1
|
Alshehri RS, Abuzinadah AR, Alrawaili MS, Alotaibi MK, Alsufyani HA, Alshanketi RM, AlShareef AA. A Review of Biomarkers of Amyotrophic Lateral Sclerosis: A Pathophysiologic Approach. Int J Mol Sci 2024; 25:10900. [PMID: 39456682 PMCID: PMC11507293 DOI: 10.3390/ijms252010900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of upper and lower motor neurons. The heterogeneous nature of ALS at the clinical, genetic, and pathological levels makes it challenging to develop diagnostic and prognostic tools that fit all disease phenotypes. Limitations associated with the functional scales and the qualitative nature of mainstay electrophysiological testing prompt the investigation of more objective quantitative assessment. Biofluid biomarkers have the potential to fill that gap by providing evidence of a disease process potentially early in the disease, its progression, and its response to therapy. In contrast to other neurodegenerative diseases, no biomarker has yet been validated in clinical use for ALS. Several fluid biomarkers have been investigated in clinical studies in ALS. Biofluid biomarkers reflect the different pathophysiological processes, from protein aggregation to muscle denervation. This review takes a pathophysiologic approach to summarizing the findings of clinical studies utilizing quantitative biofluid biomarkers in ALS, discusses the utility and shortcomings of each biomarker, and highlights the superiority of neurofilaments as biomarkers of neurodegeneration over other candidate biomarkers.
Collapse
Affiliation(s)
- Rawiah S. Alshehri
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Ahmad R. Abuzinadah
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Moafaq S. Alrawaili
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Muteb K. Alotaibi
- Neurology Department, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia;
| | - Hadeel A. Alsufyani
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Rajaa M. Alshanketi
- Internal Medicine Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia;
| | - Aysha A. AlShareef
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| |
Collapse
|
2
|
Huang L, Liu M, Tang J, Gong Z, Li Z, Yang Y, Zhang M. The role of ALDH2 rs671 polymorphism and C-reactive protein in the phenotypes of male ALS patients. Front Neurosci 2024; 18:1397991. [PMID: 39290715 PMCID: PMC11405379 DOI: 10.3389/fnins.2024.1397991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024] Open
Abstract
Background The aldehyde dehydrogenase 2 (ALDH2) rs671 (A) allele has been implicated in neurodegeneration, potentially through oxidative and inflammatory pathways. The study aims to investigate the effects of the ALDH2 rs671 (A) allele and high sensitivity C-reactive protein (hs-CRP) on the clinical phenotypes of amyotrophic lateral sclerosis (ALS) in male and female patients. Methods Clinical data and ALDH2 rs671 genotype of 143 ALS patients, including 85 males and 58 females, were collected from January 2018 to December 2022. All patients underwent assessment using the Chinese version of the Edinburgh Cognitive and Behavioral ALS Screen (ECAS). Complete blood count and metabolic profiles were measured. Clinical and laboratory parameters were compared between carriers and non-carriers of the rs671 (A) allele in males and females, respectively. The significant parameters and rs671 (A) Allele were included in multivariate linear regression models to identify potential contributors to motor and cognitive impairment. Mediation analysis was employed to evaluate any mediation effects. Results Male patients carrying rs671 (A) allele exhibited higher levels of hs-CRP than non-carriers (1.70 mg/L vs. 0.50 mg/L, p = 0.006). The rs671 (A) allele was identified as an independent risk factor for faster disease progression only in male patients (β = 0.274, 95% CI = 0.048-0.499, p = 0.018). The effect of the rs671 (A) allele on the executive function in male patients was fully mediated by hs-CRP (Indirect effect = -1.790, 95% CI = -4.555--0.225). No effects of the rs671 (A) allele or hs-CRP were observed in female ALS patients. The effects of the ALDH2 rs671 (A) allele and the mediating role of hs-CRP in male patients remained significant in the sensitivity analyses. Conclusion The ALDH2 rs671 (A) allele contributed to faster disease progression and hs-CRP mediated cognitive impairment in male ALS patients.
Collapse
Affiliation(s)
- Lifang Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mao Liu
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Jiahui Tang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhenxiang Gong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Yang
- Department of Neurology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Neurology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
3
|
Salunkhe J, Ugale R. Recent updates on immunotherapy in neurodegenerative diseases. Brain Res 2024; 1845:149205. [PMID: 39197568 DOI: 10.1016/j.brainres.2024.149205] [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: 05/30/2024] [Revised: 08/01/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Neurodegeneration is a progressive event leading to specific neuronal loss due to the accumulation of aberrant proteins. These pathologic forms of proteins further worsen and interfere with normal physiologic mechanisms, which can lead to abnormal proliferation of immune cells and subsequent inflammatory cascades and ultimately neuronal loss. Recently, immunotherapies targeting abnormal, pathologic forms of protein have shown a promising approach to modify the progression of neurodegeneration. Recent advances in immunotherapy have led to the development of novel antibodies against the proteinopathies which can eradicate aggregations of protein as evident from preclinical and clinical studies. Nonetheless, only a few of them have successfully received clinical approval, while others have been discontinued due to a lack of clinical efficacy endpoints. The current review discusses the status of investigational antibodies under clinical trials, their targets for therapeutic action, and evidence for failure or success.
Collapse
Affiliation(s)
- Jotiram Salunkhe
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India.
| |
Collapse
|
4
|
Reis PM, Vargas BS, Rebelo RA, Massafera MP, Prado FM, Oreliana H, de Oliveira HV, Freitas F, Ronsein GE, Miyamoto S, Di Mascio P, Medeiros MHG. Quantitative Analysis of Glutathione and Carnosine Adducts with 4-Hydroxy-2-nonenal in Muscle in a hSOD1 G93A ALS Rat Model. Chem Res Toxicol 2024; 37:1306-1314. [PMID: 39066735 PMCID: PMC11337210 DOI: 10.1021/acs.chemrestox.4c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the dysfunction and death of motor neurons through multifactorial mechanisms that remain unclear. ALS has been recognized as a multisystemic disease, and the potential role of skeletal muscle in disease progression has been investigated. Reactive aldehydes formed as secondary lipid peroxidation products in the redox processes react with biomolecules, such as DNA, proteins, and amino acids, resulting in cytotoxic effects. 4-Hydroxy-2-nonenal (HNE) levels are elevated in the spinal cord motor neurons of ALS patients, and HNE-modified proteins have been identified in the spinal cord tissue of an ALS transgenic mice model, suggesting that reactive aldehydes can contribute to motor neuron degeneration in ALS. One biological pathway of aldehyde detoxification involves conjugation with glutathione (GSH) or carnosine (Car). Here, the detection and quantification of Car, GSH, GSSG (glutathione disulfide), and the corresponding adducts with HNE, Car-HNE, and GS-HNE, were performed in muscle and liver tissues of a hSOD1G93A ALS rat model by reverse-phase high-performance liquid chromatography coupled to electrospray ion trap tandem mass spectrometry in the selected reaction monitoring mode. A significant increase in the levels of GS-HNE and Car-HNE was observed in the muscle tissue of the end-stage ALS animals. Therefore, analyzing variations in the levels of these adducts in ALS animal tissue is crucial from a toxicological perspective and can contribute to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo
V. M. Reis
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Bianca S. Vargas
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Rafael A. Rebelo
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Mariana P. Massafera
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Fernanda M. Prado
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Hector Oreliana
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Henrique V. de Oliveira
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Florêncio
P. Freitas
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Graziella E. Ronsein
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Paolo Di Mascio
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Marisa H. G. Medeiros
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| |
Collapse
|
5
|
Chmielarz M, Sobieszczańska B, Środa-Pomianek K. Metabolic Endotoxemia: From the Gut to Neurodegeneration. Int J Mol Sci 2024; 25:7006. [PMID: 39000116 PMCID: PMC11241432 DOI: 10.3390/ijms25137006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Metabolic endotoxemia is a severe health problem for residents in developed countries who follow a Western diet, disrupting intestinal microbiota and the whole organism's homeostasis. Although the effect of endotoxin on the human immune system is well known, its long-term impact on the human body, lasting many months or even years, is unknown. This is due to the difficulty of conducting in vitro and in vivo studies on the prolonged effect of endotoxin on the central nervous system. In this article, based on the available literature, we traced the path of endotoxin from the intestines to the blood through the intestinal epithelium and factors promoting the development of metabolic endotoxemia. The presence of endotoxin in the bloodstream and the inflammation it induces may contribute to lowering the blood-brain barrier, potentially allowing its penetration into the central nervous system; although, the theory is still controversial. Microglia, guarding the central nervous system, are the first line of defense and respond to endotoxin with activation, which may contribute to the development of neurodegenerative diseases. We traced the pro-inflammatory role of endotoxin in neurodegenerative diseases and its impact on the epigenetic regulation of microglial phenotypes.
Collapse
Affiliation(s)
- Mateusz Chmielarz
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Beata Sobieszczańska
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Wroclaw University of Medicine, Chalubinskiego 3a, 50-368 Wroclaw, Poland
| |
Collapse
|
6
|
Sha Y, Zhang D, Tu J, Zhang R, Shao Y, Chen J, Lu S, Liu X. Chronic exposure to tris(1,3-dichloro-2-propyl) phosphate: Effects on intestinal microbiota and serum metabolism in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116469. [PMID: 38772141 DOI: 10.1016/j.ecoenv.2024.116469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
Tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) is a widely used organophosphate ester that can adversely affect animal or human health. The intestinal microbiota is critical to human health. High-dose exposure to TDCIPP can markedly affect the intestinal ecosystem of mice, but the effects of long-term exposure to lower concentrations of TDCIPP on the intestinal flora and body metabolism remain unclear. In this study, TDCIPP was administered to Sprague-Dawley rats by gavage at a dose of 13.3 mg/kg bw/day for 90 days. TDCIPP increased the relative weight of the kidneys (P = 0.017), but had no effect on the relative weight of the heart, liver, spleen, lungs, testes, and ovaries (P > 0.05). 16 S rRNA gene sequencing revealed that long-term TDCIPP exposure affected the diversity, relative abundance, and functions of rat gut microbes. The serum metabolomics of the rats showed that TDCIPP can disrupt the serum metabolic profiles, result in the up-regulation of 26 metabolites and down-regulation of 3 metabolites, and affect multiple metabolic pathways in rat sera. In addition, the disturbed genera and metabolites were correlated. The functions of some disturbed gut microbes were consistent with the affected metabolic pathways in the sera, and these metabolic pathways were all associated with kidney disease, suggesting that TDCIPP may cause kidney injury in rats by affecting the intestinal flora and serum metabolism.
Collapse
Affiliation(s)
- Yujie Sha
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Duo Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Jiazichao Tu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou 510080, China
| | - Ruyue Zhang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou 510080, China
| | - Yijia Shao
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou 510080, China
| | - Shaoyou Lu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Xiang Liu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou 510080, China.
| |
Collapse
|
7
|
Koehn LM, Steele JR, Schittenhelm RB, Turner BJ, Nicolazzo JA. Sex-Dependent Changes to the Intestinal and Hepatic Abundance of Drug Transporters and Metabolizing Enzymes in the SOD1 G93A Mouse Model of Amyotrophic Lateral Sclerosis. Mol Pharm 2024; 21:1756-1767. [PMID: 38415587 DOI: 10.1021/acs.molpharmaceut.3c01089] [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: 02/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by death and dysfunction of motor neurons that result in a rapidly progressing loss of motor function. While there are some data on alterations at the blood-brain barrier (BBB) in ALS and their potential impact on CNS trafficking of drugs, little is reported on the impact of this disease on the expression of drug-handling proteins in the small intestine and liver. This may impact the dosing of the many medicines that individuals with ALS are prescribed. In the present study, a proteomic evaluation was performed on small intestine and liver samples from postnatal day 120 SOD1G93A mice (a model of familial ALS that harbors a human mutant form of superoxide dismutase 1) and wild-type (WT) littermates (n = 7/genotype/sex). Untargeted, quantitative proteomics was undertaken using either label-based [tandem mass tag (TMT)] or label-free [data-independent acquisition (DIA)] acquisition strategies on high-resolution mass spectrometric instrumentation. Copper chaperone for superoxide dismutase (CCS) was significantly higher in SOD1G93A samples compared to the WT samples for both sexes and tissues, therefore representing a potential biomarker for ALS in this mouse model. Relative to WT mice, male SOD1G93A mice had significantly different proteins (Padj < 0.05, |fold-change|>1.2) in the small intestine (male 22, female 1) and liver (male 140, female 3). This included an up-regulation of intestinal transporters for dietary glucose [solute carrier (SLC) SLC5A1] and cholesterol (Niemann-Pick c1-like 1), as well as for several drugs (e.g., SLC15A1), in the male SOD1G93A mice. There was both an up-regulation (e.g., SLCO2A1) and down-regulation (ammonium transporter rh type b) of transporters in the male SOD1G93A liver. In addition, there was both an up-regulation (e.g., phosphoenolpyruvate carboxykinase) and down-regulation (e.g., carboxylesterase 1) of metabolizing enzymes in the male SOD1G93A liver. This proteomic data set identified male-specific changes to key small intestinal and hepatic transporters and metabolizing enzymes that may have important implications for the bioavailability of nutrients and drugs in individuals with ALS.
Collapse
Affiliation(s)
- Liam M Koehn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| |
Collapse
|
8
|
Darabi S, Ariaei A, Rustamzadeh A, Afshari D, Charkhat Gorgich EA, Darabi L. Cerebrospinal fluid and blood exosomes as biomarkers for amyotrophic lateral sclerosis; a systematic review. Diagn Pathol 2024; 19:47. [PMID: 38429818 PMCID: PMC10908104 DOI: 10.1186/s13000-024-01473-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/25/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive and fatal motor neuron disease. Due to the limited knowledge about potential biomarkers that help in early diagnosis and monitoring disease progression, today's diagnoses are based on ruling out other diseases, neurography, and electromyography examination, which takes a time-consuming procedure. METHODS PubMed, ScienceDirect, and Web of Science were explored to extract articles published from January 2015 to June 2023. In the searching strategy following keywords were included; amyotrophic lateral sclerosis, biomarkers, cerebrospinal fluid, serum, and plama. RESULTS A total number of 6 studies describing fluid-based exosomal biomarkers were included in this study. Aggregated proteins including SOD1, TDP-43, pTDP-43, and FUS could be detected in the microvesicles (MVs). Moreover, TDP-43 and NFL extracted from plasma exosomes could be used as prognostic biomarkers. Also, downregulated miR-27a-3p detected through exoEasy Maxi and exoQuick Kit in the plasma could be measured as a diagnostic biomarker. Eventually, the upregulated level of CORO1A could be used to monitor disease progression. CONCLUSION Based on the results, each biomarker alone is insufficient to evaluate ALS. CNS-derived exosomes contain multiple ALS-related biomarkers (SOD1, TDP-43, pTDP-43, FUS, and miRNAs) that are detectable in cerebrospinal fluid and blood is a proper alternation. Exosome detecting kits listed as exoEasy, ExoQuick, Exo-spin, ME kit, ExoQuick Plus, and Exo-Flow, are helpful to reach this purpose.
Collapse
Affiliation(s)
- Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Armin Ariaei
- Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Auob Rustamzadeh
- Cellular and Molecular Research Center, Research Institute for Non-communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Hemmat Highway, next to Milad Tower, Tehran, Iran.
| | - Dariush Afshari
- Department of Neurology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Leila Darabi
- Department of Neurology, Tehran Medical Science Branch, Amir Al Momenin Hospital, Islamic Azad University, Tehran, Iran
| |
Collapse
|
9
|
Gentile F, Maranzano A, Verde F, Bettoni V, Colombo E, Doretti A, Olivero M, Scheveger F, Colombrita C, Bulgarelli I, Spinelli EG, Torresani E, Messina S, Maderna L, Agosta F, Morelli C, Filippi M, Silani V, Ticozzi N. The value of routine blood work-up in clinical stratification and prognosis of patients with amyotrophic lateral sclerosis. J Neurol 2024; 271:794-803. [PMID: 37801095 PMCID: PMC10827966 DOI: 10.1007/s00415-023-12015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND There is an unmet need in amyotrophic lateral sclerosis (ALS) to provide specific biomarkers for the disease. Due to their easy availability, we aimed to investigate whether routine blood parameters provide useful clues for phenotypic classification and disease prognosis. METHODS We analyzed a large inpatient cohort of 836 ALS patients who underwent deep phenotyping with evaluation of the clinical and neurophysiological burden of upper (UMN) and lower (LMN) motor neuron signs. Disability and progression rate were measured through the revised ALS Functional Rating Scale (ALSFRS-R) and its changes during time. Cox regression analysis was performed to assess survival associations. RESULTS Creatinine significantly correlated with LMN damage (r = 0.38), active (r = 0.18) and chronic (r = 0.24) denervation and baseline ALSFRS-R (r = 0.33). Creatine kinase (CK), alanine (ALT) and aspartate (AST) transaminases correlated with active (r = 0.35, r = 0.27, r = 0.24) and chronic (r = 0.37, r = 0.20, r = 0.19) denervation, while albumin and C-reactive protein significantly correlated with LMN score (r = 0.20 and r = 0.17). Disease progression rate showed correlations with chloride (r = -0.19) and potassium levels (r = -0.16). After adjustment for known prognostic factors, total protein [HR 0.70 (95% CI 0.57-0.86)], creatinine [HR 0.86 (95% CI 0.81-0.92)], chloride [HR 0.95 (95% CI 0.92-0.99)], lactate dehydrogenase [HR 0.99 (95% CI 0.99-0.99)], and AST [HR 1.02 (95% CI 1.01-1.02)] were independently associated with survival. CONCLUSIONS Creatinine is a reliable biomarker for ALS, associated with clinical features, disability and survival. Markers of nutrition/inflammation may offer additional prognostic information and partially correlate with clinical features. AST and chloride could further assist in predicting progression rate and survival.
Collapse
Affiliation(s)
- Francesco Gentile
- Neurology Residency Program, Università degli Studi di Milano, Milan, Italy
| | - Alessio Maranzano
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
| | - Federico Verde
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bettoni
- Department of Brain and Behavioral Sciences, IRCCS Mondino Foundation, Università degli Studi di Pavia, Pavia, Italy
| | - Eleonora Colombo
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
| | - Alberto Doretti
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
| | - Marco Olivero
- Neurology Residency Program, Università degli Studi di Milano, Milan, Italy
| | | | - Claudia Colombrita
- Department of Laboratory Medicine, Laboratory of Clinical Chemistry and Microbiology, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Ilaria Bulgarelli
- Department of Laboratory Medicine, Laboratory of Clinical Chemistry and Microbiology, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Edoardo Gioele Spinelli
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Erminio Torresani
- Department of Laboratory Medicine, Laboratory of Clinical Chemistry and Microbiology, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Stefano Messina
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
| | - Luca Maderna
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
| | - Federica Agosta
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Claudia Morelli
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
| | - Massimo Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vincenzo Silani
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology, IRCCS Istituto Auxologico Italiano, P. Le Brescia 20, 20149, Milan, Italy.
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
10
|
Donini L, Tanel R, Zuccarino R, Basso M. Protein biomarkers for the diagnosis and prognosis of Amyotrophic Lateral Sclerosis. Neurosci Res 2023; 197:31-41. [PMID: 37689321 DOI: 10.1016/j.neures.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/11/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common motor neuron disease, still incurable. The disease is highly heterogenous both genetically and phenotypically. Therefore, developing efficacious treatments is challenging in many aspects because it is difficult to predict the rate of disease progression and stratify the patients to minimize statistical variability in clinical studies. Moreover, there is a lack of sensitive measures of therapeutic effect to assess whether a pharmacological intervention ameliorates the disease. There is also urgency of markers that reflect a molecular mechanism dysregulated by ALS pathology and can be rescued when a treatment relieves the condition. Here, we summarize and discuss biomarkers tested in multicentered studies and across different laboratories like neurofilaments, the most used marker in ALS clinical studies, neuroinflammatory-related proteins, p75ECD, p-Tau/t-Tau, and UCHL1. We also explore the applicability of muscle proteins and extracellular vesicles as potential biomarkers.
Collapse
Affiliation(s)
- Luisa Donini
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Italy.
| | - Raffaella Tanel
- Clinical Center NeMO, APSS Ospedale Riabilitativo Villa Rosa, Pergine 38057, TN, Italy.
| | - Riccardo Zuccarino
- Clinical Center NeMO, APSS Ospedale Riabilitativo Villa Rosa, Pergine 38057, TN, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Italy.
| |
Collapse
|
11
|
Batty GD, Kivimäki M, Frank P, Gale CR, Wright L. Systemic inflammation and subsequent risk of amyotrophic lateral sclerosis: Prospective cohort study. Brain Behav Immun 2023; 114:46-51. [PMID: 37543248 PMCID: PMC10937260 DOI: 10.1016/j.bbi.2023.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/14/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND While systemic inflammation has been implicated in the etiology of selected neurodegenerative disorders, its role in the development of amyotrophic lateral sclerosis (ALS), a condition with high case-fatality, is untested. Accordingly, we quantified the relationship of C-reactive protein (CRP), an acute-phase reactant and marker of systemic inflammation, with subsequent ALS occurrence. METHODS We used data from UK Biobank, a prospective cohort study of 502,649 participants who were aged 37 to 73 years when examined at research centers between 2006 and 2010. Venous blood was collected at baseline in the full cohort and assayed for CRP, and repeat measurement was made 3-7 years later in a representative subgroup (N = 14,514) enabling correction for regression dilution. ALS was ascertained via national hospitalization and mortality registries until 2021. We computed multivariable hazard ratios with accompanying 95% confidence intervals for log-transformed CRP expressed as standard deviation and tertiles. RESULTS In an analytical sample of 400,884 initially ALS-free individuals (218,203 women), a mean follow-up of 12 years gave rise to 231 hospitalizations and 223 deaths ascribed to ALS. After adjustment for covariates which included health behaviors, comorbidity, and socio-economic status, a one standard deviation higher log-CRP was associated with elevated rates of both ALS mortality (hazard ratios; 95% confidence intervals: 1.32; 1.13, 1.53) and hospitalizations (1.20; 1.00, 1.39). There was evidence of dose-response effects across tertiles of CRP for both outcomes (p for trend ≤ 0.05). Correction for regression dilution led to a strengthening of the relationship with CRP for both mortality (1.62; 1.27, 2.08) and hospitalizations (1.37; 1.05, 1.76). CONCLUSIONS Higher levels of CRP, a blood-based biomarker widely captured in clinical practice, is associated with moderately increased future risk of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- G David Batty
- Department of Epidemiology and Public Health, University College London, UK.
| | - Mika Kivimäki
- UCL Brain Sciences, University College London, UK; Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Philipp Frank
- Department of Epidemiology and Public Health, University College London, UK.
| | - Catharine R Gale
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, UK.
| | - Liam Wright
- Centre for Longitudinal Studies, University College London, UK.
| |
Collapse
|
12
|
Rogers ML, Schultz DW, Karnaros V, Shepheard SR. Urinary biomarkers for amyotrophic lateral sclerosis: candidates, opportunities and considerations. Brain Commun 2023; 5:fcad287. [PMID: 37946793 PMCID: PMC10631861 DOI: 10.1093/braincomms/fcad287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/23/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Amyotrophic lateral sclerosis is a relentless neurodegenerative disease that is mostly fatal within 3-5 years and is diagnosed on evidence of progressive upper and lower motor neuron degeneration. Around 15% of those with amyotrophic lateral sclerosis also have frontotemporal degeneration, and gene mutations account for ∼10%. Amyotrophic lateral sclerosis is a variable heterogeneous disease, and it is becoming increasingly clear that numerous different disease processes culminate in the final degeneration of motor neurons. There is a profound need to clearly articulate and measure pathological process that occurs. Such information is needed to tailor treatments to individuals with amyotrophic lateral sclerosis according to an individual's pathological fingerprint. For new candidate therapies, there is also a need for methods to select patients according to expected treatment outcomes and measure the success, or not, of treatments. Biomarkers are essential tools to fulfil these needs, and urine is a rich source for candidate biofluid biomarkers. This review will describe promising candidate urinary biomarkers of amyotrophic lateral sclerosis and other possible urinary candidates in future areas of investigation as well as the limitations of urinary biomarkers.
Collapse
Affiliation(s)
- Mary-Louise Rogers
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia
| | - David W Schultz
- Neurology Department and MND Clinic, Flinders Medical Centre, Adelaide 5042, South Australia, Australia
| | - Vassilios Karnaros
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia
| | - Stephanie R Shepheard
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia
| |
Collapse
|
13
|
De Marchi F, Tondo G, Corrado L, Menegon F, Aprile D, Anselmi M, D’Alfonso S, Comi C, Mazzini L. Neuroinflammatory Pathways in the ALS-FTD Continuum: A Focus on Genetic Variants. Genes (Basel) 2023; 14:1658. [PMID: 37628709 PMCID: PMC10454262 DOI: 10.3390/genes14081658] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal dementia (FDT) are progressive neurodegenerative disorders that, in several cases, overlap in clinical presentation, and genetic and pathological disease mechanisms. About 10-15% of ALS cases and up to 40% of FTD are familial, usually with dominant traits. ALS and FTD, in several cases, share common gene mutations, such as in C9ORF72, TARDBP, SQSTM-1, FUS, VCP, CHCHD10, and TBK-1. Also, several mechanisms are involved in ALS and FTD pathogenesis, such as protein misfolding, oxidative stress, and impaired axonal transport. In addition, neuroinflammation and neuroinflammatory cells, such as astrocytes, oligodendrocytes, microglia, and lymphocytes and, overall, the cellular microenvironment, have been proposed as pivotal players in the pathogenesis the ALS-FTD spectrum disorders. This review overviews the current evidence regarding neuroinflammatory markers in the ALS/FTD continuum, focusing on the neuroinflammatory pathways involved in the genetic cases, moving from post-mortem reports to in vivo biofluid and neuroimaging data. We further discuss the potential link between genetic and autoimmune disorders and potential therapeutic implications.
Collapse
Affiliation(s)
- Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy; (G.T.); (D.A.); (C.C.)
| | - Lucia Corrado
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy; (L.C.); (S.D.)
| | - Federico Menegon
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (F.M.); (M.A.)
| | - Davide Aprile
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy; (G.T.); (D.A.); (C.C.)
| | - Matteo Anselmi
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (F.M.); (M.A.)
| | - Sandra D’Alfonso
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy; (L.C.); (S.D.)
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy; (G.T.); (D.A.); (C.C.)
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
| |
Collapse
|
14
|
Mandrioli J, D'Amico R, Zucchi E, De Biasi S, Banchelli F, Martinelli I, Simonini C, Lo Tartaro D, Vicini R, Fini N, Gianferrari G, Pinti M, Lunetta C, Gerardi F, Tarlarini C, Mazzini L, De Marchi F, Scognamiglio A, Sorarù G, Fortuna A, Lauria G, Bella ED, Caponnetto C, Meo G, Chio A, Calvo A, Cossarizza A. Randomized, double-blind, placebo-controlled trial of rapamycin in amyotrophic lateral sclerosis. Nat Commun 2023; 14:4970. [PMID: 37591957 PMCID: PMC10435464 DOI: 10.1038/s41467-023-40734-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
In preclinical studies rapamycin was found to target neuroinflammation, by expanding regulatory T cells, and affecting autophagy, two pillars of amyotrophic lateral sclerosis (ALS) pathogenesis. Herein we report a multicenter, randomized, double-blind trial, in 63 ALS patients who were randomly assigned in a 1:1:1 ratio to receive rapamycin 2 mg/m2/day,1 mg/m2/day or placebo (EUDRACT 2016-002399-28; NCT03359538). The primary outcome, the number of patients exhibiting an increase >30% in regulatory T cells from baseline to treatment end, was not attained. Secondary outcomes were changes from baseline of T, B, NK cell subpopulations, inflammasome mRNA expression and activation status, S6-ribosomal protein phosphorylation, neurofilaments; clinical outcome measures of disease progression; survival; safety and quality of life. Of the secondary outcomes, rapamycin decreased mRNA relative expression of the pro-inflammatory cytokine IL-18, reduced plasmatic IL-18 protein, and increased the percentage of classical monocytes and memory switched B cells, although no corrections were applied for multiple tests. In conclusion, we show that rapamycin treatment is well tolerated and provides reassuring safety findings in ALS patients, but further trials are necessary to understand the biological and clinical effects of this drug in ALS.
Collapse
Affiliation(s)
- Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy.
| | - Roberto D'Amico
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Neurosciences PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Ilaria Martinelli
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Cecilia Simonini
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Vicini
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Nicola Fini
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Gianferrari
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Christian Lunetta
- NEuroMuscular Omnicenter, Serena Onlus Foundation, Milan, Italy
- Istituto Maugeri IRCCS Milano, Milan, Italy
| | | | | | - Letizia Mazzini
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Fabiola De Marchi
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Ada Scognamiglio
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padua, Padua, Italy
- Centro Regionale Specializzato Malattie del Motoneurone, Azienda Ospedale Università di Padova, Padua, Italy
| | - Andrea Fortuna
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Giuseppe Lauria
- 3rd Neurology Unit and ALS Centre, IRCCS 'Carlo Besta' Neurological Institute, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit and ALS Centre, IRCCS 'Carlo Besta' Neurological Institute, Milan, Italy
| | - Claudia Caponnetto
- Department of Neurosciences, Rehabilitatioņ Ophthalmology, Genetics, Mother and Child Disease, Ospedale Policlinico San Martino, Genova, Italy
| | - Giuseppe Meo
- Department of Neurosciences, Rehabilitatioņ Ophthalmology, Genetics, Mother and Child Disease, Ospedale Policlinico San Martino, Genova, Italy
| | - Adriano Chio
- 'Rita Levi Montalcini' Department of Neurosciences, ALS Centre, University of Turin and Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neurosciences, ALS Centre, University of Turin and Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
- National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy
| |
Collapse
|
15
|
Zhou J, Zeng Q, Liao Q, Niu Q, Gu W, Su D, Li S, Xiao B, Bi F. Biomarkers in cerebrospinal fluid for amyotrophic lateral sclerosis phenotypes. Ann Clin Transl Neurol 2023; 10:1467-1480. [PMID: 37350306 PMCID: PMC10424661 DOI: 10.1002/acn3.51836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVE Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease involving both upper and lower motor neurons. The motor phenotypes of ALS are highly clinically heterogeneous, and the underlying mechanisms are poorly understood. METHODS A comparative proteomic analysis was performed in the cerebrospinal fluid (CSF) of bulbar-onset (BO) and spinal-onset (SO) ALS patients and controls (n = 14). Five biomarker candidates were selected from a differentially regulated protein pool, and further validation was performed in a larger independent cohort (n = 92) using enzyme-linked immunosorbent assay (ELISA). RESULTS A total of 1732 CSF proteins were identified, and 78 differentially expressed proteins were found among BO-ALS patients, SO-ALS patients, and controls. Five promising biomarker candidates were selected for further validation, and lipopolysaccharide-binding protein (LBP) and HLA class II histocompatibility antigen, DR alpha chain (HLA-DRA) were validated. CSF LBP levels were increased in ALS patients compared with controls and higher in BO-ALS versus SO-ALS. The increased CSF LBP levels were correlated with the revised ALS Functional Scale (ALSFRS-R) score. CSF HLA-DRA levels were specifically elevated in BO-ALS patients, and there was no significant difference between SO-ALS patients and controls. Increased HLA-DRA expression was correlated with decreased survival. INTERPRETATION Our data shows that elevated CSF LBP is a good biomarker for ALS and correlates with clinical severity, and increased HLA-DRA is a specific biomarker for BO-ALS and may predict short survival. It also suggests that the microglial pathway and HLA-II-related adaptive immunity may be differentially involved in ALS phenotypes and may be new therapeutic targets for ALS.
Collapse
Affiliation(s)
- Jinxia Zhou
- Department of Neurology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
- Hunan Key Laboratary of Aging Biology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
| | - Qianqian Zeng
- Department of Neurology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
| | - Qiao Liao
- Department of Neurology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
| | - Qi Niu
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical University, Nanjing Medical UniversityNanjing210029JiangsuChina
| | - Wenping Gu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
| | - Dandan Su
- Department of Neurology928 Hospital of Joint Logistics Support Force of PLAHaikou571100HainanChina
| | - Sizhuo Li
- Department of Neurology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
| | - Bo Xiao
- Department of Neurology, Xiangya HospitalCentral South UniversityChangsha410008HunanChina
| | - Fangfang Bi
- Department of Neurology, The Fifth Affiliated HospitalSun Yat‐Sen UniversityZhuhai519000GuangdongChina
| |
Collapse
|
16
|
McGrath MS, Zhang R, Bracci PM, Azhir A, Forrest BD. Regulation of the Innate Immune System as a Therapeutic Approach to Supporting Respiratory Function in ALS. Cells 2023; 12:cells12071031. [PMID: 37048104 PMCID: PMC10093136 DOI: 10.3390/cells12071031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a clinical diagnosis used to define a neurodegenerative process that involves progressive loss of voluntary muscle function and leads to death within 2–5 years after diagnosis, in most cases because of respiratory function failure. Respiratory vital capacity (VC) measurements are reproducible and strong predictors of survival. To understand the role of the innate immune response in progressive VC loss we evaluated ALS clinical trial and biomarker results from a 6-month phase 2 study of NP001, a regulator of innate immune function. All ALS baseline values were similar between treated and controls except for those > 65 years old who were excluded from analysis. Treated patients with plasma CRP ≥ 1.13 mg/L (high CRP) showed a 64% slower rate of VC decline compared with placebo and those with plasma CRP < 1.13 mg/L (low CRP) who showed no response. High CRP patients showed no age associated loss of VC whereas low CRP patients showed an age dependent loss of VC function. Plasma levels of serum amyloid A (SAA) were similarly elevated in high CRP patients consistent with ongoing innate immune activation. Plasma TGFB1 in high CRP treated patients was 95% higher than placebo at 6-months, confirming the activation and release of this anti-inflammatory factor by the innate immune alpha 2 macroglobulin (A2M) system. This report is the first to link a biomarker confirmed regulation of the innate immune system with a therapeutic approach for controlling VC loss in ALS patients.
Collapse
Affiliation(s)
- Michael S. McGrath
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
- Neuvivo, Inc., Palo Alto, CA 94301, USA
- Correspondence:
| | - Rongzhen Zhang
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Paige M. Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ari Azhir
- Neuvivo, Inc., Palo Alto, CA 94301, USA
| | - Bruce D. Forrest
- Neuvivo, Inc., Palo Alto, CA 94301, USA
- Hudson Innovations, LLC, Nyack, NY 10960, USA
| |
Collapse
|
17
|
Batty GD, Kivimäki M, Frank P, Gale CR, Wright L. Systemic inflammation and subsequent risk of amyotrophic lateral sclerosis: prospective cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.06.23286852. [PMID: 36945398 PMCID: PMC10029031 DOI: 10.1101/2023.03.06.23286852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Importance While systemic inflammation has been implicated in the aetiology of selected neurodegenerative disorders, its role in the development of amyotrophic lateral sclerosis (ALS) is untested. Objective To quantify the relationship of C-reactive protein (CRP), an acute-phase reactant and marker of systemic inflammation, with ALS occurrence. Design Setting Participants UK Biobank, a prospective cohort study of 502,649 participants who were aged 37 to 73 years when examined at research centres between 2006 and 2010. Exposure Venous blood was collected at baseline in the full cohort and assayed for CRP. Repeat measurement was made 3-7 years later in a representative subgroup (N=14,514) enabling correction for regression dilution. Main Outcomes and Measures ALS as ascertained via national hospitalisation and mortality registries. We computed multi-variable hazard ratios with accompanying 95% confidence intervals for log-transformed CRP expressed as standard deviation and tertiles. Results In an analytical sample of 400,884 individuals (218,203 women), a mean follow-up of 12 years gave rise to 231 hospitalisations and 223 deaths ascribed to ALS. After adjustment for covariates which included health behaviours, comorbidity, and socio-economic status, a one standard deviation higher log-CRP was associated with elevated rates of both ALS mortality (hazard ratios; 95% confidence intervals: 1.32; 1.13, 1.53) and hospitalisations (1.20; 1.00, 1.39). There was evidence of dose-response effects across tertiles of CRP for both outcomes (p for trend≤0.05). Correction for regression dilution led to a strengthening of the relationship with CRP for both mortality (1.62; 1.27, 2.08) and hospitalisations (1.37; 1.05, 1.76) ascribed to ALS. Conclusions and Relevance Higher levels of CRP, a blood-based biomarker widely captured in clinical practice, were associated with a higher subsequent risk of ALS. Key Points Question: Is C-reactive protein (CRP), a marker of systemic inflammation widely used in clinical practice, associated with later risk of amyotrophic lateral sclerosis (ALS)?Findings: Following 11 years disease surveillance in 400,884 individuals (218,203 women), after adjustment for covariates and correction for regression dilution, a one standard deviation higher CRP levels were associations with both mortality (hazard ratio; 95% confidence interval: 1.62; 1.27, 2.08) and hospitalisations (1.37; 1.05, 1.76) ascribed to ALS.Meaning: In the present study, CRP has a dose-response relationship with the risk of later ALS.
Collapse
Affiliation(s)
- G David Batty
- Department of Epidemiology and Public Health, University College London, UK
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, UK Clinicum, Department of Public Health, University of Helsinki, Finland
| | - Philipp Frank
- Department of Epidemiology and Public Health, University College London, UK
| | - Catharine R Gale
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, UK
| | - Liam Wright
- Centre for Longitudinal Studies, University College London, UK
| |
Collapse
|
18
|
Jiang Z, Wang Z, Wei X, Yu XF. Inflammatory checkpoints in amyotrophic lateral sclerosis: From biomarkers to therapeutic targets. Front Immunol 2022; 13:1059994. [PMID: 36618399 PMCID: PMC9815501 DOI: 10.3389/fimmu.2022.1059994] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive motor neuron damage. Due to the complexity of the ALS, so far the etiology and underlying pathogenesis of sporadic ALS are not completely understood. Recently, many studies have emphasized the role of inflammatory networks, which are comprised of various inflammatory molecules and proteins in the pathogenesis of ALS. Inflammatory molecules and proteins may be used as independent predictors of patient survival and might be used in patient stratification and in evaluating the therapeutic response in clinical trials. This review article describes the latest advances in various inflammatory markers in ALS and its animal models. In particular, this review discusses the role of inflammatory molecule markers in the pathogenesis of the disease and their relationship with clinical parameters. We also highlight the advantages and disadvantages of applying inflammatory markers in clinical manifestations, animal studies, and drug clinical trials. Further, we summarize the potential application of some inflammatory biomarkers as new therapeutic targets and therapeutic strategies, which would perhaps expand the therapeutic interventions for ALS.
Collapse
|
19
|
Gelevski D, Addy G, Rohrer M, Cohen C, Roderick A, Winter A, Carey J, Scalia J, Yerton M, Weber H, Doyle M, Parikh N, Kane G, Ellrodt A, Burke K, D'Agostino D, Sinani E, Yu H, Sherman A, Agosti J, Redlich G, Charmley P, Crowe D, Appleby M, Ziegelaar B, Hanus K, Li Z, Babu S, Nicholson K, Luppino S, Berry J, Baecher-Allan C, Paganoni S, Cudkowicz M. Safety and activity of anti-CD14 antibody IC14 (atibuclimab) in ALS: Experience with expanded access protocol. Muscle Nerve 2022; 67:354-362. [PMID: 36533976 DOI: 10.1002/mus.27775] [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: 07/04/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION/AIMS IC14 (atibuclimab) is a monoclonal anti-CD14 antibody. A previous phase 1 trial of 10 participants with amyotrophic lateral sclerosis (ALS) demonstrated initial safety of IC14 in an acute treatment setting. We provided long-term treatment with IC14 to individuals with ALS via an expanded access protocol (EAP) and documented target engagement, biomarker, safety, and disease endpoints. METHODS Participants received intravenous IC14 every 2 weeks. Consistent with United States Food and Drug Administration guidelines, participants were not eligible for clinical trials and the EAP was inclusive of a broad population. Whole blood and serum were collected to determine monocyte CD14 receptor occupancy (RO), IC14 levels, and antidrug antibodies. Ex vivo T-regulatory functional assays were performed in a subset of participants. RESULTS Seventeen participants received IC14 for up to 103 weeks (average, 30.1 weeks; range, 1 to 103 weeks). Treatment-emergent adverse events (TEAEs) were uncommon, mild, and self-limiting. There were 18 serious adverse events (SAEs), which were related to disease progression and unrelated or likely unrelated to IC14. Three participants died due to disease progression. Monocyte CD14 RO increased for all participants after IC14 infusion. One individual required more frequent dosing (every 10 days) to achieve over 80% RO. Antidrug antibodies were detected in only one participant and were transient, low titer, and non-neutralizing. DISCUSSION Administration of IC14 in ALS was safe and well-tolerated in this intermediate-size EAP. Measuring RO guided dosing frequency. Additional placebo-controlled trials are required to determine the efficacy of IC14 in ALS.
Collapse
Affiliation(s)
- Dario Gelevski
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Grace Addy
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Margot Rohrer
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Caroline Cohen
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Aimee Roderick
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Allison Winter
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Judith Carey
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Jennifer Scalia
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Megan Yerton
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Harli Weber
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Michael Doyle
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Neil Parikh
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Geli Kane
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Amy Ellrodt
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Katherine Burke
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Derek D'Agostino
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | | | - Hong Yu
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Alexander Sherman
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Jan Agosti
- Implicit Bioscience, Ltd, Brisbane, Australia
| | | | | | - David Crowe
- Implicit Bioscience, Ltd, Brisbane, Australia
| | | | | | - Katherine Hanus
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Zhenhua Li
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Suma Babu
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Katharine Nicholson
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Sarah Luppino
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - James Berry
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Clare Baecher-Allan
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Sabrina Paganoni
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States.,Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Merit Cudkowicz
- Department of Neurology, Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
20
|
de Luna N, Carbayo Á, Dols-Icardo O, Turon-Sans J, Reyes-Leiva D, Illan-Gala I, Jericó I, Pagola-Lorz I, Lleixà C, Querol L, Rubio-Guerra S, Alcolea D, Fortea J, Lleó A, Cortés-Vicente E, Rojas-Garcia R. Neuroinflammation-Related Proteins NOD2 and Spp1 Are Abnormally Upregulated in Amyotrophic Lateral Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 10:10/2/e200072. [PMID: 36460480 PMCID: PMC9720732 DOI: 10.1212/nxi.0000000000200072] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND AND OBJECTIVES Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of unknown etiology and poorly understood pathophysiology. There is no specific biomarker either for diagnosis or prognosis. The aim of our study was to investigate differentially expressed proteins in the CSF and serum from patients with ALS to determine their role in the disease process and evaluate their utility as diagnostic or prognostic biomarkers. METHODS We performed mass spectrometry in the CSF from 3 patients with ALS and 3 healthy controls (HCs). The results were compared with motor cortex dysregulated transcripts obtained from 11patients with sporadic ALS and 8 HCs. Candidate proteins were tested using ELISA in the serum of 123 patients with ALS, 30 patients with Alzheimer disease (AD), 28 patients with frontotemporal dementia (FTD), and 102 HCs. Patients with ALS, AD, and FTD were prospectively recruited from January 2003 to December 2020. A group of age-matched HCs was randomly selected from the Sant Pau Initiative on Neurodegeneration cohort of the Sant Pau Memory Unit. RESULTS Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) and osteopontin (Spp1) were differentially expressed in the CSF and the motor cortex transcriptome of patients with ALS compared with that in HCs (p < 0.05). NOD2 and Spp1 levels were significantly higher in sera from patients with ALS than in HCs (p < 0.001). Receiver operating characteristic analysis showed an area under the curve of 0.63 for NOD2 and 0.81 for Spp1. NOD2 levels were significantly lower in patients with AD and FTD than in patients with ALS (p < 0.0001), but we found no significant differences in Spp1 levels between patients with ALS, AD (p = 0.51), and FTD (p = 0.42). We found a negative correlation between Spp1 levels and ALS functional rating scale (r = -0.24, p = 0.009). DISCUSSION Our discovery-based approach identified NOD2 as a novel biomarker in ALS and adds evidence to the contribution of Spp1 in the disease process. Both proteins are involved in innate immunity and autophagy and are increased in the serum from patients with ALS. Our data support a relevant role of neuroinflammation in the pathophysiology of the disease and may identify targets for disease-modifying treatments in ALS. Further longitudinal studies should investigate the diagnostic and prognostic value of NOD2 and Spp1 in clinical practice.
Collapse
Affiliation(s)
- Noemí de Luna
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Álvaro Carbayo
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Oriol Dols-Icardo
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Janina Turon-Sans
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - David Reyes-Leiva
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Ignacio Illan-Gala
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Ivonne Jericó
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Inma Pagola-Lorz
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Cinta Lleixà
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Luis Querol
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Sara Rubio-Guerra
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Daniel Alcolea
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Juan Fortea
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Alberto Lleó
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Elena Cortés-Vicente
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.)
| | - Ricardo Rojas-Garcia
- From the Neuromuscular Diseases Laboratory (N.d.C., A.C., D.R.-L., C.L., L.Q., E.C.-V., R.R.-G.), Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant-Pau), Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (N.d.C., A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Madrid; Motor Neuron Diseases Clinic (A.C., J.T.-S., D.R.-L., L.Q., E.C.-V., R.R.-G.), Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Departament de Medicina, Universitat Autònoma de Barcelona; Sant Pau Memory Unit (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) (O.D.-I., I.I.-G., S.R.-G., D.A., J.F., A.L.), Madrid, Spain; and Neuromuscular and Motor Neuron Diseases Research Group-Health Research Institute of Navarra (IdisNA) (I.J., I.P.-L.).
| |
Collapse
|
21
|
Caputi V, Bastiaanssen TFS, Peterson V, Sajjad J, Murphy A, Stanton C, McNamara B, Shorten GD, Cryan JF, O'Mahony SM. Sex, pain, and the microbiome: The relationship between baseline gut microbiota composition, gender and somatic pain in healthy individuals. Brain Behav Immun 2022; 104:191-204. [PMID: 35688340 DOI: 10.1016/j.bbi.2022.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/19/2022] [Accepted: 06/05/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND AIM Relative to men, women present with pain conditions more commonly. Although consistent differences exist between men and women in terms of physiological pain sensitivity, the underlying mechanisms are incompletely understood and yet could inform the development of effective sex specific treatments for pain. The gut microbiota can modulate nervous system functioning, including pain signaling pathways. We hypothesized that the gut microbiota and critical components of the gut-brain axis might influence electrical pain thresholds. Further, we hypothesized that sex, menstrual cycle, and hormonal contraceptive use might account for inter-sex differences in pain perception. METHODS Healthy, non-obese males (N = 15) and females (N = 16), (nine of whom were using hormonal contraceptives), were recruited. Male subjects were invited to undergo testing once, whereas females were invited three times across the menstrual cycle, based on self-reported early follicular (EF), late follicular (LF), or mid-luteal (ML) phase. On test days, electrical stimulation on the right ankle was performed; salivary cortisol levels were measured in the morning; levels of lipopolysaccharide-binding protein (LBP), soluble CD14 (sCD14), pro-inflammatory cytokines were assessed in plasma, and microbiota composition and short-chain fatty acids (SCFAs) levels were determined in fecal samples. RESULTS We observed that the pain tolerance threshold/pain sensation threshold (PTT/PST) ratio was significantly lesser in women than men, but not PST or PTT alone. Further, hormonal contraceptive use was associated with increased LBP levels (LF & ML phase), whilst sCD14 levels or inflammatory cytokines were not affected. Interestingly, in women, hormonal contraceptive use was associated with an increase in the relative abundance of Erysipelatoclostridium, and the relative abundances of certain bacterial genera correlated positively with pain sensation thresholds (Prevotella and Megasphera) during the LF phase and cortisol awakening response (Anaerofustis) during the ML phase. In comparison with men, women displayed overall stronger associations between i) SCFAs data, ii) cortisol data, iii) inflammatory cytokines and PTT and PST. DISCUSSION AND CONCLUSION Our findings support the hypothesis that the gut microbiota may be one of the factors determining the physiological inter-sex differences in pain perception. Further research is needed to investigate the molecular mechanisms by which specific sex hormones and gut microbes modulate pain signaling pathways, but this study highlights the possibilities for innovative individual targeted therapies for pain management.
Collapse
Affiliation(s)
- Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Jahangir Sajjad
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Amy Murphy
- Teagasc Food Research Centre, Moorepark, Co. Cork, Ireland
| | | | - Brian McNamara
- Department of Clinical Neurophysiology, Cork University Hospital, Co. Cork, Ireland
| | - George D Shorten
- Department of Anaesthesia and Intensive Care Medicine, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
22
|
Systemic inflammation biomarkers in 6-OHDA- and LPS-induced Parkinson’s disease in rats. UKRAINIAN BIOCHEMICAL JOURNAL 2022. [DOI: 10.15407/ubj94.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
23
|
Beers DR, Thonhoff JR, Faridar A, Thome AD, Zhao W, Wen S, Appel SH. Tregs Attenuate Peripheral Oxidative Stress and Acute Phase Proteins in ALS. Ann Neurol 2022; 92:195-200. [PMID: 35445431 PMCID: PMC9545429 DOI: 10.1002/ana.26375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
Abstract
Oxidative stress (OS) induces inflammation, which in turn exacerbates OS and the expression of acute phase proteins (APPs). Regulatory T lymphocyte (Treg) therapy was assessed for suppression of OS and APP responses in longitudinal serum samples from subjects with amyotrophic lateral sclerosis (ALS) enrolled in a phase I clinical trial. The first round of Treg therapy suppressed levels of oxidized low‐density lipoprotein (ox‐LDL). During a 6‐month washout period, ox‐LDL levels increased. A second round of therapy again suppressed ox‐LDL levels and then rose following the cessation of treatment. Serum levels of APPs, soluble CD14, lipopolysaccharide binding protein, and C‐reactive protein, were stabilized during Treg administrations, but rose during the washout period and again after therapy was discontinued. Treg therapy potentially suppresses peripheral OS and the accompanying circulating pro‐inflammatory induced APPs, both of which may serve as peripheral candidates for monitoring efficacies of immunomodulating therapies. ANN NEUROL 2022;92:195–200
Collapse
Affiliation(s)
- David R Beers
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Jason R Thonhoff
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Alireza Faridar
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Aaron D Thome
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Weihua Zhao
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Shixiang Wen
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Stanley H Appel
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
24
|
Dreger M, Steinbach R, Otto M, Turner MR, Grosskreutz J. Cerebrospinal fluid biomarkers of disease activity and progression in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2022; 93:422-435. [PMID: 35105727 PMCID: PMC8921583 DOI: 10.1136/jnnp-2021-327503] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/29/2021] [Indexed: 12/04/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive neurodegenerative disease, and only modest disease-modifying strategies have been established to date. Numerous clinical trials have been conducted in the past years, but have been severely hampered by the wide-ranging heterogeneity of both the biological origins and clinical characteristics of the disease. Thus, reliable biomarkers of disease activity are urgently needed to stratify patients into homogenous groups with aligned disease trajectories to allow a more effective design of clinical trial. In this review, the most promising candidate biomarkers in the cerebrospinal fluid (CSF) of patients with ALS will be summarised. Correlations between biomarker levels and clinical outcome parameters are discussed, while highlighting potential pitfalls and intercorrelations of these clinical parameters. Several CSF molecules have shown potential as biomarkers of progression and prognosis, but large, international, multicentric and longitudinal studies are crucial for validation. A more standardised choice of clinical endpoints in these studies, as well as the application of individualised models of clinical progression, would allow the quantification of disease trajectories, thereby allowing a more accurate analysis of the clinical implications of candidate biomarkers. Additionally, a comparative analysis of several biomarkers and ideally the application of a multivariate analysis including comprehensive genotypic, phenotypic and clinical characteristics collectively contributing to biomarker levels in the CSF, could promote their verification. Thus, reliable prognostic markers and markers of disease activity may improve clinical trial design and patient management in the direction of precision medicine.
Collapse
Affiliation(s)
- Marie Dreger
- Department of Neurology, Jena University Hospital, Jena, Thüringen, Germany
| | - Robert Steinbach
- Department of Neurology, Jena University Hospital, Jena, Thüringen, Germany
| | - Markus Otto
- Department of Neurology, University of Halle (Saale), Halle (Saale), Germany
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, Oxfordshire, UK
| | - Julian Grosskreutz
- Precision Neurology, Department of Neurology, University of Luebeck Human Medicine, Luebeck, Schleswig-Holstein, Germany
| |
Collapse
|
25
|
Kharel S, Ojha R, Preethish-Kumar V, Bhagat R. C-reactive protein levels in patients with amyotrophic lateral sclerosis: A systematic review. Brain Behav 2022; 12:e2532. [PMID: 35201675 PMCID: PMC8933772 DOI: 10.1002/brb3.2532] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/31/2021] [Accepted: 02/06/2022] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting cortical and spinal motor neurons. There is a lack of optimal biomarkers to diagnose and prognosticate the ALS patients. C-reactive protein (CRP), an inflammatory marker, has shown promising results in ALS patients. MATERIALS AND METHODS PubMed, Embase, and Google Scholar databases were searched from 2000 to June 1, 2021 for suitable studies showing the relationship between CRP and ALS. The concentration of CRP levels was assessed between ALS patients and controls. Further, end outcomes like ALS functional rating scale (ALSFRS-R), survival status, and mortality risks were assessed in relation to CRP levels. RESULTS Eleven studies including five case-control, five cohorts, and one randomized control study were assessed. There were 2785 ALS patients and 3446 healthy controls. A significant increment in CRP levels among ALS patients in comparison with healthy controls were seen in most of the studies. ALSFRS-R and disease progression were found to be significantly correlated with CRP levels. Overall accuracy of CRP in CSF was 62% described in a single study. CONCLUSION Although CRP has shown promise as a prognostic biomarker, extensive cohort studies are required to assess its prognostic value and accuracy in diagnosing ALS taking into account the confounding factors.
Collapse
Affiliation(s)
- Sanjeev Kharel
- Department of Internal Medicine, Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj, Kathmandu, Nepal
| | - Rajeev Ojha
- Department of Neurology, Tribhuvan University Institute of Medicine, Maharajgunj, Kathmandu, Nepal
| | | | - Riwaj Bhagat
- Department of Neurology, Boston University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Miller RG, Zhang R, Bracci PM, Azhir A, Barohn R, Bedlack R, Benatar M, Berry JD, Cudkowicz M, Kasarskis EJ, Mitsumoto H, Walk D, Shefner J, McGrath MS. Phase
2B
randomized controlled trial of
NP001
in amyotrophic lateral sclerosis: pre‐specified and post‐hoc analyses. Muscle Nerve 2022; 66:39-49. [PMID: 35098554 PMCID: PMC9327716 DOI: 10.1002/mus.27511] [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: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 11/19/2022]
Abstract
Introduction/Aims ALS is a heterogeneous disease that may be complicated or in part driven by inflammation. NP001, a regulator of macrophage activation, was associated with slowing disease progression in those with higher levels of the plasma inflammatory marker C‐reactive protein (CRP) in phase 2A studies in ALS. Here, we evaluate the effects of NP001 in a phase 2B trial, and perform a post hoc analysis with combined data from the preceding phase 2A trial. Methods The phase 2B trial enrolled 138 participants within 3 y of symptom onset and with plasma hs‐CRP values >1.13 mg/L. They were randomized 1:1 to receive either placebo or NP001 for 6 mo. Change from baseline ALSFRS‐R scores was the primary efficacy endpoint. Secondary endpoints included vital capacity (VC) change from baseline and percentage of participants showing no decline of ALSFRS‐R score over 6 mo (non‐progressor). Results The phase 2B study did not show significant differences between placebo and active treatment with respect to change in ALSFRS‐R scores, or VC. The drug was safe and well tolerated. A post hoc analysis identified a 40‐ to 65‐y‐old subset in which NP001‐treated patients demonstrated slower declines in ALSFRS‐R score by 36% and VC loss by 51% compared with placebo. A greater number of non‐progressors were NP001‐treated compared with placebo (p = .004). Discussion Although the phase 2B trial failed to meet its primary endpoints, post hoc analyses identified a subgroup whose decline in ALSFRS‐R and VC scores were significantly slower than placebo. Further studies will be required to validate these findings.
Collapse
Affiliation(s)
| | - Rongzhen Zhang
- Department of Medicine University of California San Francisco San Francisco CA USA
| | - Paige M. Bracci
- Department of Epidemiology and Biostatistics University of California San Francisco San Francisco CA USA
| | | | | | | | | | | | | | | | | | - David Walk
- University of Minnesota Medical School Minneapolis MN USA
| | - Jeremy Shefner
- Barrow Neurological Institute, University of Arizona College of Medicine Phoenix Creighton University College of Medicine Phoenix Phoenix AZ USA
| | - Michael S. McGrath
- Department of Medicine University of California San Francisco San Francisco CA USA
- Neuvivo, Inc. Palo Alto CA USA
| |
Collapse
|
27
|
Staats KA, Borchelt DR, Tansey MG, Wymer J. Blood-based biomarkers of inflammation in amyotrophic lateral sclerosis. Mol Neurodegener 2022; 17:11. [PMID: 35073950 PMCID: PMC8785449 DOI: 10.1186/s13024-022-00515-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease in which many processes are detected including (neuro)inflammation. Many drugs have been tested for ALS in clinical trials but most have failed to reach their primary endpoints. The development and inclusion of different types of biomarkers in diagnosis and clinical trials can assist in determining target engagement of a drug, in distinguishing between ALS and other diseases, and in predicting disease progression rate, drug responsiveness, or an adverse event. Ideally, among other characteristics, a biomarker in ALS correlates highly with a disease process in the central nervous system or with disease progression and is conveniently obtained in a peripheral tissue. Here, we describe the state of biomarkers of inflammation in ALS by focusing on peripherally detectable and cellular responses from blood cells, and provide new (combinatorial) directions for exploration that are now feasible due to technological advancements.
Collapse
Affiliation(s)
- Kim A. Staats
- Staats Life Sciences Consulting, LLC, Los Angeles, CA USA
| | - David R. Borchelt
- Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida USA
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease at The University of Florida College of Medicine, Gainesville, Florida USA
| | - James Wymer
- Department of Neurology, University of Florida College of Medicine, Gainesville, Florida USA
| |
Collapse
|
28
|
Thompson AG, Gray E, Verber N, Bobeva Y, Lombardi V, Shepheard SR, Yildiz O, Feneberg E, Farrimond L, Dharmadasa T, Gray P, Edmond EC, Scaber J, Gagliardi D, Kirby J, Jenkins TM, Fratta P, McDermott CJ, Manohar SG, Talbot K, Malaspina A, Shaw PJ, Turner MR. OUP accepted manuscript. Brain Commun 2022; 4:fcac029. [PMID: 35224491 PMCID: PMC8870425 DOI: 10.1093/braincomms/fcac029] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 11/25/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
The routine clinical integration of individualized objective markers of disease activity in those diagnosed with the neurodegenerative disorder amyotrophic lateral sclerosis is a key requirement for therapeutic development. A large, multicentre, clinic-based, longitudinal cohort was used to systematically appraise the leading candidate biofluid biomarkers in the stratification and potential therapeutic assessment of those with amyotrophic lateral sclerosis. Incident patients diagnosed with amyotrophic lateral sclerosis (n = 258), other neurological diseases (n = 80) and healthy control participants (n = 101), were recruited and followed at intervals of 3–6 months for up to 30 months. Cerebrospinal fluid neurofilament light chain and chitotriosidase 1 and blood neurofilament light chain, creatine kinase, ferritin, complement C3 and C4 and C-reactive protein were measured. Blood neurofilament light chain, creatine kinase, serum ferritin, C3 and cerebrospinal fluid neurofilament light chain and chitotriosidase 1 were all significantly elevated in amyotrophic lateral sclerosis patients. First-visit plasma neurofilament light chain level was additionally strongly associated with survival (hazard ratio for one standard deviation increase in log10 plasma neurofilament light chain 2.99, 95% confidence interval 1.65–5.41, P = 0.016) and rate of disability progression, independent of other prognostic factors. A small increase in level was noted within the first 12 months after reported symptom onset (slope 0.031 log10 units per month, 95% confidence interval 0.012–0.049, P = 0.006). Modelling the inclusion of plasma neurofilament light chain as a therapeutic trial outcome measure demonstrated that a significant reduction in sample size and earlier detection of disease-slowing is possible, compared with using the revised Amyotrophic Lateral Sclerosis Functional Rating Scale. This study provides strong evidence that blood neurofilament light chain levels outperform conventional measures of disease activity at the group level. The application of blood neurofilament light chain has the potential to radically reduce the duration and cost of therapeutic trials. It might also offer a first step towards the goal of more personalized objective disease activity monitoring for those living with amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
| | - Elizabeth Gray
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Nick Verber
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Yoana Bobeva
- Blizard Institute, Queen Mary University of London, London, UK
| | | | - Stephanie R. Shepheard
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Ozlem Yildiz
- Blizard Institute, Queen Mary University of London, London, UK
| | - Emily Feneberg
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Lucy Farrimond
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Thanuja Dharmadasa
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Pamela Gray
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Evan C. Edmond
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jakub Scaber
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Delia Gagliardi
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Thomas M. Jenkins
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Pietro Fratta
- Blizard Institute, Queen Mary University of London, London, UK
| | | | - Sanjay G. Manohar
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Andrea Malaspina
- Blizard Institute, Queen Mary University of London, London, UK
- Correspondence may also be addressed to: Prof Andrea Malaspina Blizard Institute 4 Newark St, Whitechapel London, E1 2AT, UK E-mail:
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
- Correspondence may also be addressed to: Prof Dame Pamela Shaw Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield, 385a Glossop Rd Broomhall, Sheffield, S10 2HQ, UK E-mail:
| | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Correspondence to: Prof Martin Turner Nuffield Department of Clinical Neurosciences Level 6, West Wing, John Radcliffe Hospital Oxford, OX3 9DU, UK E-mail:
| |
Collapse
|
29
|
Targeted drug delivery systems to control neuroinflammation in central nervous system disorders. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
30
|
Thompson AG, Oeckl P, Feneberg E, Bowser R, Otto M, Fischer R, Kessler B, Turner MR. Advancing mechanistic understanding and biomarker development in amyotrophic lateral sclerosis. Expert Rev Proteomics 2021; 18:977-994. [PMID: 34758687 DOI: 10.1080/14789450.2021.2004890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Proteomic analysis has contributed significantly to the study of the neurodegenerative disease amyotrophic lateral sclerosis (ALS). It has helped to define the pathological change common to nearly all cases, namely intracellular aggregates of phosphorylated TDP-43, shifting the focus of pathogenesis in ALS toward RNA biology. Proteomics has also uniquely underpinned the delineation of disease mechanisms in model systems and has been central to recent advances in human ALS biomarker development. AREAS COVERED The contribution of proteomics to understanding the cellular pathological changes, disease mechanisms, and biomarker development in ALS are covered. EXPERT OPINION Proteomics has delivered unique insights into the pathogenesis of ALS and advanced the goal of objective measurements of disease activity to improve therapeutic trials. Further developments in sensitivity and quantification are expected, with application to the presymptomatic phase of human disease offering the hope of prevention strategies.
Collapse
Affiliation(s)
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (Dzne e.V.), Ulm, Germany
| | - Emily Feneberg
- Department of Neurology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Robert Bowser
- Departments of Neurology and Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.,Department of Neurology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Benedikt Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Henderson RD, Agosti JM, McCombe PA, Thorpe K, Heggie S, Heshmat S, Appleby MW, Ziegelaar BW, Crowe DT, Redlich GL. Phase 1b dose-escalation, safety, and pharmacokinetic study of IC14, a monoclonal antibody against CD14, for the treatment of amyotrophic lateral sclerosis. Medicine (Baltimore) 2021; 100:e27421. [PMID: 34678870 PMCID: PMC8542123 DOI: 10.1097/md.0000000000027421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/16/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The primary objective was to demonstrate the safety and tolerability of monoclonal antibody against CD14 (IC14) (atibuclimab) in amyotrophic lateral sclerosis patients. The secondary objectives were pharmacokinetics, pharmacodynamics, and preliminary effects on disease status and biomarkers. METHODS In this open-label, dose-escalation trial, IC14 was administered at 2 mg/kg intravenous (IV) followed by 1 mg/kg/d IV × 3 (n = 3) and in subsequent patients at 4 mg/kg IV followed by 2 mg/kg/d IV × 3 (n = 7) (NCT03487263). Disease status was measured using the Revised Amyotrophic Lateral Sclerosis Functional Rating Scale, forced vital capacity, sniff nasal pressure, Edinburgh Cognitive and Behavioural ALS Screen, and Revised ALS-Specific Quality-of-Life Score. Disease biomarkers included cerebrospinal fluid and serum levels of neurofilament light chain (NfL) and urinary p75 neurotrophin receptor. RESULTS IC14 was safe and well tolerated. No antidrug antibodies were detected. The drug target saturation of monocyte CD14 receptors was rapid and sustained through day 8. There was no significant change in Revised Amyotrophic Lateral Sclerosis Functional Rating Scale, forced vital capacity, sniff nasal pressure, or Revised ALS-Specific Quality-of-Life Score following a single cycle of treatment. Cerebrospinal fluid NfL levels decreased in 6 of 9 patients sampled with declines of 15% to 40% between baseline (not significant [ns]) and day 8 in 3 patients. Serum NfL modestly decreased in 5 of 10 patients (ns) at day 8 and was sustained in 4 (4%-37%, ns) over 33 days of follow up. CONCLUSION IC14 quickly and durably saturated its target in all patients. This study demonstrated safety and tolerability in patients with amyotrophic lateral sclerosis. Even though only a single cycle of treatment was given, there were promising beneficial trends in the neurofilament light chain, a disease biomarker. The emerging understanding of the role of systemic inflammation in neurodegenerative diseases, and the potential for IC14 to serve as a safe, potent, and broad-spectrum inhibitor of immune dysregulation merits further clinical study. CLINICAL TRIAL REGISTRATION NCT03487263.
Collapse
Affiliation(s)
- Robert D. Henderson
- Royal Brisbane & Women's Hospital, Herston, Queensland, Australia
- University of Queensland, Centre for Clinical Research, Herston, Queensland, Australia
| | - Jan M. Agosti
- Implicit Bioscience, Seattle, WA
- Implicit Bioscience, Brisbane, Australia
| | - Pamela A. McCombe
- Royal Brisbane & Women's Hospital, Herston, Queensland, Australia
- University of Queensland, Centre for Clinical Research, Herston, Queensland, Australia
| | - Kathryn Thorpe
- Royal Brisbane & Women's Hospital, Herston, Queensland, Australia
| | - Susan Heggie
- Royal Brisbane & Women's Hospital, Herston, Queensland, Australia
| | - Saman Heshmat
- Royal Brisbane & Women's Hospital, Herston, Queensland, Australia
| | - Mark W. Appleby
- Implicit Bioscience, Seattle, WA
- Implicit Bioscience, Brisbane, Australia
| | - Brian W. Ziegelaar
- Implicit Bioscience, Seattle, WA
- Implicit Bioscience, Brisbane, Australia
| | - David T. Crowe
- Implicit Bioscience, Seattle, WA
- Implicit Bioscience, Brisbane, Australia
| | - Garry L. Redlich
- Implicit Bioscience, Seattle, WA
- Implicit Bioscience, Brisbane, Australia
| |
Collapse
|
32
|
Beers DR, Zhao W, Thonhoff JR, Faridar A, Thome AD, Wen S, Wang J, Appel SH. Serum programmed cell death proteins in amyotrophic lateral sclerosis. Brain Behav Immun Health 2021; 12:100209. [PMID: 34589734 PMCID: PMC8474632 DOI: 10.1016/j.bbih.2021.100209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a multifactorial, multisystem pro-inflammatory neuromuscular disorder. Activation of programmed cell death-1 (PD-1), and its ligands, programmed cell death-ligand 1 and 2 (PD-L1/L2), leads to immune suppression. Serum soluble forms of these proteins, sPD-1/sPD-L1/sPD-L2, inhibit this suppression and promote pro-inflammatory responses. The purpose of this study was to determine if sPD-1, sPD-L1, and sPD-L2 were increased in sera of patients with ALS. sPD-1 and sPD-L2 were elevated in sera of patients and accurately reflected patients’ disease burdens. Increased sera levels of programmed cell death proteins reinforce the concept that peripheral pro-inflammatory responses contribute to systemic inflammation in patients with ALS. Immune regulatory checkpoint pathways play important roles in maintaining the homeostasis of the immune system. ALS and tumor pathobiologies may be thought of as opposite ends of a detrimental versus beneficial spectrum of pro-inflammatory immune responses. Programmed cell death-1 (PD-1) protein, and its ligands, programmed cell death-ligand 1 and 2 (PD-L1/PD-L2), three proteins involved in these regulatory pathways that suppress activation of immune cells. Serum soluble forms of these proteins, sPD-1/sPD-L1/sPD-L2, inhibit this suppression. sPD-1 levels were elevated in the sera of patients with ALS. sPD-1 was only elevated in sera from fast progressing patients with ALS. sPD-L1 was not increased in patients with ALS. sPD-L2 was increased in the sera of patients with ALS. sPD-L2 was increased in fast and slowly progressing patients with ALS. Sera sPD-1 and sPD-L2 positively correlated with patients’ disease burdens. sPD-1 positively correlated with sPD-L2. Sera sPD-1 and sPD-L2 positively correlated with patients’ sera LBP levels. Increased sera levels of programmed cell death proteins reinforce the concept that peripheral pro-inflammatory responses contribute to systemic inflammation in patients with ALS.
Collapse
Affiliation(s)
- David R Beers
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Weihua Zhao
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Jason R Thonhoff
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Alireza Faridar
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Aaron D Thome
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Shixiang Wen
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Jinghong Wang
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Stanley H Appel
- Peggy and Gary Edwards ALS Laboratory, Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
33
|
Wei QQ, Hou YB, Zhang LY, Ou RW, Cao B, Chen YP, Shang HF. Neutrophil-to-lymphocyte ratio in sporadic amyotrophic lateral sclerosis. Neural Regen Res 2021; 17:875-880. [PMID: 34472488 PMCID: PMC8530123 DOI: 10.4103/1673-5374.322476] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) is considered a robust prognostic biomarker for predicting patient survival outcomes in many diseases. However, it remains unclear whether it can be used as a biomarker for amyotrophic lateral sclerosis (ALS). To correlate NLR with disease progression and survival in sporadic ALS, 1030 patients with ALS between January 2012 and December 2018 were included in this study. These patients were assigned into three groups according to their NLR values: Group 1 (NLR < 2, n = 544 [52.8%]), Group 2 (NLR = 2-3, n = 314 [30.5%]), and Group 3 (NLR > 3, n = 172 [16.7%]). All patients were followed up until April 2020. Patients in Group 3 had a significantly older onset age, a lower score on the Revised ALS Functional Rating Scale, and rapidly progressing disease conditions. Furthermore, faster disease progression rates were associated with higher NLR values (odds ratio = 1.211, 95% confidence interval [CI]: 1.090-1.346, P < 0.001) after adjusting for other risk factors. Compared with Groups 1 and 2, the survival time in Group 3 was significantly shorter (log-rank P = 0.002). The NLR value was considered an independent parameter for the prediction of survival in ALS patients after normalizing for all other potential parameters (hazard ratio [HR] = 1.079, 95% CI: 1.016-1.146, P = 0.014). The effects on ALS survival remained significant when adjusted for treatment (HR = 1.074, 95% CI: 1.012-1.141, Ptrend = 0.019) or when considering the stratified NLR value (HR = 1.115, 95% CI: 1.009-1.232, Ptrend = 0.033). Thus, the NLR may help to predict the rate of disease progression and survival in patients with sporadic ALS. The study was approved by the Institutional Ethics Committee of West China Hospital of Sichuan University, China (approval No. 2015 (236)) on December 23, 2015.
Collapse
Affiliation(s)
- Qian-Qian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yan-Bing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ling-Yu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ru-Wei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yong-Ping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui-Fang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
34
|
Amyotrophic lateral sclerosis is a systemic disease: peripheral contributions to inflammation-mediated neurodegeneration. Curr Opin Neurol 2021; 34:765-772. [PMID: 34402459 DOI: 10.1097/wco.0000000000000983] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Neuroinflammation is an important mediator of the pathogenesis of disease in amyotrophic lateral sclerosis (ALS). Genetic mutations such as C9orf72 have begun to define the numerous cell autonomous pathways that initiate motor neuron injury. Yet, it is the signalling to surrounding glia and peripherally derived immune cells that initiates the noncell autonomous inflammatory process and promotes self-propagating motor neuron cell death. The purpose of this review is to explore the systemic immune/inflammatory contributions to the pathogenesis of ALS: what are the peripheral pro-inflammatory signatures, what initiates their presence and do they represent potential therapeutic targets. RECENT FINDINGS In ALS, motor neuron cell death is initiated by multiple cell autonomous pathways leading to misfolded proteins, oxidative stress, altered mitochondria, impaired autophagy and altered RNA metabolism, which collectively promote noncell autonomous inflammatory reactivity. The resulting disease is characterized by activated microglia and astrocytes as well as peripherally derived pro-inflammatory innate and adaptive immune cells. In this unrelenting disorder, circulating blood monocytes and natural killer cells are pro-inflammatory. Furthermore, regulatory T lymphocytes are dysfunctional, and pro-inflammatory cytokines and acute phase proteins are elevated. SUMMARY The collective dysregulation of cells and cytokines in patients with ALS accurately reflect increased disease burdens, more rapid progression rates and reduced survival times, reinforcing the concept of ALS as a disorder with extensive systemic pro-inflammatory responses. These increased systemic pro-inflammatory immune constituents provide potentially meaningful therapeutic targets.
Collapse
|
35
|
Mcgill RB, Steyn FJ, Ngo ST, Thorpe KA, Heggie S, Henderson RD, Mccombe PA, Woodruff TM. Monocyte CD14 and HLA-DR expression increases with disease duration and severity in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2021; 23:430-437. [PMID: 34396845 DOI: 10.1080/21678421.2021.1964531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objective: To investigate changes in immune markers and frequencies throughout disease progression in patients with amyotrophic lateral sclerosis (ALS). Methods: In this longitudinal study, serial blood samples were collected from 21 patients with ALS over a time period of up to 16 months. Flow cytometry was used to quantitate CD14, HLA-DR, and CD16 marker expression on monocyte subpopulations and neutrophils, as well as their cell population frequencies. A Generalized Estimating Equation model was used to assess the association between changes in these immune parameters and disease duration and the Revised Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS-R). Results: CD14 expression on monocyte subpopulations increased with both disease duration and a decrease in ALSFRS-R score in patients with ALS. HLA-DR expression on monocyte subpopulations also increased with disease severity and/or duration. The expression of CD16 did not change relative to disease duration or ALSFRS-R. Finally, patients had a reduction in non-classical monocytes and an increase in the classical to non-classical monocyte ratio throughout disease duration. Conclusion: The progressive immunological changes observed in this study provide further support that monocytes are implicated in ALS pathology. Monocytic CD14 and HLA-DR surface proteins may serve as a therapeutic target or criteria for the recruitment of patients with ALS into clinical trials for immunomodulatory therapies.
Collapse
Affiliation(s)
- R B Mcgill
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - F J Steyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia.,Wesley Medical Research, The Wesley Hospital, Brisbane, Australia.,University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - S T Ngo
- Wesley Medical Research, The Wesley Hospital, Brisbane, Australia.,University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Australia, and.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - K A Thorpe
- Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - S Heggie
- Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - R D Henderson
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - P A Mccombe
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - T M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia.,Wesley Medical Research, The Wesley Hospital, Brisbane, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Australia, and
| |
Collapse
|
36
|
Chen SJ, Chi YC, Ho CH, Yang WS, Lin CH. Plasma Lipopolysaccharide-Binding Protein Reflects Risk and Progression of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2021; 11:1129-1139. [PMID: 33720853 DOI: 10.3233/jpd-212574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Lipopolysaccharide-binding protein (LBP) presents bacterial endotoxin, lipopolysaccharides, to cellular surface pattern receptors for immune responses in the gut-brain axis of Parkinson's disease (PD). OBJECTIVE We investigated whether plasma LBP levels were associated with PD severity and progression. METHODS This study included 397 participants (248 PD patients and 149 controls). We measured participants' plasma levels of LBP and pro-inflammatory cytokines, including TNF-α, IL-6, andIL-17A. PD patients underwent motor and cognition evaluations at baseline and at a mean follow-up interval of 4.7±2.3 years. We assessed the progression of motor and cognition symptoms based on changes in the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) part III motor score and Mini-Mental State Examination (MMSE) score, respectively. RESULTS Plasma LBP levels were lower in PD patients than controls (9.08±2.91 vs. 10.10±3.00μg/ml, p < 0.01). A multiple logistic regression model with adjustment for age, sex, and plasma cytokine levels revealed that reduced plasma LBP levels were associated with increased PD risk (odds ratio 0.816, [95% CI 0.717-0.929], p = 0.002). Among PD patients, LBP levels were correlated with MDS-UPDRS part III motor score after adjustment for confounders (coefficient = 0.636, p = 0.017), but not with MMSE score. Adjusted Cox regression analysis showed that higher plasma LBP levels associated with faster motor progression (adjusted hazard ratio 1.084 [95% CI 1.011-1.163], p = 0.024) during follow-up. CONCLUSION Our results demonstrated that plasma LBP levels reflect risk, motor symptom severity and progression in patients with PD.
Collapse
Affiliation(s)
- Szu-Ju Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Yu-Chiao Chi
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chang-Han Ho
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Shiung Yang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|