1
|
Babu S, Nicholson KA, Rothstein JD, Swenson A, Sampognaro PJ, Pant P, Macklin EA, Spruill S, Paganoni S, Gendron TF, Prudencio M, Petrucelli L, Nix D, Landrette S, Nkrumah E, Fandrick K, Edwards J, Young PR. Apilimod dimesylate in C9orf72 amyotrophic lateral sclerosis: a randomized phase 2a clinical trial. Brain 2024; 147:2998-3008. [PMID: 38606777 DOI: 10.1093/brain/awae109] [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: 11/28/2023] [Revised: 02/18/2024] [Accepted: 03/03/2024] [Indexed: 04/13/2024] Open
Abstract
Apilimod dimesylate is a first-in-class phosphoinositide kinase, FYVE-type zinc finger-containing (PIKfyve) inhibitor with a favourable clinical safety profile and has demonstrated activity in preclinical C9orf72 and TDP-43 amyotrophic lateral sclerosis (ALS) models. In this ALS clinical trial, the safety, tolerability, CNS penetrance and modulation of pharmacodynamic target engagement biomarkers were evaluated. This phase 2a, randomized, double-blind, placebo-controlled, biomarker-end-point clinical trial was conducted in four US centres (ClinicalTrials.gov NCT05163886). Participants with C9orf72 repeat expansions were randomly assigned (2:1) to receive twice-daily oral treatment with 125 mg apilimod dimesylate capsules or matching placebo for 12 weeks, followed by a 12-week open-label extension. Safety was measured as the occurrence of treatment-emergent or serious adverse events attributable to the study drug and tolerability at trial completion or treatment over 12 weeks. Changes from baseline in plasma and CSF and concentrations of apilimod dimesylate and its active metabolites and of pharmacodynamic biomarkers of PIKfyve inhibition [soluble glycoprotein nonmetastatic melanoma protein B (sGPNMB) upregulation] and disease-specific CNS target engagement [poly(GP)] were measured. Between 16 December 2021 and 7 July 2022, 15 eligible participants were enrolled. There were no drug-related serious adverse events reported in the trial. Fourteen (93%) participants completed the double-blind period with 99% dose compliance [n = 9 (90%) apilimod dimesylate; n = 5 (100%) placebo]. At Week 12, apilimod dimesylate was measurable in CSF at 1.63 ng/ml [standard deviation (SD): 0.937]. At Week 12, apilimod dimesylate increased plasma sGPNMB by >2.5-fold (P < 0.001), indicating PIKfyve inhibition, and lowered CSF poly(GP) protein levels by 73% (P < 0.001), indicating CNS tissue-level proof of mechanism. Apilimod dimesylate met prespecified key safety and biomarker end-points in this phase 2a trial and demonstrated CNS penetrance and pharmacodynamic target engagement. Apilimod dimesylate was observed to result in the greatest reduction in CSF poly(GP) levels observed to date in C9orf72 clinical trials.
Collapse
Affiliation(s)
- Suma Babu
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Katharine A Nicholson
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Sanofi, Cambridge, MA 02139, USA
| | - Jeffrey D Rothstein
- Department of Neurology, Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrea Swenson
- Department of Neurology, University of Iowa, Iowa city, IA 52242, USA
| | - Paul J Sampognaro
- Neuromuscular Division, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Pravin Pant
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Eric A Macklin
- Biostatistics Center at Massachusetts General Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Susan Spruill
- Applied Statistics and Consulting, Spruce Pine, NC 28777, USA
| | - Sabrina Paganoni
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Garau J, Garofalo M, Dragoni F, Scarian E, Di Gerlando R, Diamanti L, Zucca S, Bordoni M, Pansarasa O, Gagliardi S. RNA expression profiling in lymphoblastoid cell lines from mutated and non-mutated amyotrophic lateral sclerosis patients. J Gene Med 2024; 26:e3711. [PMID: 38967638 DOI: 10.1002/jgm.3711] [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: 10/04/2023] [Revised: 04/18/2024] [Accepted: 06/02/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the death of upper and lower motor neurons with an unknown etiology. The difficulty of recovering biological material from patients led to employ lymphoblastoid cell lines (LCLs) as a model for ALS because many pathways, typically located in neurons, are also activated in these cells. METHODS To investigate the expression of coding and long non-coding RNAs in LCLs, a transcriptomic profiling of sporadic ALS (SALS) and mutated patients (FUS, TARDBP, C9ORF72 and SOD1) and matched controls was realized. Thus, differentially expressed genes (DEGs) were investigated among the different subgroups of patients. Peripheral blood mononuclear cells (PBMCs) were isolated and immortalized into LCLs via Epstein-Barr virus infection; RNA was extracted, and RNA-sequencing analysis was performed. RESULTS Gene expression profiles of LCLs were genetic-background-specific; indeed, only 12 genes were commonly deregulated in all groups. Nonetheless, pathways enriched by DEGs in each group were also compared, and a total of 89 Kyoto Encyclopedia of Genes and Genomes (KEGG) terms were shared among all patients. Eventually, the similarity of affected pathways was also assessed when our data were matched with a transcriptomic profile realized in the PBMCs of the same patients. CONCLUSIONS We conclude that LCLs are a good model for the study of RNA deregulation in ALS.
Collapse
Affiliation(s)
| | | | - Francesca Dragoni
- IRCCS Mondino Foundation, Pavia, Italy
- Department of Biology and Biotechnology "L. Spallanzani, University of Pavia, Pavia, Italy
| | - Eveljn Scarian
- IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Rosalinda Di Gerlando
- IRCCS Mondino Foundation, Pavia, Italy
- Department of Biology and Biotechnology "L. Spallanzani, University of Pavia, Pavia, Italy
| | | | | | | | | | | |
Collapse
|
3
|
Alexovič M, Uličná C, Sabo J, Davalieva K. Human peripheral blood mononuclear cells as a valuable source of disease-related biomarkers: Evidence from comparative proteomics studies. Proteomics Clin Appl 2024; 18:e2300072. [PMID: 37933719 DOI: 10.1002/prca.202300072] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/08/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE The discovery of specific and sensitive disease-associated biomarkers for early diagnostic purposes of many diseases is still highly challenging due to various complex molecular mechanisms triggered, high variability of disease-related interactions, and an overlap of manifestations among diseases. Human peripheral blood mononuclear cells (PBMCs) contain protein signatures corresponding to essential immunological interplay. Certain diseases stimulate PBMCs and contribute towards modulation of their proteome which can be effectively identified and evaluated via the comparative proteomics approach. EXPERIMENTAL DESIGN In this review, we made a detailed survey of the PBMCS-derived protein biomarker candidates for a variety of diseases, published in the last 15 years. Articles were preselected to include only comparative proteomics studies. RESULTS PBMC-derived biomarkers were investigated for cancer, glomerular, neurodegenerative/neurodevelopmental, psychiatric, chronic inflammatory, autoimmune, endocrinal, infectious, and other diseases. A detailed review of these studies encompassed the proteomics platforms, proposed candidate biomarkers, their immune cell type specificity, and potential clinical application. CONCLUSIONS Overall, PBMCs have shown a solid potential in giving early diagnostic and prognostic biomarkers for many diseases. The future of PBMC biomarker research should reveal its full potential through well-designed comparative studies and extensive testing of the most promising protein biomarkers identified so far.
Collapse
Affiliation(s)
- Michal Alexovič
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Csilla Uličná
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Ján Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| |
Collapse
|
4
|
Scarian E, Viola C, Dragoni F, Di Gerlando R, Rizzo B, Diamanti L, Gagliardi S, Bordoni M, Pansarasa O. New Insights into Oxidative Stress and Inflammatory Response in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2698. [PMID: 38473944 DOI: 10.3390/ijms25052698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Oxidative stress (OS) and inflammation are two important and well-studied pathological hallmarks of neurodegenerative diseases (NDDs). Due to elevated oxygen consumption, the high presence of easily oxidizable polyunsaturated fatty acids and the weak antioxidant defenses, the brain is particularly vulnerable to oxidative injury. Uncertainty exists over whether these deficits contribute to the development of NDDs or are solely a consequence of neuronal degeneration. Furthermore, these two pathological hallmarks are linked, and it is known that OS can affect the inflammatory response. In this review, we will overview the last findings about these two pathways in the principal NDDs. Moreover, we will focus more in depth on amyotrophic lateral sclerosis (ALS) to understand how anti-inflammatory and antioxidants drugs have been used for the treatment of this still incurable motor neuron (MN) disease. Finally, we will analyze the principal past and actual clinical trials and the future perspectives in the study of these two pathological mechanisms.
Collapse
Affiliation(s)
- Eveljn Scarian
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Camilla Viola
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Via Agostino Bassi 21, 27100 Pavia, Italy
| | - Francesca Dragoni
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Rosalinda Di Gerlando
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Bartolo Rizzo
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Luca Diamanti
- Neuroncology Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Matteo Bordoni
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Orietta Pansarasa
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| |
Collapse
|
5
|
Mohammadi F, Rahimi K, Ahmadi A, Hooshmandi Z, Amini S, Mohammadi A. Anti-inflammatory effects of Mentha pulegium L. extract on human peripheral blood mononuclear cells are mediated by TLR-4 and NF-κB suppression. Heliyon 2024; 10:e24040. [PMID: 38234883 PMCID: PMC10792569 DOI: 10.1016/j.heliyon.2024.e24040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
There is great interest in evaluating the anti-inflammatory properties of new herbal products. Thus, the effects of Mentha pulegium L. extract on gene and protein expressions of pro-inflammatory mediators and transcription factors were determined. The hydro-ethanolic extract of Mentha pulegium L. was obtained and optimal non-cytotoxic concentrations of the extract were determined by MTT assay. Then, three different concentrations of Mentha pulegium L. (10, 30, and 90 μg/mL) were used to pre-treat the lipopolysaccharide (LPS)-stimulated and non-stimulated peripheral blood mononuclear cells (PBMCs) of 10 healthy individuals. Finally, the tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, Toll-like receptor-4 (TLR-4), nuclear factor-kappa B (NF-κB) p65, activator protein-1 (AP-1), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) gene expressions and TNF-α, IL-1β, IL-6, TLR-4, prostaglandin E2 (PGE2), and COX-2 protein levels were measured. MTT results showed that there is no significant difference in cell viability among 10, 20, 40, and 80 μg/mL concentrations of Mentha pulegium L. extract at 24, 48, and 72 h (P > 0.05). The IC50 values were 236.1, 147.0, and 118.0 μg/mL after 24, 48, and 72 h respectively. TNF-α, IL-1β, IL-6, TLR-4, iNOS, and NF-κB p65 mRNA levels in the pre-treated LPS-stimulated PBMCs were concentration-dependently reduced (P < 0.01 for TNF-α, TLR-4, and NF-κB p65; P < 0.05 for IL-1β, IL-6, and iNOS). Also, the protein levels of pro-inflammatory mediators decreased and these differences were significant for TNF-α, IL-1β, and TLR-4 (P < 0.001, P < 0.01, and P < 0.001, respectively). Mentha pulegium L. extract decreased the expression and biosynthesis of pro-inflammatory mediators. These effects are mainly mediated by TLR-4 and NF-κB suppression. Thus, Mentha pulegium L. could be useful in treating or ameliorating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Firouz Mohammadi
- Department of Biology, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Kaveh Rahimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Abbas Ahmadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Zahra Hooshmandi
- Department of Biology, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Sabrieh Amini
- Department of Biology, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
6
|
Jia F, Zhang B, Yu W, Chen Z, Xu W, Zhao W, Wang Z. Exploring the cuproptosis-related molecular clusters in the peripheral blood of patients with amyotrophic lateral sclerosis. Comput Biol Med 2024; 168:107776. [PMID: 38056214 DOI: 10.1016/j.compbiomed.2023.107776] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/08/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive and lethal neurodegenerative disease. Several studies have suggested the involvement of cuproptosis in its pathogenesis. In this research, we intend to explore the cuproptosis-related molecular clusters in ALS and develop a novel cuproptosis-related genes prediction model. METHODS The peripheral blood gene expression data was downloaded from the Gene Expression Omnibus (GEO) online database. Based on the GSE112681 dataset, we investigated the critical cuproptosis-related genes (CuRGs) and pathological clustering of ALS. The immune microenvironment features of the peripheral blood in ALS patients were also examined using the CIBERSORT algorithm. Cluster-specific hub genes were determined by the WGCNA. The most accurate prediction model was selected by comparing the performance of four machine learning techniques. ROC curves and two independent datasets were applied to validate the prediction accuracy. The available compounds targeting these hub genes were filtered to investigate their efficacy in treating ALS. RESULTS We successfully determined four critical cuproptosis-related genes and two pathological clusters with various immune profiles and biological characteristics in ALS. Functional analysis showed that genes in Cluster1 were primarily enriched in pathways closely associated with immunity. The Support Vector Machine (SVM) model exhibited the best discrimination properties with a large area under the curve (AUC = 0.862). Five hub prediction genes (BAP1, SMG1, BCLAF1, DHX15, EIF4G2) were selected to establish a nomogram model, suggesting significant risk prediction potential for ALS. The accuracy of this model in predicting ALS incidence was also demonstrated through calibration curves, nomograms, and decision curve analysis. Finally, three drugs targeting BAP1 were determined through drug-gene interactions. CONCLUSION This study elucidated the complex associations between cuproptosis and ALS and constructed a satisfactory predictive model to explore the pathological characteristics of different clusters in ALS patients.
Collapse
Affiliation(s)
- Fang Jia
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bingchang Zhang
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Weijie Yu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Zheng Chen
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenbin Xu
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenpeng Zhao
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhanxiang Wang
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
7
|
Lualdi M, Casale F, Rizzone MG, Zibetti M, Monti C, Colugnat I, Calvo A, De Marco G, Moglia C, Fuda G, Comi C, Chiò A, Lopiano L, Fasano M, Alberio T. Shared and Unique Disease Pathways in Amyotrophic Lateral Sclerosis and Parkinson's Disease Unveiled in Peripheral Blood Mononuclear Cells. ACS Chem Neurosci 2023; 14:4240-4251. [PMID: 37939393 DOI: 10.1021/acschemneuro.3c00629] [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] [Indexed: 11/10/2023] Open
Abstract
Recent evidence supports an association between amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD). Indeed, prospective population-based studies demonstrated that about one-third of ALS patients develop parkinsonian (PK) signs, even though different neuronal circuitries are involved. In this context, proteomics represents a valuable tool to identify unique and shared pathological pathways. Here, we used two-dimensional electrophoresis to obtain the proteomic profile of peripheral blood mononuclear cells (PBMCs) from PD and ALS patients including a small cohort of ALS patients with parkinsonian signs (ALS-PK). After the removal of protein spots correlating with confounding factors, we applied a sparse partial least square discriminant analysis followed by recursive feature elimination to obtain two protein classifiers able to discriminate (i) PD and ALS patients (30 spots) and (ii) ALS-PK patients among all ALS subjects (20 spots). Functionally, the glycolysis pathway was significantly overrepresented in the first signature, while extracellular interactions and intracellular signaling were enriched in the second signature. These results represent molecular evidence at the periphery for the classification of ALS-PK as ALS patients that manifest parkinsonian signs, rather than comorbid patients suffering from both ALS and PD. Moreover, we confirmed that low levels of fibrinogen in PBMCs is a characteristic feature of PD, also when compared with another movement disorder. Collectively, we provide evidence that peripheral protein signatures are a tool to differentially investigate neurodegenerative diseases and highlight altered biochemical pathways.
Collapse
Affiliation(s)
- Marta Lualdi
- Department of Science and High Technology and Center for Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, Varese, Italy
| | - Federico Casale
- Neurology 1, ALS Expert Center, "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Mario Giorgio Rizzone
- "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Maurizio Zibetti
- "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Chiara Monti
- Department of Science and High Technology and Center for Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, Varese, Italy
| | - Ilaria Colugnat
- Department of Science and High Technology and Center for Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, Varese, Italy
| | - Andrea Calvo
- Neurology 1, ALS Expert Center, "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Giovanni De Marco
- Neurology 1, ALS Expert Center, "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Cristina Moglia
- Neurology 1, ALS Expert Center, "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Giuseppe Fuda
- Neurology 1, ALS Expert Center, "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, University of Piemonte Orientale, and Sant'Andrea Hospital, I-13100 Vercelli, Italy
| | - Adriano Chiò
- Neurology 1, ALS Expert Center, "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Leonardo Lopiano
- "Rita Levi Montalcini" Department of Neuroscience, University of Torino, and AOU Città della Salute e della Scienza, I-10126 Torino, Italy
| | - Mauro Fasano
- Department of Science and High Technology and Center for Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, Varese, Italy
| | - Tiziana Alberio
- Department of Science and High Technology and Center for Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, Varese, Italy
| |
Collapse
|
8
|
Proteomics Analysis of Lymphoblastoid Cell Lines from Patients with Amyotrophic Lateral Sclerosis. Molecules 2023; 28:molecules28052014. [PMID: 36903260 PMCID: PMC10004326 DOI: 10.3390/molecules28052014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) consists of the progressive degeneration of motor neurons, caused by poorly understood mechanisms for which there is no cure. Some of the cellular perturbations associated with ALS can be detected in peripheral cells, including lymphocytes from blood. A related cell system that is very suitable for research consists of human lymphoblastoid cell lines (LCLs), which are immortalized lymphocytes. LCLs that can be easily expanded in culture and can be maintained for long periods as stable cultures. We investigated, on a small set of LCLs, if a proteomics analysis using liquid chromatography followed by tandem mass spectrometry reveals proteins that are differentially present in ALS versus healthy controls. We found that individual proteins, the cellular and molecular pathways in which these proteins participate, are detected as differentially present in the ALS samples. Some of these proteins and pathways are already known to be perturbed in ALS, while others are new and present interest for further investigations. These observations suggest that a more detailed proteomics analysis of LCLs, using a larger number of samples, represents a promising approach for investigating ALS mechanisms and to search for therapeutic agents. Proteomics data are available via ProteomeXchange with identifier PXD040240.
Collapse
|
9
|
Stabel JR, Wherry TLT. Comparison of methods to isolate peripheral blood mononuclear cells from cattle blood. J Immunol Methods 2023; 512:113407. [PMID: 36528086 DOI: 10.1016/j.jim.2022.113407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/23/2022] [Accepted: 12/12/2022] [Indexed: 12/16/2022]
Abstract
Peripheral blood mononuclear cells (PBMCs) are critical for assessment of host immune responses to infectious disease. The isolation of PBMCs from whole blood is a laborious process involving density gradients and multiple centrifugation steps. In the present study we compared a more traditional method of PBMC isolation used in our laboratory to two novel methods of cell isolation for efficiency, cell viability, and enumeration of cell subsets. Our laboratory method uses Histopaque-1077 density gradient in standard conical tubes and this was compared with isolation of cells using SepMate™ tubes, a novel conical tube containing an insert to separate the density gradient. Multiple experiments were performed to optimize the SepMate™ tubes for use with cattle blood. A final experiment was conducted to compare traditional methodology, the optimized SepMate™ method with a more novel method using cell preparation tubes (CPT-10 vacutainers containing density gradient). Results demonstrated that optimization of the SepMate™ tube methodology was necessary, including dilution of blood and addition of centrifugation steps to reduce platelet contamination. The CPT-10 tubes worked well but cell recovery was lower compared to other methods. Both of the newer methods were comparable to a modified version of our traditional laboratory method of PBMC isolation in terms of numbers of recovered viable cells and the frequency of immune cell subsets. Additionally, efficiency was improved, particularly with the SepMate™ tube method, resulting in reduced time in the laboratory as well as reduced usage of plasticware.
Collapse
Affiliation(s)
- Judith R Stabel
- USDA-ARS, National Animal Disease Center, Ames, IA 50010, USA.
| | - Taylor L T Wherry
- Department of Veterinary Pathology, Iowa State University, Ames, IA 50010, USA
| |
Collapse
|
10
|
Biological aspects of nitrogen heterocycles for amyotrophic lateral sclerosis. Appl Microbiol Biotechnol 2022; 107:43-56. [DOI: 10.1007/s00253-022-12317-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022]
|
11
|
Banack SA, Dunlop RA, Stommel EW, Mehta P, Cox PA. miRNA extracted from extracellular vesicles is a robust biomarker of amyotrophic lateral sclerosis. J Neurol Sci 2022; 442:120396. [PMID: 36081303 DOI: 10.1016/j.jns.2022.120396] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND AND OBJECTIVES We examined miRNA biomarkers for ALS extracted from extracellular vesicles in blood samples using a large and diverse patient and control population. Different blood collection and storage protocols by different investigators could impact repeatability of miRNA analysis. We tested the hypotheses that miRNA extracted from extracellular vesicles using immunoaffinity purification techniques are robust and repeatable across investigators, laboratories and in a broad ALS population. METHODS De-identified patient blood plasma samples obtained from the U.S. National ALS Biorepository were compared with plasma from non-ALS controls. Extracellular vesicles were extracted and isolated using L1CAM immunoaffinity purification. Total RNA was extracted, and miRNA quantified using qPCR following careful quality control measures. Gene fold expressions of eight miRNAs were compared using a Mann-Whitney two-tailed test. RESULTS One hundred blinded, blood plasma samples were analyzed. Thirty-five men and 15 women with ALS were compared with controls consisting of 30 men and 20 women. None of the ALS patient cohort reported family members with ALS suggesting sporadic ALS. Five of the eight biomarkers previously published were found to significantly discriminate ALS patient samples from control samples. DISCUSSION The methods used in this study provide a repeatable measure of miRNA biomarkers that statistically differentiate ALS patient samples from control samples. The broad inclusion criteria for both the ALS patient cohort and controls along with the collection of blood samples by different investigators suggest that these methods are robust and represent good candidates for further research and development aimed at clinical application.
Collapse
Affiliation(s)
| | | | - Elijah W Stommel
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Paul Mehta
- Centers for Disease Control and Prevention/Agency for Toxic Substances and Disease Registry, National ALS Registry (CDC/ATSDR), Atlanta, GA, USA
| | | |
Collapse
|
12
|
Bordoni M, Pansarasa O, Scarian E, Cristofani R, Leone R, Fantini V, Garofalo M, Diamanti L, Bernuzzi S, Gagliardi S, Carelli S, Poletti A, Cereda C. Lysosomes Dysfunction Causes Mitophagy Impairment in PBMCs of Sporadic ALS Patients. Cells 2022; 11:cells11081272. [PMID: 35455952 PMCID: PMC9030813 DOI: 10.3390/cells11081272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondria alterations are present in tissues derived from patients and animal models, but no data are available for peripheral blood mononuclear cells (PBMCs) of ALS patients. This work aims to investigate mitophagy in PBMCs of sporadic (sALS) patients and how this pathway can be tuned by using small molecules. We found the presence of morphologically atypical mitochondria by TEM and morphological abnormalities by MitoTracker™. We found a decreased number of healthy mitochondria in sALS PBMCs and an impairment of mitophagy with western blot and immunofluorescence. After rapamycin treatment, we found a higher increase in the LC3 marker in sALS PBMCs, while after NH4Cl treatment, we found a lower increase in the LC3 marker. Finally, mTOR-independent autophagy induction with trehalose resulted in a significant decrease in the lysosomes level sALS PBMCs. Our data suggest that the presence of morphologically altered mitochondria and an inefficient turnover of damaged mitochondria in PBMCs of sALS patients rely on the impairment of the mitophagy pathway. We also found that the induction of the mTOR-independent autophagy pathway leads to a decrease in lysosomes level, suggesting a more sensitivity of sALS PBMCs to trehalose. Such evidence suggests that trehalose could represent an effective treatment for ALS patients.
Collapse
Affiliation(s)
- Matteo Bordoni
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Orietta Pansarasa
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Correspondence: ; Tel.: +0382-380-248
| | - Eveljn Scarian
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | | | - Valentina Fantini
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy;
| | - Maria Garofalo
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Luca Diamanti
- Neuroncology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy;
| | - Stefano Bernuzzi
- Immunohematological and Transfusional Service and Centre of Transplantation Immunology, IRCCS “San Matteo Foundation”, 27100 Pavia, Italy;
| | - Stella Gagliardi
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, 20157 Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | - Cristina Cereda
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| |
Collapse
|