury is caused by hepatotoxic retention of polymerized mutant alpha 1-antitrypsin molecules within the endoplasmic reticulum. Currently, there is no curative therapy for AATD. In this study, we selected small molecules with the potential to bind mutant alpha 1-antitrypsin (Z-variant) to inhibit its accumulation in hepatocytes. We used molecular docking to select candidate compounds that were validated in cell and animal models of disease. A crystal structure of polymerized alpha 1-antitrypsin molecule was used as the basis for docking 139,735 compounds. Effects of the top scoring compounds were investigated in a cell model that stably expresses Z-variant alpha 1-antitrypsin and in PiZ mice expressing Z-variant human alpha 1-antitrypsin (Z-hAAT), encoded by SERPINA1*E342K. 4′,′5-(Methylenedioxy)-2-nitrocinnamic acid was predicted to bind cleaved alpha 1-antitrypsin at the polymerization interface, and observed to co-localize with Z-hAAT, increase Z-hAAT degradation, inhibit intracellular accumulation of Z-hAAT, and alleviate liver fibrosis.
Collapse
|
46
|
|
47
|
Schuster R, Bar-Nathan O, Tiosano A, Lewis EC, Silberstein E. Enhanced Survival and Accelerated Perfusion of Skin Flap to Recipient Site Following Administration of Human α1-Antitrypsin in Murine Models. Adv Wound Care (New Rochelle) 2019; 8:281-290. [PMID: 31737418 DOI: 10.1089/wound.2018.0889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/23/2018] [Indexed: 12/18/2022] Open
Abstract
Objective: Skin flaps are routinely used in reconstructive surgery yet remain susceptible to ischemia and necrosis. Distant flaps require lengthy time to detach causing patient discomfort. Human α1-antitrypsin (hAAT) is a clinically available serum glycoprotein. hAAT was shown to support mature vessel formation and enhance tissue survival following ischemia-reperfusion injuries. The purpose of the presented study was to examine the effect of hAAT on skin flap survival and distant "tube" flap perfusion through its recipient site. Approach: Random-pattern skin flaps were performed on mice treated with clinical-grade hAAT using three unique routes of administration (transgenic, i.p. and s.c. infiltration); necrotic area and tissue perfusion were assessed. Blockade of vascular endothelial growth factor (VEGF) and nitric oxide synthase (NOS) were used to explore aspects of mechanism of action. A distant tube flap model was performed to examine time to perfusion. Results: hAAT-treated mice displayed approximately two-fold smaller necrotic flap areas versus controls across all hAAT administration routes. Flaps displayed greater perfusion as early as 3 days postsurgery (64.6% ± 4.0% vs. 43.7% ± 1.7% in controls; p = 0.007). hAAT-mediated flap survival was prominently NOS dependent, but only partially VEGF dependent. Finally, distant flaps treated with hAAT displayed significantly earlier perfusion versus controls (mean 9.6 ± 1.6 vs. 13.1 ± 1.0 days; p = 0.0005). Innovation: The established safety record of hAAT renders it an attractive candidate toward improving skin flap surgery outcomes, particularly during VEGF blockade. Conclusions: hAAT treatment enhances survival and accelerates perfusion of skin flaps in animal models in a NOS-dependent manner, partially circumventing VEGF blockade. Further mechanistic studies are required.
Collapse
Affiliation(s)
- Ronen Schuster
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Or Bar-Nathan
- Department of Plastic and Reconstructive Surgery, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Tiosano
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eldad Silberstein
- Department of Plastic and Reconstructive Surgery, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
48
|
Barragán-Méndez C, Sobrino I, Marín-Rincón A, Fernández-Boo S, Costas B, Mancera JM, Ruiz-Jarabo I. Acute-Stress Biomarkers in Three Octopodidae Species After Bottom Trawling. Front Physiol 2019; 10:784. [PMID: 31293450 PMCID: PMC6603232 DOI: 10.3389/fphys.2019.00784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/04/2019] [Indexed: 11/20/2022] Open
Abstract
Several Octopodidae species have a great potential for the diversification of worldwide aquaculture. Unfortunately, the lack of stress-related biomarkers in this taxon results an obstacle for its maintenance in conditions where animal welfare is of paramount relevance. In this study, we made a first approach to uncover physiological responses related to fishing capture in Eledone moschata, Eledone cirrhosa, and Octopus vulgaris. Captured octopus from all three species were individually maintained in an aquaculture system onboard of oceanographic vessel in south-western waters of Europe. Haemolymph plasma and muscle were collected in animals at the moment of capture, and recovery was evaluated along a time-course of 48 h in Eledone spp., and 24 h for O. vulgaris. Survival rates of these species captured in spring and autumn were evaluated. Physiological parameters such as plasma pH, total CO2, peroxidase activity, lysozyme, hemocyanin, proteases, pro-phenoloxidase, anti-proteases, free amino acids, lactate and glucose levels, as well as muscle water percentage, free amino acids, lactate, glycogen and glucose values were analyzed. The immune system appears to be compromised in these species due to capture processes, while energy metabolites were mobilized to face the acute-stress situation, but recovery of all described parameters occurs within the first 24 h after capture. Moreover, this situation exerts hydric balance changes, as observed in the muscle water, being these responses depending on the species assessed. In conclusion, three Octopodidae species from south-western waters of Europe have been evaluated for stress-related biomarkers resulting in differentiated mechanisms between species. This study may pave the way to further study the physiology of stress in adult octopuses and develop new methodologies for their growth in aquaculture conditions.
Collapse
Affiliation(s)
- Cristina Barragán-Méndez
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Ignacio Sobrino
- Instituto Español de Oceanografía, Centro Oceanográfico de Cádiz, Cádiz, Spain
| | - Adrián Marín-Rincón
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Sergio Fernández-Boo
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Matosinhos, Portugal
| | - Benjamin Costas
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Matosinhos, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Juan Miguel Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Ignacio Ruiz-Jarabo
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar, Universidad de Cádiz, Cádiz, Spain
| |
Collapse
|
49
|
Kaner Z, Engelman R, Schuster R, Rider P, Greenberg D, Av-Gay Y, Benhar M, Lewis EC. S-Nitrosylation of α1-Antitrypsin Triggers Macrophages Toward Inflammatory Phenotype and Enhances Intra-Cellular Bacteria Elimination. Front Immunol 2019; 10:590. [PMID: 31001247 PMCID: PMC6454134 DOI: 10.3389/fimmu.2019.00590] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/05/2019] [Indexed: 01/01/2023] Open
Abstract
Background: Human α1-antitrypsin (hAAT) is a circulating anti-inflammatory serine-protease inhibitor that rises during acute phase responses. in vivo, hAAT reduces bacterial load, without directly inhibiting bacterial growth. In conditions of excess nitric-oxide (NO), hAAT undergoes S-nitrosylation (S-NO-hAAT) and gains antibacterial capacity. The impact of S-NO-hAAT on immune cells has yet to be explored. Aim: Study the effects of S-NO-hAAT on immune cells during bacterial infection. Methods: Clinical-grade hAAT was S-nitrosylated and then compared to unmodified hAAT, functionally, and structurally. Intracellular bacterial clearance by THP-1 macrophages was assessed using live Salmonella typhi. Murine peritoneal macrophages were examined, and signaling pathways were evaluated. S-NO-hAAT was also investigated after blocking free mambranal cysteine residues on cells. Results: S-NO-hAAT (27.5 uM) enhances intracellular bacteria elimination by immunocytes (up to 1-log reduction). S-NO-hAAT causes resting macrophages to exhibit a pro-inflammatory and antibacterial phenotype, including release of inflammatory cytokines and induction of inducible nitric oxide synthase (iNOS) and TLR2. These pro-inflammatory effects are dependent upon cell surface thiols and activation of MAPK pathways. Conclusions: hAAT duality appears to be context-specific, involving S-nitrosylation in a nitric oxide rich environment. Our results suggest that S-nitrosylation facilitates the antibacterial activity of hAAT by promoting its ability to activate innate immune cells. This pro-inflammatory effect may involve transferring of nitric oxide from S-NO-hAAT to a free cysteine residue on cellular targets.
Collapse
Affiliation(s)
- Ziv Kaner
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rotem Engelman
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronen Schuster
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - David Greenberg
- The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Yossef Av-Gay
- Division of Infectious Diseases, Departments of Medicine and Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
50
|
Chen X, Yao J, Liu L, Zheng W, Hu X, Zhu Y, Wang H, Guo J. Serum Alpha1-Globulin as a Novel Prognostic Factor in Metastatic Renal Cell Carcinoma Treated with Tyrosine Kinase Inhibitors. Target Oncol 2019; 14:187-195. [PMID: 30887420 DOI: 10.1007/s11523-019-00625-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Serum protein fraction (SPF) is a common parameter reflecting the nutritional and inflammatory status of the human body. However, its role in patients with cancer, particularly those treated with targeted agents, is unknown. OBJECTIVE We conducted this study to explore the prognostic value of SPF in patients with metastatic renal cell carcinoma (mRCC) treated with tyrosine kinase inhibitors and its association with clinical characteristics. METHODS Patients with mRCC (n = 213) who initiated first-line sunitinib or sorafenib systemic therapy for metastatic disease between March 2007 and June 2017 at Zhongshan Hospital, Fudan University, were retrospectively included in our analysis. Clinical and pathological data were collected. SPF was measured by capillary electrophoresis. Prognostic factors of overall survival (OS) and progression-free survival (PFS) were analyzed using the Cox proportional hazards model. Correlation was estimated with Spearman's correlation coefficient. RESULTS Among all SPF components, high α1-globulin was an independent prognostic factor for OS and PFS (dichotomized by median, hazard ratio [HR] 2.356; 95% confidence interval [CI] 1.399-3.966, p = 0.001; and HR 1.994; 95% CI 1.360-2.923, p < 0.001, respectively). In our cohort, α1-globulin showed better predictive value for OS than the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) model (C-index 0.682 vs. 0.597; p = 0.005). Moreover, serum α1-globulin was positively correlated with International Society of Urological Pathology (ISUP) grade (r = 0.237; p < 0.001), tumor size (r = 0.242; p < 0.001), initial tumor/node/metastasis (TNM) stage (r = 0.185; p = 0.007), and IMDC risk group (r = 0.485; p < 0.001). CONCLUSIONS High serum α1-globulin correlates with high tumor load. Serum α1-globulin is an independent prognostic factor of OS and PFS in mRCC and demonstrates better predictive value for OS than does the IMDC model.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - JiaXi Yao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - WenZhong Zheng
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - XiaoYi Hu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - YanJun Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hang Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - JianMing Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|