1
|
Piñeiro-Llanes J, Suzuki-Hatano S, Jain A, Venigalla S, Kamat M, Basso KB, Cade WT, Simmons CS, Pacak CA. Rescue of mitochondrial dysfunction through alteration of extracellular matrix composition in barth syndrome cardiac fibroblasts. Biomaterials 2025; 315:122922. [PMID: 39509858 PMCID: PMC11625619 DOI: 10.1016/j.biomaterials.2024.122922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Fibroblast-ECM (dys)regulation is associated with a plethora of diseases. The ECM acts as a reservoir of inflammatory factors and cytokines that mediate molecular mechanisms within cardiac cell populations. The role of ECM-mitochondria crosstalk in the development and progression of cardiac disorders remains uncertain. We evaluated the influence of ECM produced by stromal cells from patients with the mitochondrial cardiomyopathy (Barth syndrome, BTHS) and unaffected healthy controls on cardiac fibroblast (CF) metabolic function. To do this, cell-derived matrices CDMs were generated from BTHS and healthy human pluripotent stem cell-derived CFs (hPSC-CF) and used as cell culture substrates. BTHS CDMs negatively impacted the mitochondrial function of healthy hPSC-CFs while healthy CDMs improved mitochondrial function in BTHS hPSC-CFs. Mass spectrometry comparisons identified 5 matrisome proteins differentially expressed in BTHS compared to healthy CDM. Our results highlight a key role for the ECM in disease through its impact on mitochondrial function.
Collapse
Affiliation(s)
- Janny Piñeiro-Llanes
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA.
| | - Silveli Suzuki-Hatano
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - Ananya Jain
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA.
| | - Sree Venigalla
- Department of Neurology and Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, 55455, USA.
| | - Manasi Kamat
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| | - Kari B Basso
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| | - William T Cade
- Doctor of Physical Therapy Division, Duke University, Durham, NC, 27710, USA.
| | - Chelsey S Simmons
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA; Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA.
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA; Department of Neurology and Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, 55455, USA.
| |
Collapse
|
2
|
Lee J, Sasaki F, Koike E, Cho M, Lee Y, Dho SH, Lee J, Lee E, Toyohara E, Sunakawa M, Ishibashi M, Hung HH, Nishioka S, Komine R, Okura C, Shimizu M, Ikawa M, Yoshimura A, Morita R, Kim LK. Gelsolin alleviates rheumatoid arthritis by negatively regulating NLRP3 inflammasome activation. Cell Death Differ 2024; 31:1679-1694. [PMID: 39179640 PMCID: PMC11618363 DOI: 10.1038/s41418-024-01367-6] [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: 04/30/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 08/26/2024] Open
Abstract
Despite numerous biomarkers being proposed for rheumatoid arthritis (RA), a gap remains in our understanding of their mechanisms of action. In this study, we discovered a novel role for gelsolin (GSN), an actin-binding protein whose levels are notably reduced in the plasma of RA patients. We elucidated that GSN is a key regulator of NLRP3 inflammasome activation in macrophages, providing a plausible explanation for the decreased secretion of GSN in RA patients. We found that GSN interacts with NLRP3 in LPS-primed macrophages, hence modulating the formation of the NLRP3 inflammasome complex. Reducing GSN expression significantly enhanced NLRP3 inflammasome activation. GSN impeded NLRP3 translocation to the mitochondria; it contributed to the maintenance of intracellular calcium equilibrium and mitochondrial stability. This maintenance is crucial for controlling the inflammatory response associated with RA. Furthermore, the exacerbation of arthritic symptoms in GSN-deficient mice indicates the potential of GSN as both a diagnostic biomarker and a therapeutic target. Moreover, not limited to RA models, GSN has demonstrated a protective function in diverse disease models associated with the NLRP3 inflammasome. Myeloid cell-specific GSN-knockout mice exhibited aggravated inflammatory responses in models of MSU-induced peritonitis, folic acid-induced acute tubular necrosis, and LPS-induced sepsis. These findings suggest novel therapeutic approaches that modulate GSN activity, offering promise for more effective management of RA and a broader spectrum of inflammatory conditions.
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Fumiyuki Sasaki
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Eri Koike
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Minjeong Cho
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeongun Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Hee Dho
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jina Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunji Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eri Toyohara
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Mika Sunakawa
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Mariko Ishibashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Huynh Hiep Hung
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Saki Nishioka
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ritsuko Komine
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Chiaki Okura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masumi Shimizu
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Masahito Ikawa
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan.
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
DiNubile MJ, Parra S, Salomó AC, Levinson SL. Adjunctive Recombinant Human Plasma Gelsolin for Severe Coronavirus Disease 2019 Pneumonia. Open Forum Infect Dis 2022; 9:ofac357. [PMID: 35928505 PMCID: PMC9345409 DOI: 10.1093/ofid/ofac357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/21/2022] [Indexed: 12/15/2022] Open
Abstract
Background Excessive inflammation contributes to the morbidity and mortality of severe coronavirus disease 2019 (COVID-19) pneumonia. Recombinant human plasma gelsolin (rhu-pGSN) improves disease outcomes in diverse experimental models of infectious and noninfectious inflammation. Methods In a blinded, randomized study, 61 subjects with documented COVID-19 pneumonia having a World Health Organization (WHO) Severity Score of 4 to 6 and evidence of a hyperinflammatory state were treated with standard care and either adjunctive rhu-pGSN 12 mg/kg or an equal volume of saline placebo given intravenously at entry, 12 hours, and 36 hours. The prespecified coprimary outcomes were survival without major respiratory, hemodynamic, or renal support on Day 14 and the incidence of serious adverse events (SAEs) during the 90-day study period. Results All subjects receiving ≥1 dose of study drug were analyzed. Fifty-four of 61 subjects (88.5%) were WHO severity level 4 at entry. The proportions of subjects alive without support on Day 14 were 25 of 30 rhu-pGSN recipients (83.3%) and 27 of 31 placebo recipients (87.1%). Over the duration of the study, WHO Severity Scores improved similarly in both treatment groups. No statistically significant differences were observed between treatment groups at any time point examined. Two subjects died in each group. Numerically fewer subjects in the rhu-pGSN group had SAEs (5 subjects; 16.7%) or ≥ Grade 3 adverse events (5 subjects; 16.7%) than in the placebo group (8 subjects [25.8%] and 9 subjects [29.0%], respectively), mostly involving the lungs. Three rhu-pGSN recipients (10.0%) were intubated compared to 6 placebo recipients (19.4%). Conclusions Overall, subjects in this study did well irrespective of treatment arm. When added to dexamethasone and remdesivir, no definitive benefit was demonstrated for rhu-pGSN relative to placebo. Safety signals were not identified after the administration of 3 doses of 12 mg/kg rhu-pGSN over 36 hours. The frequencies of SAEs and intubation were numerically fewer in the rhu-pGSN group compared with placebo.
Collapse
Affiliation(s)
| | - Sandra Parra
- Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili , Reus , Spain
| | - Antoni Castro Salomó
- Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili , Reus , Spain
| | | |
Collapse
|
4
|
Maeda K, Nakamura M, Yamamura T, Sawada T, Ishikawa E, Oishi A, Ikegami S, Kakushima N, Furukawa K, Iida T, Mizutani Y, Ishikawa T, Ohno E, Honda T, Ishigami M, Kawashima H. Gelsolin as a Potential Biomarker for Endoscopic Activity and Mucosal Healing in Ulcerative Colitis. Biomedicines 2022; 10:872. [PMID: 35453622 PMCID: PMC9029534 DOI: 10.3390/biomedicines10040872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
The therapeutic goal in ulcerative colitis is mucosal healing, which requires improved non-invasive biomarkers to evaluate disease activity. Gelsolin is associated with several autoimmune diseases, and here, we aimed to analyze its usefulness as a serological biomarker for clinical and endoscopic activities in ulcerative colitis. Patients with ulcerative colitis (n = 138) who had undergone blood tests and colonoscopy were included. Serum gelsolin was measured using enzyme-linked immunosorbent assay, and correlation between the gelsolin level and clinical and endoscopic activities was examined. The serum gelsolin level in patients with ulcerative colitis was significantly lower than that in healthy subjects, and it decreased in proportion to increasing Mayo score and Mayo endoscopic subscore. The area under the curve for correlation between clinical and endoscopic remission and serum gelsolin level was higher than that for C-reactive protein. Furthermore, in C-reactive protein-negative patients, the serum gelsolin level was lower in the active phase than in remission. Our findings indicate that the serum gelsolin level correlates with clinical and endoscopic activities in ulcerative colitis, has a higher sensitivity and specificity than C-reactive protein, and can detect mucosal healing, suggesting that gelsolin can be used as a biomarker for ulcerative colitis.
Collapse
Affiliation(s)
- Keiko Maeda
- Department of Endoscopy, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan;
| | - Masanao Nakamura
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Takeshi Yamamura
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Tsunaki Sawada
- Department of Endoscopy, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan;
| | - Eri Ishikawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Akina Oishi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Shuji Ikegami
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Naomi Kakushima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Kazuhiro Furukawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Tadashi Iida
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Yasuyuki Mizutani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Eizaburo Ohno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.); (T.Y.); (E.I.); (A.O.); (S.I.); (N.K.); (K.F.); (T.I.); (Y.M.); (T.I.); (E.O.); (T.H.); (M.I.); (H.K.)
| |
Collapse
|
5
|
Troyer B, Rodgers J, Wolf BJ, Oates JC, Drake RR, Nowling TK. Glycosphingolipid Levels in Urine Extracellular Vesicles Enhance Prediction of Therapeutic Response in Lupus Nephritis. Metabolites 2022; 12:134. [PMID: 35208209 PMCID: PMC8876142 DOI: 10.3390/metabo12020134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/28/2022] Open
Abstract
The development of nephritis increases the risk of morbidity and mortality in systemic lupus erythematosus (SLE) patients. While standard induction therapies, such as mycophenolate mofetil (MMF) induce clinical remission (i.e., complete response) in approximately 50% of SLE patients with nephritis, many patients fail to respond. Therapeutic response is often not assessed until 6-12 months after beginning treatment. Those patients that fail to respond to treatment continue to accumulate organ damage, thus, there is a critical need to predict which patients will fail therapy before beginning treatment, allowing physicians to optimize therapy. Our previous studies demonstrated elevated urine, but not serum, glycosphingolipids (GSLs) in SLE patients with nephritis compared to SLE patients without nephritis, suggesting the urine GSLs were derived from the kidney. In this study, we measured the GSLs hexosylceramide and lactosylceramide in extracellular vesicles isolated from longitudinal urine samples of LN patients that were treated with MMF for 12 months. GSL levels were significantly elevated in the baseline samples (prior to treatment) of non-responders compared to complete responders. While a few other proteins measured in the whole urine were higher in non-responders at baseline, only GSLs demonstrated a significant ability to discriminate treatment response in lupus nephritis patients.
Collapse
Affiliation(s)
- Brian Troyer
- Department of Medicine, Rheumatology Division, Medical University of South Carolina, Charleston, SC 29425, USA; (B.T.); (J.R.); (J.C.O.)
| | - Jessalyn Rodgers
- Department of Medicine, Rheumatology Division, Medical University of South Carolina, Charleston, SC 29425, USA; (B.T.); (J.R.); (J.C.O.)
| | - Bethany J. Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - James C. Oates
- Department of Medicine, Rheumatology Division, Medical University of South Carolina, Charleston, SC 29425, USA; (B.T.); (J.R.); (J.C.O.)
- Rheumatology Section, Ralph H. Johnson VA Medical Center, Charleston, SC 29403, USA
| | - Richard R. Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Tamara K. Nowling
- Department of Medicine, Rheumatology Division, Medical University of South Carolina, Charleston, SC 29425, USA; (B.T.); (J.R.); (J.C.O.)
| |
Collapse
|
6
|
Mosaad GM, Abdel moneam SM, Soliman AF, Ameen SG, Amer AS. Implication of plasma gelsolin in systemic lupus erythematosus patients. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2022. [DOI: 10.1186/s43166-021-00103-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder with more than one organ involvement. Kidney is the foremost commonly affected one. Gelsolin is a protein that induces depolymerization of actin filaments thus preventing downstream stimulation of inflammatory reactions. The aim of this work was to detect the relation of plasma gelsolin to SLE disease activity and severity indices in order to find out if plasma gelsolin could be used as a biomarker of the disease. This study was conducted on 50 SLE female patients and 30 matched control. SLE disease activity Index (SLEDAI) and SLE damage index (SDI) were assessed. All lupus nephritis (LN) patients were subjected to an ultrasound-guided kidney biopsy. Plasma gelsolin level was measured.
Results
The mean age of the patients was 38.5 ± 6.3 years (26–51 years) with median disease duration of 5 (3–9.3) years. Eighteen patients had LN, 11 had cardiac manifestations and 12 had chest manifestations. The mean SLEDAI was 13.1 ± 4.5 (4–22) and the median SDI was 2 (1–3). Plasma gelsolin level was significantly lower in SLE patients (74.9 mg/l; 57.5–98.8 mg/l) compared to control (801.5 mg/l; 225–1008.3 mg/l) (p < 0.001). There were significant negative correlations of gelsolin levels with anti-ds DNA (r = − 0.63, p < 0.001), SLEDAI (r = − 0.79, p < 0.001), and SDI (r = − 0.74, p = 0.001). Plasma gelsolin level was significantly lower in SLE patients with high/very high activity grades compared to those with low and moderate (p = 0.007 and p < 0.001 respectively). A gelsolin level of ≤ 78.95 mg/l significantly predicted renal affection (p < 0.001), with a sensitivity of 100%, specificity 71.9%, and a positive predictive value 66.7%.
Conclusion
A decreased gelsolin level is associated with disease activity in SLE patients. Plasma gelsolin was well related to disease activity and severity with a high predictive value for renal affection comparable to anti-ds DNA titre. Plasma gelsolin is a potentially important predictive biomarker for SLE and LN.
Collapse
|
7
|
Safety and Pharmacokinetics of Recombinant Human Plasma Gelsolin in Patients Hospitalized for Nonsevere Community-Acquired Pneumonia. Antimicrob Agents Chemother 2020; 64:AAC.00579-20. [PMID: 32690640 DOI: 10.1128/aac.00579-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/11/2020] [Indexed: 12/30/2022] Open
Abstract
There remains an unmet need to address the substantial morbidity and mortality associated with severe community-acquired pneumonia (sCAP). Recombinant human plasma gelsolin (rhu-pGSN) improves disease outcomes in diverse animal models of infectious and noninfectious inflammation. This blinded dose-escalation safety study involved non-intensive care unit (ICU) patients admitted for mild CAP and randomized 3:1 to receive adjunctive rhu-pGSN or placebo intravenously. Thirty-three subjects were treated: 8 in the single-dose phase and 25 in the multidose phase. For the single-dose phase, rhu-pGSN at 6 mg/kg of body weight was administered once. For the multidose phase, a daily rhu-pGSN dose of 6, 12, or 24 mg/kg was given on 3 consecutive days. Adverse events (AEs) were generally mild in both treatment groups irrespective of dose. The only serious AE (SAE) in the single-dose phase was a non-drug-related pneumonia in a rhu-pGSN recipient who died after institution of comfort care. One single-dose placebo recipient had a drug-related AE (maculo-papular rash). In the multidose phase, there were 2 SAEs in 1 placebo recipient, including a fatal pulmonary embolism. In the 18 rhu-pGSN recipients in the multidose phase, there were no serious or drug-related AEs, and nausea and increased blood pressure were each reported in 2 patients. The median rhu-pGSN half-life exceeded 17 h with all dosing regimens, and supraphysiologic levels were maintained throughout the 24-h dosing interval in the 2 highest dosing arms. Rhu-pGSN was well tolerated overall in CAP patients admitted to non-ICU beds, justifying a larger proof-of-concept trial in an ICU population admitted with sCAP. (This study has been registered at ClinicalTrials.gov under identifier NCT03466073.).
Collapse
|
8
|
DiNubile MJ, Levinson SL, Stossel TP, Lawrenz MB, Warawa JM. Recombinant Human Plasma Gelsolin Improves Survival and Attenuates Lung Injury in a Murine Model of Multidrug-Resistant Pseudomonas aeruginosa Pneumonia. Open Forum Infect Dis 2020; 7:ofaa236. [PMID: 32766380 PMCID: PMC7397834 DOI: 10.1093/ofid/ofaa236] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Background Plasma gelsolin (pGSN) is an abundant circulating protein quickly consumed by extensive tissue damage. Marked depletion is associated with later poor outcomes in diverse clinical circumstances. Repletion with recombinant human (rhu)-pGSN in animal models of inflammation lessens mortality and morbidity. Methods Neutropenic mice were treated with different meropenem doses ±12 mg of rhu-pGSN commencing 1 day before an intratracheal challenge with multidrug-resistant Pseudomonas aeruginosa. Survival, bacterial counts, and pulmonary pathology were compared between corresponding meropenem groups with and without rhu-pGSN. Results Overall survival was 35/64 (55%) and 46/64 (72%) in mice given meropenem without and with rhu-pGSN, respectively (Δ = 17%; 95% CI, 1-34). In control mice receiving meropenem 1250 mg/kg/d where the majority died, the addition of rhu-pGSN increased survival from 5/16 (31%) to 12/16 (75%) (Δ = 44%; 95% CI, 13-75). Survival with minor lung injury was found in 26/64 (41%) mice receiving only meropenem, vs 38/64 (59%) in mice given meropenem plus rhu-pGSN (Δ = 19%; 95% CI, 2-36). Conclusions In a series of dose-ranging experiments, both mortality and lung injury were reduced by the addition of rhu-pGSN to meropenem against carbapenem-resistant P. aeruginosa. Rhu-pGSN offers a novel candidate therapy for antibiotic-resistant pneumonia.
Collapse
Affiliation(s)
| | | | | | - Matthew B Lawrenz
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Jonathan M Warawa
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
9
|
Piktel E, Wnorowska U, Cieśluk M, Deptuła P, Prasad SV, Król G, Durnaś B, Namiot A, Markiewicz KH, Niemirowicz-Laskowska K, Wilczewska AZ, Janmey PA, Reszeć J, Bucki R. Recombinant Human Plasma Gelsolin Stimulates Phagocytosis while Diminishing Excessive Inflammatory Responses in Mice with Pseudomonas aeruginosa Sepsis. Int J Mol Sci 2020; 21:ijms21072551. [PMID: 32272559 PMCID: PMC7177774 DOI: 10.3390/ijms21072551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/29/2020] [Accepted: 04/03/2020] [Indexed: 12/22/2022] Open
Abstract
Plasma gelsolin (pGSN) is a highly conserved abundant circulating protein, characterized by diverse immunomodulatory activities including macrophage activation and the ability to neutralize pro-inflammatory molecules produced by the host and pathogen. Using a murine model of Gram-negative sepsis initiated by the peritoneal instillation of Pseudomonas aeruginosa Xen 5, we observed a decrease in the tissue uptake of IRDye®800CW 2-deoxyglucose, an indicator of inflammation, and a decrease in bacterial growth from ascitic fluid in mice treated with intravenous recombinant human plasma gelsolin (pGSN) compared to the control vehicle. Pretreatment of the murine macrophage line RAW264.7 with pGSN, followed by addition of Pseudomonas aeruginosa Xen 5, resulted in a dose-dependent increase in the proportion of macrophages with internalized bacteria. This increased uptake was less pronounced when cells were pretreated with pGSN and then centrifuged to remove unbound pGSN before addition of bacteria to macrophages. These observations suggest that recombinant plasma gelsolin can modulate the inflammatory response while at the same time augmenting host antibacterial activity.
Collapse
Affiliation(s)
- Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
| | - Mateusz Cieśluk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
| | - Piotr Deptuła
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
| | - Suhanya V. Prasad
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
| | - Grzegorz Król
- Department of Microbiology and Immunology, the Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Stefana Żeromskiego 5, 25-001 Kielce, Poland; (G.K.); (B.D.)
| | - Bonita Durnaś
- Department of Microbiology and Immunology, the Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Stefana Żeromskiego 5, 25-001 Kielce, Poland; (G.K.); (B.D.)
| | - Andrzej Namiot
- Department of Anatomy, Medical University of Bialystok, Mickiewicza 2b, 15-222 Bialystok, Poland;
| | - Karolina H. Markiewicz
- Institute of Chemistry, University of Białystok, Ciołkowskiego 1K, 15-245 Bialystok, Poland; (K.H.M.); (A.Z.W.)
| | - Katarzyna Niemirowicz-Laskowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
| | - Agnieszka Z. Wilczewska
- Institute of Chemistry, University of Białystok, Ciołkowskiego 1K, 15-245 Bialystok, Poland; (K.H.M.); (A.Z.W.)
| | - Paul A. Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA 19104, USA;
| | - Joanna Reszeć
- Department of Pathology, Medical University of Bialystok, Waszyngtona 13, 15-269 Bialystok, Poland;
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland; (E.P.); (U.W.); (M.C.); (P.D.); (S.V.P.); (K.N.-L.)
- Department of Microbiology and Immunology, the Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Stefana Żeromskiego 5, 25-001 Kielce, Poland; (G.K.); (B.D.)
- Correspondence: ; Tel.: +48-748-54-93
| |
Collapse
|
10
|
Parra S, Heras M, Herrero P, Amigó N, Garcés E, Girona J, Correig X, Canela N, Castro A. Gelsolin: a new biomarker of disease activity in SLE patients associated with HDL-c. Rheumatology (Oxford) 2020; 59:650-661. [PMID: 31504936 DOI: 10.1093/rheumatology/kez293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/28/2019] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVES To identify potential biomarkers of disease activity analysing the proteome of high-density lipoprotein (HDL) particles from SLE patients in clinical remission and when they develop a flare compared with a healthy control group. METHODS Quantitative proteomic analyses of purified HDL were performed using Tandem Mass Tag isobaric tag-labelling and nanoLC-Orbitrap (nLC-MS/MS) from nine SLE patients in clinical remission when they developed a flare and from nine healthy controls (9-9-9). We verified the identified proteins by Western blot and ELISA in a cohort of 104 SLE women patients, 46 healthy women and 14 SLE patients when a flare developed. RESULTS We found 17 proteins with a significant fold-change (>1.1) compared with the control group. In lupus patients experiencing a flare compared with those in remission, we identified four proteins with a significant fold-change (C4, Indian Hedgehog protein, S100A8 and gelsolin). Plasma gelsolin (pGSN) levels were decreased in the 104 SLE patients (176.02(74.9) mcg/l) compared with the control group (217.13(86.7) mcg/l); P=0.005 and when they developed a clinical flare (104.84(41.7) mcg/l); P=0.002). pGSN levels were associated with HDL cholesterol levels (r = 0.316, P<0.001). Antimalarial treated patients showed significant higher levels of pGSN (214.56(88.94) mcg/l regarding 170.35(66.36) mcg/l); P = 0.017. CONCLUSION Decreased pGSN are associated with clinical disease activity in SLE patients. Antimalarial treatment and HDL cholesterol are associated with higher levels of pGSN.
Collapse
Affiliation(s)
- Sandra Parra
- Autoimmune Diseases Unit, Internal Medicine Department, 'Sant Joan' University Hospital (Reus, Spain), Spain.,Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Reus, Spain
| | - Mercedes Heras
- Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Reus, Spain.,Unitat de Recerca de Lipids i Arteriosclerosis (URLA), 'Sant Joan' University Hospital (Reus-Spain), Spain
| | - Pol Herrero
- Centre for Omic Science, Joint Unit Universitat Rovira i Virgili-Eurecat, Spain
| | - Nuria Amigó
- Department of Electronic Engineering, Universitat Rovira i Virgili (URV), Reus, Spain, Rovira i Virgili University, IISPV, Spain
| | - Esperanza Garcés
- Autoimmune Diseases Unit, Internal Medicine Department, 'Sant Joan' University Hospital (Reus, Spain), Spain
| | - Josefa Girona
- Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Reus, Spain.,Unitat de Recerca de Lipids i Arteriosclerosis (URLA), 'Sant Joan' University Hospital (Reus-Spain), Spain
| | - Xavier Correig
- Department of Electronic Engineering, Universitat Rovira i Virgili (URV), Reus, Spain, Rovira i Virgili University, IISPV, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Nuria Canela
- Centre for Omic Science, Joint Unit Universitat Rovira i Virgili-Eurecat, Spain
| | - Antoni Castro
- Autoimmune Diseases Unit, Internal Medicine Department, 'Sant Joan' University Hospital (Reus, Spain), Spain.,Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Reus, Spain
| |
Collapse
|
11
|
Plasma gelsolin levels in patients with psoriatic arthritis: a possible novel marker. Clin Rheumatol 2020; 39:1881-1888. [PMID: 32002760 DOI: 10.1007/s10067-020-04959-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 01/06/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Psoriatic arthritis (PsA) is an inflammatory disorder affecting the joints of psoriatic patients. Gelsolin regulated the actin assembly and disassembly. Reduction in plasma gelsolin levels was detected in tissue damages, including trauma, sepsis, and chronic inflammatory disorders. OBJECTIVES The study aims to investigate the potential role of gelsolin in PsA and to determine the association between gelsolin and the disease activity. METHODS Plasma gelsolin levels were measured in 76 PsA patients in comparison with 40 patients having psoriasis only and 40 age- and sex -matched healthy controls. RESULTS Plasma gelsolin levels were decreased in PsA patients compared to controls and psoriasis-only patients (p ˂ 0.0001). The optimal cutoff point of gelsolin was 172.5 mg/L. Gelsolin showed 92.1% sensitivity and 95% specificity in detecting PsA. But, it had 92.1% sensitivity and 80% specificity in differentiating between psoriasis and PsA. Plasma gelsolin showed a significant negative correlation with inflammatory markers as C-reactive protein and erythrocyte sedimentation rate (p < 0.0001 and p = 0.039; respectively). A significant negative correlation between plasma gelsolin and PsA activity was detected (p < 0.0001). The PsA activity was defined by the Disease Activity for Psoriatic Arthritis Score and the Composite Psoriatic Disease Activity Index. CONCLUSIONS The plasma gelsolin levels were decreased in PsA patients, suggesting that gelsolin may be implicated in the chronic joint inflammation process. Plasma gelsolin seems to be a useful predictive biomarker for diagnosing PsA and monitoring the disease activity.Key Points• This study introduces an unprecedented focus within which the relationship between the levels of plasma gelsolin and PsA is investigated• The study examines the potential role of gelsolin in PsA, and detects the association between gelsolin and the arthritis activity.• There were decreased plasma gelsolin levels in PsA patients. So, gelsolin can constitute a role in the chronic joint inflammation process.• Gelsolin may be a useful biomarker for diagnosing of PsA and monitoring the disease activity.
Collapse
|
12
|
Dinsdale RJ, Hazeldine J, Al Tarrah K, Hampson P, Devi A, Ermogenous C, Bamford AL, Bishop J, Watts S, Kirkman E, Dalle Lucca JJ, Midwinter M, Woolley T, Foster M, Lord JM, Moiemen N, Harrison P. Dysregulation of the actin scavenging system and inhibition of DNase activity following severe thermal injury. Br J Surg 2019; 107:391-401. [PMID: 31502663 PMCID: PMC7079039 DOI: 10.1002/bjs.11310] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/21/2019] [Accepted: 06/08/2019] [Indexed: 01/25/2023]
Abstract
Background Circulating cell‐free DNA (cfDNA) is not found in healthy subjects, but is readily detected after thermal injury and may contribute to the risk of multiple organ failure. The hypothesis was that a postburn reduction in DNase protein/enzyme activity could contribute to the increase in cfDNA following thermal injury. Methods Patients with severe burns covering at least 15 per cent of total body surface area were recruited to a prospective cohort study within 24 h of injury. Blood samples were collected from the day of injury for 12 months. Results Analysis of blood samples from 64 patients revealed a significant reduction in DNase activity on days 1–28 after injury, compared with healthy controls. DNase protein levels were not affected, suggesting the presence of an enzyme inhibitor. Further analysis revealed that actin (an inhibitor of DNase) was present in serum samples from patients but not those from controls, and concentrations of the actin scavenging proteins gelsolin and vitamin D‐binding protein were significantly reduced after burn injury. In a pilot study of ten military patients with polytrauma, administration of blood products resulted in an increase in DNase activity and gelsolin levels. Conclusion The results of this study suggest a novel biological mechanism for the accumulation of cfDNA following thermal injury by which high levels of actin released by damaged tissue cause a reduction in DNase activity. Restoration of the actin scavenging system could therefore restore DNase activity, and reduce the risk of cfDNA‐induced host tissue damage and thrombosis.
Collapse
Affiliation(s)
- R J Dinsdale
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - J Hazeldine
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - K Al Tarrah
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - P Hampson
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - A Devi
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - C Ermogenous
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - A L Bamford
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - J Bishop
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - S Watts
- Chemical, Biological and Radiological (CBR) Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, UK
| | - E Kirkman
- Chemical, Biological and Radiological (CBR) Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, UK
| | - J J Dalle Lucca
- Translational Medical Division, Department of Chemical and Biological Technologies, Defense Threat Reduction Agency, Fort Belvoir, Virginia, USA
| | - M Midwinter
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - T Woolley
- ICT Centre, Birmingham Research Park, Birmingham, UK.,Chemical, Biological and Radiological (CBR) Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, UK
| | - M Foster
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - J M Lord
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - N Moiemen
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - P Harrison
- Scar Free Foundation, Birmingham Centre for Burns Research, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
13
|
Kopecki Z, Yang G, Treloar S, Mashtoub S, Howarth GS, Cummins AG, Cowin AJ. Flightless I exacerbation of inflammatory responses contributes to increased colonic damage in a mouse model of dextran sulphate sodium-induced ulcerative colitis. Sci Rep 2019; 9:12792. [PMID: 31488864 PMCID: PMC6728368 DOI: 10.1038/s41598-019-49129-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by cytokine driven inflammation that disrupts the mucosa and impedes intestinal structure and functions. Flightless I (Flii) is an immuno-modulatory protein is a member of the gelsolin family of actin-remodelling proteins that regulates cellular and inflammatory processes critical in tissue repair. Here we investigated its involvement in UC and show that Flii is significantly elevated in colonic tissues of patients with inflammatory bowel disease. Using an acute murine model of colitis, we characterised the contribution of Flii to UC using mice with low (Flii+/-), normal (Flii+/+) and high Flii (FliiTg/Tg). High levels of Flii resulted in significantly elevated disease severity index scores, increased rectal bleeding and degree of colon shortening whereas, low Flii expression decreased disease severity, reduced tissue inflammation and improved clinical indicators of UC. Mice with high levels of Flii had significantly increased histological disease severity and elevated mucosal damage with significantly increased inflammatory cell infiltrate and significantly higher levels of TNF-α, IFN-γ, IL-5 and IL-13 pro-inflammatory cytokines. Additionally, Flii overexpression resulted in decreased β-catenin levels, inhibited Wnt/β-catenin signalling and impaired regeneration of colonic crypts. These studies suggest that high levels of Flii, as is observed in patients with UC, may adversely affect mucosal healing via mechanisms involving Th1 and Th2 mediated tissue inflammation and Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Z Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia.
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.
| | - G Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
| | - S Treloar
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Adelaide, South Australia, Australia
| | - S Mashtoub
- Department of Gastroenterology, Women's and Children's Hospital, North Adelaide, South Australia, Australia
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - G S Howarth
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - A G Cummins
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville South, Adelaide, South Australia, Australia
| | - A J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Development of a Novel Diagnostic Biomarker Set for Rheumatoid Arthritis Using a Proteomics Approach. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7490723. [PMID: 30662913 PMCID: PMC6312602 DOI: 10.1155/2018/7490723] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/28/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
Abstract
Background Rheumatoid arthritis (RA) is an autoimmune disease that starts with inflammation of the synovial membrane. Studies have been conducted to develop methods for efficient diagnosis of RA and to identify the mechanisms underlying RA development. Blood samples can be useful for detecting disturbance of homeostasis in patients with RA. Nanoliquid chromatography-tandem mass spectrometry (LC-MS/MS) is an efficient proteomics approach to analyze blood sample and quantify serum proteins. Methods Serum samples of 18 healthy controls and 18 patients with RA were analyzed by LC-MS/MS. Selected candidate biomarkers were validated by enzyme-linked immunosorbent assay (ELISA) using sera from 43 healthy controls and 44 patients with RA. Results Thirty-eight proteins were significantly differentially expressed by more than 2-fold in healthy controls and patients with RA. Based on a literature survey, we selected six candidate RA biomarkers. ELISA was used to evaluate whether these proteins effectively allow distinguishing patients with RA from healthy controls and monitoring drug efficacy. SAA4, gelsolin, and vitamin D-binding protein were validated as potential biomarkers of RA for screening and drug efficacy monitoring of RA. Conclusions We identified a panel of three biomarkers for RA which has potential for application in RA diagnosis and drug efficacy monitoring. Further, our findings will aid in understanding the pathogenesis of RA.
Collapse
|
15
|
Plasma Gelsolin: Indicator of Inflammation and Its Potential as a Diagnostic Tool and Therapeutic Target. Int J Mol Sci 2018; 19:ijms19092516. [PMID: 30149613 PMCID: PMC6164782 DOI: 10.3390/ijms19092516] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/14/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022] Open
Abstract
Gelsolin, an actin-depolymerizing protein expressed both in extracellular fluids and in the cytoplasm of a majority of human cells, has been recently implicated in a variety of both physiological and pathological processes. Its extracellular isoform, called plasma gelsolin (pGSN), is present in blood, cerebrospinal fluid, milk, urine, and other extracellular fluids. This isoform has been recognized as a potential biomarker of inflammatory-associated medical conditions, allowing for the prediction of illness severity, recovery, efficacy of treatment, and clinical outcome. A compelling number of animal studies also demonstrate a broad spectrum of beneficial effects mediated by gelsolin, suggesting therapeutic utility for extracellular recombinant gelsolin. In the review, we summarize the current data related to the potential of pGSN as an inflammatory predictor and therapeutic target, discuss gelsolin-mediated mechanisms of action, and highlight recent progress in the clinical use of pGSN.
Collapse
|
16
|
Horváth-Szalai Z, Kustán P, Szirmay B, Lakatos Á, Christensen PH, Huber T, Bugyi B, Mühl D, Ludány A, Miseta A, Kovács GL, Kőszegi T. Validation of an automated immune turbidimetric assay for serum gelsolin and its possible clinical utility in sepsis. J Clin Lab Anal 2017; 32. [PMID: 28872708 DOI: 10.1002/jcla.22321] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/10/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Studies showing the potential predictive value of the actin-binding protein gelsolin, in critically ill patients are scarce. Moreover, even up to now a rapid automated measurement of gelsolin has still remained a challenge. Therefore, we developed and validated an automated serum gelsolin immune turbidimetric assay for possible clinical use. METHODS Validation of serum gelsolin assay was performed on a Cobas 8000/c502 analyzer (Roche) according to the second edition of Eurachem guidelines. Furthermore, we also studied the diagnostic value of serum gelsolin in sepsis when investigating sera of septic (n = 25), systemic inflammatory response syndrome (SIRS; n = 8) and control patients (n = 14). We compared our previously published Western blot data with those of the new turbidimetric assay. RESULTS The sample volume was 7 μL and the assay time was 10 minutes. The detection limit was 0.72 mg/L, intra- and inter-assay imprecision remained in most cases less than 5% expressed as CV. Recovery was found to be 84.56%-93.52% and linearity study gave an appropriate correlation coefficient by linear regression analysis (r2 = .998). Septic patients exhibited lower (P = .015) first-day serum gelsolin levels than SIRS patients, which confirmed our previous Western blot results. The determined cut-off point for serum gelsolin was 14.05 mg/L (sensitivity: 75%; specificity: 60%) when investigating its diagnostic value in sepsis. CONCLUSION Based on the results, our immune turbidimetric measurement offers a rapid and accurate quantitation of gelsolin in human serum samples. Serum gelsolin seems a promising additional diagnostic marker of sepsis which has to be further investigated.
Collapse
Affiliation(s)
| | - Péter Kustán
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary.,Department of Anaesthesiology and Intensive Therapy, University of Pécs, Pécs, Hungary
| | - Balázs Szirmay
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary
| | - Ágnes Lakatos
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary
| | | | - Tamás Huber
- Department of Biophysics, University of Pécs, Pécs, Hungary
| | - Beáta Bugyi
- Department of Biophysics, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Diána Mühl
- Department of Anaesthesiology and Intensive Therapy, University of Pécs, Pécs, Hungary
| | - Andrea Ludány
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary
| | - Gábor L Kovács
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Tamás Kőszegi
- Department of Laboratory Medicine, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| |
Collapse
|
17
|
Wongtrakul J, Thongtan T, Roytrakul S, Praparattanapan J, Wipasa J, Kumrapich B, Supparatpinyo K. Identification of novel biomarkers for adult-onset-immunodeficiency (AOID) syndrome using serum proteomics. ASIAN PAC J TROP MED 2017. [PMID: 28647184 DOI: 10.1016/j.apjtm.2017.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE To identify the candidate protein biomarkers of adult-onset-immunodeficiency (AOID) syndrome using serum proteomics. METHODS Screening and verification phases were performed in the study. A total of 97 serum samples were classified into three groups: AOID patients with opportunistic infections (active AOID), AOID patients without opportunistic infections (inactive AOID), and healthy control. In the screening phase, pooled sera collected from patients and healthy control in each group were separated by 2D-gel electrophoresis, analyzed for differentially expressed proteins and identified for biomarkers using LC/MS. In the verification phase, the protein candidates were selected for confirmation by western blotting. RESULTS The analysis revealed 35 differentially expressed proteins. Three proteins including haptoglobin, gelsolin, and transthyretin, were selected for verification. The results showed that the levels of haptoglobin in both active and inactive AOID groups were significantly higher than that in the control group, while the levels of gelsolin in the active AOID group were significantly lower than that in the inactive AOID group. The level of transthyretin in the active AOID group was also significantly lower than that in the control group. CONCLUSIONS The comparison of serum proteins between the three groups revealed three candidates which are related to chronic inflammatory diseases. Haptoglobin and transthyretin biomarkers could be applied in clinical assessment for monitor of disease outcome, including for the study of AOID pathogenesis.
Collapse
Affiliation(s)
- Jeerang Wongtrakul
- Research Institute for Health Sciences, Chiang Mai University, 110 Intavaroros Road, Sriphum, Muang District, Chiang Mai 50200, Thailand.
| | - Thananya Thongtan
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok 10330, Thailand
| | - Sittiruk Roytrakul
- Proteomics Research Laboratory, Genome Institute, National Center for Genetic Engineering and Biotechnology, 113 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathumthani 12120, Thailand
| | - Jutarat Praparattanapan
- Department of Medicine, Faculty of Medicine, Chiang Mai University, 110 Intavaroros Road, Sriphum, Muang District, Chiang Mai 50200, Thailand
| | - Jiraprapa Wipasa
- Research Institute for Health Sciences, Chiang Mai University, 110 Intavaroros Road, Sriphum, Muang District, Chiang Mai 50200, Thailand
| | - Benjawan Kumrapich
- Research Institute for Health Sciences, Chiang Mai University, 110 Intavaroros Road, Sriphum, Muang District, Chiang Mai 50200, Thailand
| | - Khuanchai Supparatpinyo
- Department of Medicine, Faculty of Medicine, Chiang Mai University, 110 Intavaroros Road, Sriphum, Muang District, Chiang Mai 50200, Thailand
| |
Collapse
|
18
|
Zhang L, Han C, Ye F, He Y, Jin Y, Wang T, Wu Y, Jiang Y, Zhang F, Jin X. Plasma Gelsolin Induced Glomerular Fibrosis via the TGF-β1/Smads Signal Transduction Pathway in IgA Nephropathy. Int J Mol Sci 2017; 18:ijms18020390. [PMID: 28208683 PMCID: PMC5343925 DOI: 10.3390/ijms18020390] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 01/29/2017] [Accepted: 02/08/2017] [Indexed: 11/16/2022] Open
Abstract
Glomerular fibrosis has been shown to be closely related to the progression and prognosis of IgA nephropathy (IgAN). However, mechanism underlying IgAN glomerular fibrosis remains unclear. Recently, our study showed that plasma gelsolin (pGSN) was decreased in the serum of an IgAN mouse model and that pGSN deposition was found in the glomeruli. Another cytokine, TGF-β1, which is closely related to glomerular fibrosis, was also found to be highly expressed in the glomeruli. In the present study, we report that pGSN induces glomerular fibrosis through the TGF-β1/Smads signal transduction pathway. This is supported by the following findings: human mesangial cells (HMCs) show remarkable morphological changes and proliferation in response to co-stimulation with pGSN and polymeric IgA1 (pIgA1) from IgAN patients compared to other controls. Moreover, ELISA assays showed that more TGF-β1 secretion was found in HMCs supernatants in the co-stimulation group. Further experiments showed increased TGF-β1, Smad3, p-Smad2/3, Smad4, and collagen 1 and decreased Smad7 expression in the co-stimulation group. Our present study implied that the synergistic effect of pGSN and pIgA induced glomerular fibrosis via the TGF-β1/Smads signal transduction pathway. This might be a potential mechanism for the glomerular fibrosis observed in IgAN patients.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Changsong Han
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Fei Ye
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yan He
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yinji Jin
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Tianzhen Wang
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yiqi Wu
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yang Jiang
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China.
| | - Xiaoming Jin
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
19
|
Kopecki Z, Ludwig RJ, Cowin AJ. Cytoskeletal Regulation of Inflammation and Its Impact on Skin Blistering Disease Epidermolysis Bullosa Acquisita. Int J Mol Sci 2016; 17:ijms17071116. [PMID: 27420054 PMCID: PMC4964491 DOI: 10.3390/ijms17071116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 01/10/2023] Open
Abstract
Actin remodelling proteins regulate cytoskeletal cell responses and are important in both innate and adaptive immunity. These responses play a major role in providing a fine balance in a cascade of biological events that results in either protective acute inflammation or chronic inflammation that leads to a host of diseases including autoimmune inflammation mediated epidermolysis bullosa acquisita (EBA). This review describes the role of the actin cytoskeleton and in particular the actin remodelling protein called Flightless I (Flii) in regulating cellular inflammatory responses and its subsequent effect on the autoimmune skin blistering disease EBA. It also outlines the potential of an antibody based therapy for decreasing Flii expression in vivo to ameliorate the symptoms associated with EBA.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Future Industries Institute, Regenerative Medicine, University of South Australia, Mawson Lakes 5095, Adelaide, Australia.
| | - Ralf J Ludwig
- Institute of Experimental Dermatology, University of Lubeck, Lubeck 23562, Germany.
| | - Allison J Cowin
- Future Industries Institute, Regenerative Medicine, University of South Australia, Mawson Lakes 5095, Adelaide, Australia.
| |
Collapse
|
20
|
Suhara K, Miyazaki Y, Okamoto T, Ishizuka M, Tsuchiya K, Inase N. Fragmented gelsolins are increased in rheumatoid arthritis-associated interstitial lung disease with usual interstitial pneumonia pattern. Allergol Int 2016; 65:88-95. [PMID: 26666486 DOI: 10.1016/j.alit.2015.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 08/06/2015] [Accepted: 08/06/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) occurs in 10%-30% of patients with RA, and interstitial lung disease (ILD) is associated with increased mortality in up to 10% of patients with RA. The pathogenesis of RA-ILD is virtually unknown. The aim of this study is to investigate the proteins related to UIP pattern by comparing to OP pattern in RA-ILD using proteome analysis of bronchoalveolar lavage fluid (BALF). METHODS Proteomic differences in BALF were compared between the UIP pattern and OP pattern by examining BALF from 5 patients with the UIP pattern and 7 patients with the OP pattern by two-dimensional gel electrophoresis and mass spectrometry. RESULTS In individual comparisons of BALF samples, the levels of the protein gelsolin and Ig kappa chain C region were significantly higher in the UIP pattern than in the OP pattern. In contrast, the levels of α-1 antitrypsin, CRP, haptoglobin β, and surfactant protein A (isoform number 5) were all significantly higher in the OP pattern than in the UIP pattern. Gelsolin was cleaved into two fragments, a C-terminal half and N-terminal half, and the levels of both were significantly higher in the UIP pattern than in the OP pattern. CONCLUSIONS Fragmented gelsolins may be associated with the pathogenesis of fibrosis in RA-ILD.
Collapse
Affiliation(s)
- Kozo Suhara
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Tsukasa Okamoto
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahiro Ishizuka
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kimitake Tsuchiya
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Naohiko Inase
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
21
|
Abstract
BACKGROUND Acute rheumatic fever is an autoimmune, inflammatory, and multi-systemic disease secondary to pharyngitis and is caused by group A streptococcus. In developing countries, acute rheumatic fever is the most common cause of acquired heart disease. Gelsolin is a calcium-dependent, multi-functional actin-regulatory protein circulating in the plasma of healthy human beings. The correlation between blood gelsolin levels and inflammatory conditions suggests the potential benefit of gelsolin as a prognostic marker. The aim of the present study was to appraise the association of gelsolin and acute rheumatic carditis in childhood. MATERIALS AND METHODS Plasma gelsolin levels were measured and echocardiographic examinations were performed in patients (n=37) with acute rheumatic carditis and compared with those of age- and gender-matched healthy controls (n=24). RESULTS The plasma gelsolin levels in children with acute rheumatic carditis were significantly lower compared with controls (197±218 versus 322±255 mg/L, p=0.039). There was a significant correlation among gelsolin levels and the grade of mitral regurgitation (p=0.030), left ventricular end-diastolic diameter (p=0.017), and left ventricular end-systolic diameter (p=0.028) at diagnosis. CONCLUSIONS Levels of the gelsolin plasma isoform were decreased in patients with acute rheumatic carditis compared with healthy controls. Gelsolin may be used as a biochemical marker for acute rheumatic carditis.
Collapse
|