Skip to main content
Erschienen in: BMC Neurology 1/2023

Open Access 01.12.2023 | Research

Neutrophil to lymphocyte ratio in parkinson’s disease: a systematic review and meta-analysis

verfasst von: Samaneh Hosseini, Nasim Shafiabadi, Monireh Khanzadeh, Arshin Ghaedi, Raziyeh Ghorbanzadeh, Amir Azarhomayoun, Aida Bazrgar, Jalil Pezeshki, Hanieh Bazrafshan, Shokoufeh Khanzadeh

Erschienen in: BMC Neurology | Ausgabe 1/2023

Abstract

Background

The goal of this research was to explore the role of Neutrophil to lymphocyte ratio (NLR) in Parkinson’s disease (PD).

Methods

From inception to 4 June 2023, PubMed, Web of Science, and ProQuest were searched for papers comparing NLR in PD to healthy individuals. Standardized mean difference (SMD) with a confidence interval (CI) of 95% were calculated.

Results

A random-effect model revealed that PD patients had elevated NLR values compared to healthy individuals (SMD = 0.81, 95% CI = 0.47 to 1.14, P < 0.001). The results of subgroup analysis were as follows: (1) study design: We observed that patients with PD had higher levels of NLR than healthy controls in either retrospective (SMD = 1.12, 95% CI = 0.58 to 1.66, P < 0.001) or prospective (SMD = 0.43, 95% CI = 0.18 to 0.68, P = 0.001) studies. (2) Ethnicity: We noticed that individuals with PD had higher levels of NLR than healthy controls, whether they were East Asian (SMD = 0.93, 95% CI = 0.22 to 1.63, P = 0.010) or Caucasian (SMD = 0.75, 95% CI = 0.40 to 1.10, P < 0.001).The pooled sensitivity of NLR in the prediction of PD was 0.67 (95% CI = 0.61–0.73), and the pooled specificity was 0.66 (95% CI, 0.61–0.70).

Conclusions

Increased levels of NLR is highly related with the presence of PD. Further research is needed to determine the potential clinical benefits of this simple and low-cost biomarker in the PD diagnosis.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12883-023-03380-7.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Parkinson’s disease (PD) is the second most common neurological illness after Alzheimer’s disease, affecting approximately 7 to 10 million individuals globally [1, 2]. The disease’s most apparent symptoms are related to movement, such as rigidity, movement slowness, tremor, and postural instability [3, 4]. Despite the fact that PD is identified by the appearance of intraneuronal proteinacious cytoplasmic inclusions defined as Lewy bodies and a considerable decrease of dopaminergic neurons in the substantia nigra pars compacta, the illness’s origin remains unknown [57]. The antiparkinson medicines, including dopamine agonists and levodopa are often used to treat PD, and they are clinically beneficial in the early stages of disease. However, as the condition worsens, these treatments become ineffective and have adverse side effects [8, 9]. As a result, there is an urgent need to comprehend the etiology of PD and create innovative medicines to prevent or halt disease development.
Inflammation appears to play a prominent role in the pathologic characteristics and symptoms of PD, according to growing research [1015]. Interleukins and tumor necrosis factors (TNFs) are crucial immune activation signaling molecules that affect both the brain and the periphery [16]. In vivo findings of positron emission tomography and postmortem in PD patients revealed elevated inflammatory reactions, such as microglial activation [17, 18], and higher levels of immunological markers in the brain [1820].
In addition, several studies have compared inflammatory biomarkers in individuals with PD to healthy controls in an effort to better understand the origin of the condition and identify potential biomarkers for it [2124].
Neutrophil to lymphocyte ratio (NLR) is a biomarker based on Complete blood count (CBC), and it shows the balance between immunity and systemic inflammation. Although several new studies found links between PD and NLR, the findings were contradictory [2543]. A meta-analysis on this topic is needed to resolve clinical evidence discrepancies. As far as we know, this is the first systematic review and meta-analysis on this matter.

Methods

Study design

We followed the most recent methodological suggestions provided by the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) to conduct this meta-analysis [44]. We registered our study in PEROSPERO (CRD42023429384).

Eligibility criteria

The following PICO terms served as our basis for inclusion:
(a)
Population: Patients suffering from PD.
 
(b)
Intervention: NLR.
 
(c)
Control: Healthy controls.
 
(d)
Outcomes: The diagnostic value of NLR.
 
(e)
Study design: case-control, cross-sectional, and cohort articles.
 
Conference abstracts, basic science investigations, animal studies, case reports and case series were all excluded.

Search strategy

We searched major data bases like Web of Science, ProQuest, and PubMed from inception until June 4, 2023. We used an extensive list of keywords related to NLR and Parkinson’s disease in our search strategy. We did not impose any language or publishing time constraints.

Study selection

Two review writers independently screened the titles, abstracts, and full texts of publications that appeared in our search to determine which met the criteria for inclusion. If there were any issues, a writer would step in and settle it. Following that, the reference and citation lists of the included papers were examined for further possibly relevant publications.

Data extraction and management

The extracted information from the selected articles are as follows:
Name of the first author, location, publication year, design of the study, demographic characteristics of the population, mean and standard deviation (SD) of the NLR, cut off point, specificity and sensitivity as effect measures.

Assessment of methodological quality

Two review authors independently assessed the methodological quality of the included papers using the Newcastle-Ottawa scale (NOS) [45].

Statistical analysis and data synthesis

Stata 11.2 (Stata Corp, College Station, TX) was employed to conduct the meta-analysis. To compare the NLR values between PD patients and controls, we employed standardized mean difference (SMD) with a 95% confidence interval (CI). To assess the heterogeneity of the included articles, the I2 and Cochran’s Q tests were used. Significant heterogeneity across included articles was noted as I2 > 50% and Q test p-value < 0.05. We also utilized the random-effects model to assess pooled effects since we noticed a substantial level of heterogeneity. We used the “metandi” command to measure negative likelihood ratio, positive likelihood ratio, diagnostic odds ratio (DOR), and pooled sensitivity and specificity of NLR for PD. In sensitivity analysis, we used “metaninf” command to investigate the influence of every single study on the overall meta-analysis estimate. We also prepared a summary receiver operating characteristic (SROC) curve. Finally, we employed the Egger test and the funnel plot to assess publication bias. GRADE (Grading of Recommendations Assessment, Development and Evaluation) approach was used to assess the certainty of evidence [46].

Results

Search results and included studies

There were 1067 total results from the manual search of the article citation list and the database search. Eventually, after removing duplicates and irrelevant records, 20 studies were included to this review [2543, 47]. The PRISMA flow diagram, which illustrates the inclusion and exclusion procedure, appears in Fig. 1.

Characteristics of the population and quality assessment

Totally, 20 articles were included in the analysis, comprising 3584 patients with PD and 2487 healthy controls [2543, 47]. The general features of the included articles are shown in Table 1. According to the NOS scale, the quality assessment revealed that all of the studies were of moderate to high quality (Table 1).
Table 1
General characteristics of included studies
Author
Year
Design
Ethnicity
Parkinson’ disease
Healthy control
SEN
SP
NOS score
N
NLR
N
NLR
Akil
2014
Prospective
Caucasian
51
3.10 ± 1.30
50
2.10 ± 0.32
73
74
7
Ucar
2016
Retrospective
Caucasian
46
2.66 ± 1.05
20
2.46 ± 1.04
  
7
Moghaddam
2018
Prospective
Caucasian
388
2.50 ± 0.90
148
2.20 ± 0.80
  
6
Pekel
2018
Prospective
Caucasian
17
2.32 ± 0.77
21
3.04 ± 1.63
  
8
Solmaz
2018
Not declared
Caucasian
101
3.07
60
2.17
65
75
7
Jiang,S.
2019
Retrospective
East Asian
116
1.87 ± 0.71
99
1.67 ± 0.61
  
8
Kenangil
2019
Not declared
Caucasian
94
2.20 ± 1.13
97
2.18 ± 0.96
  
8
Yazar
2019
Not declared
Caucasian
211
2.28 ± 0.55
200
1.71 ± 0.50
  
8
Jin
2020
Retrospective
Caucasian
183
2.91 ± 1.74
89
3.00 ± 1.50
  
7
Munoz-Delgado
2021
Retrospective
Caucasian
377
2.47 ± 1.10
355
1.98 ± 0.91
  
8
Liu
2021
Retrospective
East Asian
101
2.59 ± 0.87
97
2.02 ± 0.76
49
83
6
Kara
2021
Retrospective
Caucasian
100
2.40 ± 0.10
61
1.90 ± 0.10
70
62
8
Wang2
2021
Retrospective
East Asian
453
8.47 ± 2.74
436
9.03 ± 2.91
  
6
Contaldi
2022
Prospective
Caucasian
58
2.63 ± 1.15
58
2.23 ± 0.78
69
48
7
Jiang,L.
2022
Retrospective
Caucasian
125
2.09 ± 0.89
124
2.04 ± 0.77
  
7
Madetko
2022
Retrospective
Caucasian
98
2.26 ± 1.12
99
1.76 ± 0.71
  
8
Paul
2022
Prospective
Caucasian
562
3.02 ± 1.60
227
2.16 ± 1.20
  
8
Wang1
2022
Retrospective
East Asian
153
2.89 ± 0.15
36
2.00 ± 0.16
  
8
Xing
2022
Prospective
East Asian
69
2.83 ± 2.91
36
1.98 ± 0.86
  
7
Munoz-Delgado
2023
Retrospective
Caucasian
281
2.56 1.16
174
2.18 0.79
  
7
 N:Number; NLR: Neutrophil to lymphocyte ratio

PD patients’ NLR levels

According to a random-effects model, NLR levels were higher in PD patients than in healthy controls (SMD = 0.81, 95% CI = 0.47 to 1.14, P < 0.001, Fig. 2). However, the certainty of evidence was very low (Table 2).
Table 2
GRADE1 Evidence Profile for cohort studies of the neutrophil to lymphocyte ratio in Parkinson’s disease
Certainty assessment
No of patients
Certainty
Importance
№ of studies
Study design
Risk of bias2
Inconsistency3
Indirectness
Imprecision5
Publication bias6
Participants, n
Cases, n
20
observational studies
not serious
very serious
not serious
not serious
none
6071
3584
⨁◯◯◯
Very low
CRITICAL
1Grading of Recommendations Assessment, Development and Evaluation
2Risk of bias based on Newcastle-Ottawa Scale
3When I2 was < 30% inconsistency considered as Not serious limitation, > 50 considered as serious and more than 75% considered as very serious limitation
5Serious limitations when there was fewer than 400 participants for each outcome and very serious limitations when there was fewer than 300 participants for each outcome
6Funnel plot revealed no asymmetry; neither test of publication bias approached P < 0.10
The results of subgroup analysis were shown in Table 3. In the subgroup analysis according to study design, we observed that PD patients exhibited increased levels of NLR in comparison to healthy controls in either retrospective (SMD = 0.93, 95% CI = 0.22 to 1.63, P < 0.010) or prospective (SMD = 0.43, 95% CI = 0.18 to 0.68, P = 0.001) studies (Fig. 3).
Table 3
Results of subgroup analysis
Shared characteristic
Subsets of participants
Number of studies
SMD
95%CI
P-value
I2
Cochran’s Q tests P-value
Study design
Retrospective
11
1.12
0.58 to 1.66
< 0.001
98.1
< 0.001
Prospective
6
0.43
0.18 to 0.68
0.001
75.8
0.001
Ethnicity
East Asian
7
0.93
0.22 to 1.63
0.010
98.0
< 0.001
Caucasian
12
0.75
0.40 to 1.10
< 0.001
95.7
< 0.001
In the subgroup analysis according to ethnicity, we observed that PD patients exhibited increased levels of NLR in comparison to healthy controls in either East Asian (SMD = 0.93, 95% CI = 0.22 to 1.63, P = 0.010) or Caucasian (SMD = 0.75, 95% CI = 0.40 to 1.10, P < 0.001) patients (Fig. 4).

Sensitivity analysis

Our results were stable and not reversed after omitting each study in sensitivity analysis (Fig. 5, Supplemental Table I).

NLR’s diagnostic value in PD

The pooled sensitivity was 0.67 (95% CI = 0.61–0.73), and the pooled specificity was 0.66 (95% CI, 0.61–0.70). The pooled positive likelihood ratio, negative likelihood ratio, diagnostic odds ratio (DOR) of NLR were 2.00(95%CI = 1.66–2.42), 0.48 (95%CI = 0.38–0.60), and 2.06 (95%CI = 1.65–2.58), respectively (Fig. 6).

Publication bias

There were some signs of publication bias among the included articles (Egger test 0.001, Supplemental Figure I).

Discussion

We undertook meta-analysis of 20 studies to evaluate the correlation of NLR with PD. It was discovered that PD patients had higher levels of NLR than healthy controls.
PD is a progressive neurodegenerative illness defined pathologically by emergence of lewy bodies and dopaminergic neuron loss in the substantia nigra. Neuroinflammation has a critical role in lewy body formation and substania nigra cell loss. Both the adaptive and innate immune systems are significantly altered in PD [48]. Inflammation is silent in the brain. The response relies on the production of inflammatory components by microglia in the brain. Microglia produce inflammatory cytokines that appear to play significant roles [49]. Interleukin-1 (IL-1), IL-6, and Tumor necrosis factor-alpha (TNF) are examples of inflammatory cytokines that amplify and sustain immune responses and inflammation. It is interesting that PD patients’ CSF basal ganglia contain elevated levels of TNFα, IL-6, and IL-1b [50]. The BBB’s integrity may be lost as a result of the release of pro-inflammatory cytokines by microglia and also upregulation of adhesive molecules (VCAM and ICAM), which cause infiltration of peripheral immune cell in brain [51]. The NLR is a low-cost, simple-to-use indicator of peripheral inflammation [52, 53]. Increased NLR has been shown in many CNS conditions [54]. Several studies evaluated association of increased NLR with PD and our meta-analysis confirmed this association. This may show important role of inflammation and it may exist several years before clinical manifestation of PD and the immune system involvement is promising for therapeutic targeting. In addition to diagnostic utilities, NLR can be used as a prognostic factor in PD. For example, in 2022, Muñoz-Delgado et al. conducted a retrospective cohort study on 211 PD patients as primary-cohort. For replication goals, they also included 344 separate patients with PD in Parkinson’s Progression Markers Initiative group as PPMI-cohort. They showed that NLR is associated with nigrostriatal dopaminergic system degeneration, assessed using striatal dopamine transporter (DAT) density. Patients with higher levels of NLR had significantly lower DAT levels in the putamen (primary-cohort: P = 0.02; PPMI-cohort: P = 0.02) and the caudate (primary-cohort: P < 0.001; PPMI-cohort: P = 0.05) [55]. In addition, Song et al. prospectively followed up 26,210 participants for 21 years. During the follow-up, 486 incidences of PD were reported. The results showed that NLR (HR = 1.09; 95% CI: 1.00- 1.19), was related to an increased risk of PD [56].
Although the role of chronic inflammation is confirmed in PD, it is unclear whether inflammation is the root cause of neurodegeneration or whether it develops as a consequence of cell degeneration and selective damage process [57]. Also the trigger factor for inflammation such as viral infection or autoimmunity remain to be elucidated. The role of inflammation may be different in various subtypes of PD. Use of anti-inflammatory, anti-viral drug and immune-modulating (immunotherapy)for PD treatment and even preventing of PD can be a new era for management of neurodegenerative disease [51, 58].
Due to the extensive evidence that inflammation and immune activation are characteristics of PD, it is not strange that immune-targeting interventions and anti-inflammatory drugs have progressed in the treatments for PD, as they have in other neurodegenerative illnesses such as amyotrophic lateral sclerosis and Alzheimer’s disease [59, 60]. Trials on anti-inflammatory medication in PD have been profoundly unsatisfactory, as they have been in other neurodegenerative illnesses, and such trials need adjusting the strategy [52]. The incorporation of immune-related end points might be a significant therapeutic factor that influences the design of future, possibly more effective trials that use combination medicines that target many processes, like immunological responses [52, 61].
As the neurodegenerative field grows, it is becoming more important to investigate the role of peripheral and central inflammation in the progression and pathogenesis of PD, one of the key obstacles is the creation of innovative methods, including advanced patient-derived models, machine learning, or single-cell multi-omics that will empower the field to thoroughly examine how immune cells predispose or protect neuron injury, whether or not inflammation and the immune system are significant elements of the disease, and at which stages immune processes play crucial roles and are prognostic of illness course [62, 63]. Therefore, further research in these areas is required. Better knowledge and identification of the immunological processes behind the initial indications of PD may result in innovative medicines, and physicians may one day be able to successfully intervene with repurposed or novel immunomodulatory and anti-inflammatory medications to postpone or slow progression of the illness from the periphery to the CNS [64].
There were several limitations to this study. First, only summary data, rather than individual patient data, can be utilized. Second, we only considered studies that reported mean and SD. Third, there was a significant publication bias in our results. Forth, we could not perform trial sequential analysis (TSA), because it can be applied to analyses on dichotomous data like odds ratio (OR) and hazard ratio (HR), but not on SMD, which was reported in our meta-analysis. We tried to extract dichotomous data like OR from included studies and conduct a new meta-analysis and then, perform TAS; however, except to one study [33], no study had reported OR, so further studies reporting TSA results are needed. Finally, neutrophil and lymphocyte counts are nonspecific measures that might be affected by other circumstances such as infections, inflammation, and medicines. These contemporaneous variables may affect how NLR is measured in some of the included studies since they were not specifically controlled.

Conclusion

In conclusion, high NLR can be regarded as an indicator of inflammation in PD. Still, it needs more investigations to study its diagnostic value, association with the stage and progression of the disease, and therapeutic value. Our results expanded immunological information from Parkinson’s disease patients to build better preclinical models, with the long-term objective of allowing early detection of at-risk people to postpone, and treat the illness more properly.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Leigh RM. Parkinson’s disease. Rumi Michael Leigh; 2019. Leigh RM. Parkinson’s disease. Rumi Michael Leigh; 2019.
2.
Zurück zum Zitat Tan E-K, Chao Y-X, West A, Chan L-L, Poewe W, Jankovic J. Parkinson disease and the immune system—associations, mechanisms and therapeutics. Nat Reviews Neurol. 2020;16(6):303–18. Tan E-K, Chao Y-X, West A, Chan L-L, Poewe W, Jankovic J. Parkinson disease and the immune system—associations, mechanisms and therapeutics. Nat Reviews Neurol. 2020;16(6):303–18.
3.
Zurück zum Zitat Zesiewicz TA. Parkinson disease. CONTINUUM: Lifelong Learning in Neurology. 2019;25(4):896–918.PubMed Zesiewicz TA. Parkinson disease. CONTINUUM: Lifelong Learning in Neurology. 2019;25(4):896–918.PubMed
4.
Zurück zum Zitat Berg D, Borghammer P, Fereshtehnejad S-M, Heinzel S, Horsager J, Schaeffer E, et al. Prodromal Parkinson disease subtypes—key to understanding heterogeneity. Nat Reviews Neurol. 2021;17(6):349–61. Berg D, Borghammer P, Fereshtehnejad S-M, Heinzel S, Horsager J, Schaeffer E, et al. Prodromal Parkinson disease subtypes—key to understanding heterogeneity. Nat Reviews Neurol. 2021;17(6):349–61.
5.
Zurück zum Zitat Balestrino R, Schapira A. Parkinson disease. Eur J Neurol. 2020;27(1):27–42.PubMed Balestrino R, Schapira A. Parkinson disease. Eur J Neurol. 2020;27(1):27–42.PubMed
6.
Zurück zum Zitat Simon DK, Tanner CM, Brundin P. Parkinson disease epidemiology, pathology, genetics, and pathophysiology. Clin Geriatr Med. 2020;36(1):1–12.PubMed Simon DK, Tanner CM, Brundin P. Parkinson disease epidemiology, pathology, genetics, and pathophysiology. Clin Geriatr Med. 2020;36(1):1–12.PubMed
7.
Zurück zum Zitat Limousin P, Foltynie T. Long-term outcomes of deep brain stimulation in Parkinson disease. Nat Reviews Neurol. 2019;15(4):234–42. Limousin P, Foltynie T. Long-term outcomes of deep brain stimulation in Parkinson disease. Nat Reviews Neurol. 2019;15(4):234–42.
8.
Zurück zum Zitat Bloem BR, Okun MS, Klein C. Parkinson’s disease. The Lancet. 2021;397(10291):2284–303. Bloem BR, Okun MS, Klein C. Parkinson’s disease. The Lancet. 2021;397(10291):2284–303.
9.
Zurück zum Zitat Aarsland D, Batzu L, Halliday GM, Geurtsen GJ, Ballard C, Ray Chaudhuri K, et al. Parkinson disease-associated cognitive impairment. Nat Reviews Disease Primers. 2021;7(1):1–21. Aarsland D, Batzu L, Halliday GM, Geurtsen GJ, Ballard C, Ray Chaudhuri K, et al. Parkinson disease-associated cognitive impairment. Nat Reviews Disease Primers. 2021;7(1):1–21.
10.
Zurück zum Zitat Pajares M, Rojo I, Manda A, Boscá G, Cuadrado L. Inflammation in Parkinson’s disease: mechanisms and therapeutic implications. Cells. 2020;9(7):1687.PubMedPubMedCentral Pajares M, Rojo I, Manda A, Boscá G, Cuadrado L. Inflammation in Parkinson’s disease: mechanisms and therapeutic implications. Cells. 2020;9(7):1687.PubMedPubMedCentral
11.
Zurück zum Zitat Han X, Sun S, Sun Y, Song Q, Zhu J, Song N, et al. Small molecule-driven NLRP3 inflammation inhibition via interplay between ubiquitination and autophagy: implications for Parkinson disease. Autophagy. 2019;15(11):1860–81.PubMedPubMedCentral Han X, Sun S, Sun Y, Song Q, Zhu J, Song N, et al. Small molecule-driven NLRP3 inflammation inhibition via interplay between ubiquitination and autophagy: implications for Parkinson disease. Autophagy. 2019;15(11):1860–81.PubMedPubMedCentral
12.
Zurück zum Zitat Borsche M, König IR, Delcambre S, Petrucci S, Balck A, Brüggemann N, et al. Mitochondrial damage-associated inflammation highlights biomarkers in PRKN/PINK1 parkinsonism. Brain. 2020;143(10):3041–51.PubMedPubMedCentral Borsche M, König IR, Delcambre S, Petrucci S, Balck A, Brüggemann N, et al. Mitochondrial damage-associated inflammation highlights biomarkers in PRKN/PINK1 parkinsonism. Brain. 2020;143(10):3041–51.PubMedPubMedCentral
13.
Zurück zum Zitat Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Parkinson’s Disease. 2021;7(1):1–13. Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Parkinson’s Disease. 2021;7(1):1–13.
14.
Zurück zum Zitat Marogianni C, Sokratous M, Dardiotis E, Hadjigeorgiou GM, Bogdanos D, Xiromerisiou G. Neurodegeneration and Inflammation—An interesting interplay in Parkinson’s disease. Int J Mol Sci. 2020;21(22):8421.PubMedPubMedCentral Marogianni C, Sokratous M, Dardiotis E, Hadjigeorgiou GM, Bogdanos D, Xiromerisiou G. Neurodegeneration and Inflammation—An interesting interplay in Parkinson’s disease. Int J Mol Sci. 2020;21(22):8421.PubMedPubMedCentral
15.
Zurück zum Zitat Armstrong MJ, Okun MS. Diagnosis and treatment of Parkinson disease: a review. JAMA. 2020;323(6):548–60.PubMed Armstrong MJ, Okun MS. Diagnosis and treatment of Parkinson disease: a review. JAMA. 2020;323(6):548–60.PubMed
16.
Zurück zum Zitat Rolli-Derkinderen M, Leclair-Visonneau L, Bourreille A, Coron E, Neunlist M, Derkinderen P. Is Parkinson’s disease a chronic low-grade inflammatory bowel disease? J Neurol. 2020;267(8):2207–13.PubMed Rolli-Derkinderen M, Leclair-Visonneau L, Bourreille A, Coron E, Neunlist M, Derkinderen P. Is Parkinson’s disease a chronic low-grade inflammatory bowel disease? J Neurol. 2020;267(8):2207–13.PubMed
17.
Zurück zum Zitat Joshi N, Singh S. Updates on immunity and inflammation in Parkinson disease pathology. J Neurosci Res. 2018;96(3):379–90.PubMed Joshi N, Singh S. Updates on immunity and inflammation in Parkinson disease pathology. J Neurosci Res. 2018;96(3):379–90.PubMed
18.
Zurück zum Zitat McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:3–S7. McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:3–S7.
19.
Zurück zum Zitat Tansey MG, Wallings RL, Houser MC, Herrick MK, Keating CE, Joers V. Inflammation and immune dysfunction in Parkinson disease. Nat Rev Immunol. 2022:1–17. Tansey MG, Wallings RL, Houser MC, Herrick MK, Keating CE, Joers V. Inflammation and immune dysfunction in Parkinson disease. Nat Rev Immunol. 2022:1–17.
20.
Zurück zum Zitat Chen L, Mo M, Li G, Cen L, Wei L, Xiao Y, et al. The biomarkers of immune dysregulation and inflammation response in Parkinson disease. Translational Neurodegeneration. 2016;5(1):1–6. Chen L, Mo M, Li G, Cen L, Wei L, Xiao Y, et al. The biomarkers of immune dysregulation and inflammation response in Parkinson disease. Translational Neurodegeneration. 2016;5(1):1–6.
21.
Zurück zum Zitat Wu Y, Le W, Jankovic J. Preclinical biomarkers of Parkinson disease. Arch Neurol. 2011;68(1):22–30.PubMed Wu Y, Le W, Jankovic J. Preclinical biomarkers of Parkinson disease. Arch Neurol. 2011;68(1):22–30.PubMed
22.
23.
Zurück zum Zitat Miller DB, O’Callaghan JP. Biomarkers of Parkinson’s disease: present and future. Metabolism. 2015;64(3):40–S6. Miller DB, O’Callaghan JP. Biomarkers of Parkinson’s disease: present and future. Metabolism. 2015;64(3):40–S6.
24.
Zurück zum Zitat Delenclos M, Jones DR, McLean PJ, Uitti RJ. Biomarkers in Parkinson’s disease: advances and strategies. Parkinsonism Relat Disord. 2016;22:106–S10. Delenclos M, Jones DR, McLean PJ, Uitti RJ. Biomarkers in Parkinson’s disease: advances and strategies. Parkinsonism Relat Disord. 2016;22:106–S10.
25.
Zurück zum Zitat Akıl E, Bulut A, Kaplan İ, Özdemir HH, Arslan D, Aluçlu MU. The increase of carcinoembryonic antigen (CEA), high-sensitivity C-reactive protein, and neutrophil/lymphocyte ratio in Parkinson’s disease. Neurol Sciences: Official J Italian Neurol Soc Italian Soc Clin Neurophysiol. 2015;36(3):423–8. Akıl E, Bulut A, Kaplan İ, Özdemir HH, Arslan D, Aluçlu MU. The increase of carcinoembryonic antigen (CEA), high-sensitivity C-reactive protein, and neutrophil/lymphocyte ratio in Parkinson’s disease. Neurol Sciences: Official J Italian Neurol Soc Italian Soc Clin Neurophysiol. 2015;36(3):423–8.
26.
Zurück zum Zitat Ataç Uçar C, Gökçe Çokal B, Ünal Artık HA, İnan LE, Yoldaş TK. Comparison of neutrophil-lymphocyte ratio (NLR) in Parkinson’s disease subtypes. Neurol Sciences: Official J Italian Neurol Soc Italian Soc Clin Neurophysiol. 2017;38(2):287–93. Ataç Uçar C, Gökçe Çokal B, Ünal Artık HA, İnan LE, Yoldaş TK. Comparison of neutrophil-lymphocyte ratio (NLR) in Parkinson’s disease subtypes. Neurol Sciences: Official J Italian Neurol Soc Italian Soc Clin Neurophysiol. 2017;38(2):287–93.
27.
Zurück zum Zitat Contaldi E, Magistrelli L, Cosentino M, Marino F, Comi C. Lymphocyte Count and Neutrophil-to-lymphocyte ratio are Associated with mild cognitive impairment in Parkinson’s Disease: a Single-Center Longitudinal Study. J Clin Med. 2022;11(19):5543.PubMedPubMedCentral Contaldi E, Magistrelli L, Cosentino M, Marino F, Comi C. Lymphocyte Count and Neutrophil-to-lymphocyte ratio are Associated with mild cognitive impairment in Parkinson’s Disease: a Single-Center Longitudinal Study. J Clin Med. 2022;11(19):5543.PubMedPubMedCentral
28.
Zurück zum Zitat Jiang L, Zhong Z, Huang J, Bian H, Huang W. Monocytohigh-density lipoprotein ratio has a high predictive value for the diagnosis of multiple system atrophy and the differentiation from Parkinson’s disease. Front Aging Neurosci. 2022;14. Jiang L, Zhong Z, Huang J, Bian H, Huang W. Monocytohigh-density lipoprotein ratio has a high predictive value for the diagnosis of multiple system atrophy and the differentiation from Parkinson’s disease. Front Aging Neurosci. 2022;14.
29.
Zurück zum Zitat Jiang S, Wang Y, Gao H, Luo Q, Wang D, Li Y et al. Cell ratio differences in peripheral blood between early-and late-onset Parkinson’s disease: a case-control study. BioMed Research International. 2019;2019. Jiang S, Wang Y, Gao H, Luo Q, Wang D, Li Y et al. Cell ratio differences in peripheral blood between early-and late-onset Parkinson’s disease: a case-control study. BioMed Research International. 2019;2019.
30.
Zurück zum Zitat Jin H, Gu H-y, Mao C-j, Chen J, Liu C. -f. Association of inflammatory factors and aging in Parkinson’s disease. Neurosci Lett. 2020;736:135259.PubMed Jin H, Gu H-y, Mao C-j, Chen J, Liu C. -f. Association of inflammatory factors and aging in Parkinson’s disease. Neurosci Lett. 2020;736:135259.PubMed
31.
Zurück zum Zitat Kara SP, Altunan B, Unal A. Investigation of the peripheral inflammation (neutrophil–lymphocyte ratio) in two neurodegenerative diseases of the central nervous system. Neurol Sci. 2022;43(3):1799–807.PubMed Kara SP, Altunan B, Unal A. Investigation of the peripheral inflammation (neutrophil–lymphocyte ratio) in two neurodegenerative diseases of the central nervous system. Neurol Sci. 2022;43(3):1799–807.PubMed
32.
Zurück zum Zitat Kenangil G, Ari B, Kaya F, Demir M, Domac F. Red cell distribution width levels in Parkinson’s disease patients. Acta Neurol Belgica. 2020;120(5):1147–50. Kenangil G, Ari B, Kaya F, Demir M, Domac F. Red cell distribution width levels in Parkinson’s disease patients. Acta Neurol Belgica. 2020;120(5):1147–50.
33.
Zurück zum Zitat Liu Z, Fan Q, Wu S, Wan Y, Lei Y. Compared with the monocyte to high-density lipoprotein ratio (MHR) and the neutrophil to lymphocyte ratio (NLR), the neutrophil to high-density lipoprotein ratio (NHR) is more valuable for assessing the inflammatory process in Parkinson’s disease. Lipids Health Dis. 2021;20(1):1–12. Liu Z, Fan Q, Wu S, Wan Y, Lei Y. Compared with the monocyte to high-density lipoprotein ratio (MHR) and the neutrophil to lymphocyte ratio (NLR), the neutrophil to high-density lipoprotein ratio (NHR) is more valuable for assessing the inflammatory process in Parkinson’s disease. Lipids Health Dis. 2021;20(1):1–12.
34.
Zurück zum Zitat Madetko N, Migda B, Alster P, Turski P, Koziorowski D, Friedman A. Platelet-to-lymphocyte ratio and neutrophil-tolymphocyte ratio may reflect differences in PD and MSA-P neuroinflammation patterns. Neurol Neurochir Pol. 2022;56(2):148–55.PubMed Madetko N, Migda B, Alster P, Turski P, Koziorowski D, Friedman A. Platelet-to-lymphocyte ratio and neutrophil-tolymphocyte ratio may reflect differences in PD and MSA-P neuroinflammation patterns. Neurol Neurochir Pol. 2022;56(2):148–55.PubMed
35.
Zurück zum Zitat Muñoz-Delgado L, Macías‐García D, Jesús S, Martín‐Rodríguez JF, Labrador‐Espinosa M, Jiménez‐Jaraba MV, et al. Peripheral Immune Profile and Neutrophil‐to‐lymphocyte ratio in Parkinson’s Disease. Mov Disord. 2021;36(10):2426–30.PubMed Muñoz-Delgado L, Macías‐García D, Jesús S, Martín‐Rodríguez JF, Labrador‐Espinosa M, Jiménez‐Jaraba MV, et al. Peripheral Immune Profile and Neutrophil‐to‐lymphocyte ratio in Parkinson’s Disease. Mov Disord. 2021;36(10):2426–30.PubMed
36.
Zurück zum Zitat Paul KC, Kusters C, Furlong M, Zhang K, Yu Y, Folle AD, et al. Immune system disruptions implicated in whole blood epigenome-wide association study of depression among Parkinson’s disease patients. Brain Behav Immunity-Health. 2022;26:100530. Paul KC, Kusters C, Furlong M, Zhang K, Yu Y, Folle AD, et al. Immune system disruptions implicated in whole blood epigenome-wide association study of depression among Parkinson’s disease patients. Brain Behav Immunity-Health. 2022;26:100530.
37.
Zurück zum Zitat Pekel, NB, Yildiz 1 D, Siğirli 2 D, Yabaci 2 A, Seferoğlu1 M, Güneş1 A. Parkinson’s Disease: Is it Actually an inflammatory disorder? Turkish J Geriatr. 2018;21(4):483–9. Pekel, NB, Yildiz 1 D, Siğirli 2 D, Yabaci 2 A, Seferoğlu1 M, Güneş1 A. Parkinson’s Disease: Is it Actually an inflammatory disorder? Turkish J Geriatr. 2018;21(4):483–9.
38.
Zurück zum Zitat Sanjari Moghaddam H, Ghazi Sherbaf F, Mojtahed Zadeh M, Ashraf-Ganjouei A, Aarabi MH. Association between Peripheral inflammation and DATSCAN Data of the Striatal nuclei in different motor subtypes of Parkinson Disease. Front Neurol. 2018;9:234.PubMedPubMedCentral Sanjari Moghaddam H, Ghazi Sherbaf F, Mojtahed Zadeh M, Ashraf-Ganjouei A, Aarabi MH. Association between Peripheral inflammation and DATSCAN Data of the Striatal nuclei in different motor subtypes of Parkinson Disease. Front Neurol. 2018;9:234.PubMedPubMedCentral
39.
Zurück zum Zitat Solmaz V, Genç EP, Aksoy D, Çevik B, Kurt SG, Benli İ. Parkinson hastalarında nötrofil/lenfosit oranları, C reaktif protein ve sedimantasyon hızlarının değerlendirilmesi. Cukurova Med J (Çukurova Üniversitesi Tıp Fakültesi Dergisi).43(2):1-. Solmaz V, Genç EP, Aksoy D, Çevik B, Kurt SG, Benli İ. Parkinson hastalarında nötrofil/lenfosit oranları, C reaktif protein ve sedimantasyon hızlarının değerlendirilmesi. Cukurova Med J (Çukurova Üniversitesi Tıp Fakültesi Dergisi).43(2):1-.
40.
Zurück zum Zitat Wang L-x, Liu C, Shao Y-q, Jin H, Mao C-j, Chen J. Peripheral blood inflammatory cytokines are Associated with Rapid Eye Movement Sleep Behavior Disorder in Parkinson’s Disease. Neurosci Lett. 2022:136692. Wang L-x, Liu C, Shao Y-q, Jin H, Mao C-j, Chen J. Peripheral blood inflammatory cytokines are Associated with Rapid Eye Movement Sleep Behavior Disorder in Parkinson’s Disease. Neurosci Lett. 2022:136692.
41.
Zurück zum Zitat Wang Y, Gao H, Jiang S, Luo Q, Han X, Xiong Y, et al. Principal component analysis of routine blood test results with Parkinson’s disease: a case-control study. Exp Gerontol. 2021;144:111188.PubMed Wang Y, Gao H, Jiang S, Luo Q, Han X, Xiong Y, et al. Principal component analysis of routine blood test results with Parkinson’s disease: a case-control study. Exp Gerontol. 2021;144:111188.PubMed
42.
Zurück zum Zitat Xing N, Dong Z, Wu Q, Kan P, Han Y, Cheng X et al. Identification and validation of key molecules associated with humoral immune modulation in Parkinson’s disease based on bioinformatics. Front Immunol. 2022;13. Xing N, Dong Z, Wu Q, Kan P, Han Y, Cheng X et al. Identification and validation of key molecules associated with humoral immune modulation in Parkinson’s disease based on bioinformatics. Front Immunol. 2022;13.
43.
Zurück zum Zitat Yazar HO, Yazar T. Serum inflammation biomarkers are associated with stages of Parkinson’s disease. 2019. Yazar HO, Yazar T. Serum inflammation biomarkers are associated with stages of Parkinson’s disease. 2019.
44.
Zurück zum Zitat Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Int J Surg. 2021;88:105906.PubMed Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Int J Surg. 2021;88:105906.PubMed
45.
Zurück zum Zitat Wells GA, Shea B, O’Connell D, Peterson J, Welch V, Losos M et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. Oxford; 2000. Wells GA, Shea B, O’Connell D, Peterson J, Welch V, Losos M et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. Oxford; 2000.
46.
Zurück zum Zitat Goldet G, Howick J. Understanding GRADE: an introduction. J Evidence-Based Med. 2013;6(1):50–4. Goldet G, Howick J. Understanding GRADE: an introduction. J Evidence-Based Med. 2013;6(1):50–4.
47.
Zurück zum Zitat Muñoz-Delgado L, Macías-García D, Periñán MT, Jesús S, Adarmes-Gómez AD, Bonilla Toribio M, et al. Peripheral inflammatory immune response differs among sporadic and familial Parkinson’s disease. npj Parkinson’s Disease. 2023;9(1):12.PubMedPubMedCentral Muñoz-Delgado L, Macías-García D, Periñán MT, Jesús S, Adarmes-Gómez AD, Bonilla Toribio M, et al. Peripheral inflammatory immune response differs among sporadic and familial Parkinson’s disease. npj Parkinson’s Disease. 2023;9(1):12.PubMedPubMedCentral
48.
Zurück zum Zitat Hirsch EC, Vyas S, Hunot S. Neuroinflammation in Parkinson’s disease. Parkinsonism Relat Disord. 2012;18:210–S2. Hirsch EC, Vyas S, Hunot S. Neuroinflammation in Parkinson’s disease. Parkinsonism Relat Disord. 2012;18:210–S2.
49.
Zurück zum Zitat Hirsch EC, Standaert DG. Ten unsolved questions about neuroinflammation in Parkinson’s disease. Mov Disord. 2021;36(1):16–24.PubMed Hirsch EC, Standaert DG. Ten unsolved questions about neuroinflammation in Parkinson’s disease. Mov Disord. 2021;36(1):16–24.PubMed
50.
Zurück zum Zitat Tiwari PC, Pal R. The potential role of neuroinflammation and transcription factors in Parkinson disease. Dialogues in clinical neuroscience. 2022. Tiwari PC, Pal R. The potential role of neuroinflammation and transcription factors in Parkinson disease. Dialogues in clinical neuroscience. 2022.
51.
Zurück zum Zitat De Virgilio A, Greco A, Fabbrini G, Inghilleri M, Rizzo MI, Gallo A, et al. Parkinson’s disease: autoimmunity and neuroinflammation. Autoimmun rev. 2016;15(10):1005–11.PubMed De Virgilio A, Greco A, Fabbrini G, Inghilleri M, Rizzo MI, Gallo A, et al. Parkinson’s disease: autoimmunity and neuroinflammation. Autoimmun rev. 2016;15(10):1005–11.PubMed
52.
Zurück zum Zitat Wang Q, Liu Y, Zhou J. Neuroinflammation in Parkinson’s disease and its potential as therapeutic target. Translational Neurodegeneration. 2015;4(1):1–9. Wang Q, Liu Y, Zhou J. Neuroinflammation in Parkinson’s disease and its potential as therapeutic target. Translational Neurodegeneration. 2015;4(1):1–9.
53.
Zurück zum Zitat Song S-Y, Zhao X-X, Rajah G, Hua C, Kang R-j, Han Y-p, et al. Clinical significance of baseline neutrophil-to-lymphocyte ratio in patients with ischemic stroke or hemorrhagic stroke: an updated meta-analysis. Front Neurol. 2019;10:1032.PubMedPubMedCentral Song S-Y, Zhao X-X, Rajah G, Hua C, Kang R-j, Han Y-p, et al. Clinical significance of baseline neutrophil-to-lymphocyte ratio in patients with ischemic stroke or hemorrhagic stroke: an updated meta-analysis. Front Neurol. 2019;10:1032.PubMedPubMedCentral
54.
Zurück zum Zitat Yu S, Arima H, Bertmar C, Clarke S, Herkes G, Krause M. Neutrophil to lymphocyte ratio and early clinical outcomes in patients with acute ischemic stroke. J Neurol Sci. 2018;387:115–8.PubMed Yu S, Arima H, Bertmar C, Clarke S, Herkes G, Krause M. Neutrophil to lymphocyte ratio and early clinical outcomes in patients with acute ischemic stroke. J Neurol Sci. 2018;387:115–8.PubMed
55.
Zurück zum Zitat Muñoz-Delgado L, Labrador‐Espinosa M, Macías‐García D, Jesús S, Benitez Zamora B, Fernández‐Rodríguez P et al. Peripheral inflammation is Associated with Dopaminergic Degeneration in Parkinson’s Disease. Mov Disord. 2023. Muñoz-Delgado L, Labrador‐Espinosa M, Macías‐García D, Jesús S, Benitez Zamora B, Fernández‐Rodríguez P et al. Peripheral inflammation is Associated with Dopaminergic Degeneration in Parkinson’s Disease. Mov Disord. 2023.
56.
Zurück zum Zitat Song L, Zhang S, Li H, Hansson O, Sonestedt E, Borné Y. Comparison of risk factors for Parkinson’s disease, coronary events and ischemic stroke. npj Parkinson’s Disease. 2022;8(1):107.PubMedPubMedCentral Song L, Zhang S, Li H, Hansson O, Sonestedt E, Borné Y. Comparison of risk factors for Parkinson’s disease, coronary events and ischemic stroke. npj Parkinson’s Disease. 2022;8(1):107.PubMedPubMedCentral
57.
Zurück zum Zitat Gelders G, Baekelandt V, Van der Perren A. Linking neuroinflammation and neurodegeneration in Parkinson’s disease. Journal of immunology research. 2018;2018. Gelders G, Baekelandt V, Van der Perren A. Linking neuroinflammation and neurodegeneration in Parkinson’s disease. Journal of immunology research. 2018;2018.
58.
Zurück zum Zitat More SV, Kumar H, Kim IS, Song S-Y, Choi D-K. Cellular and molecular mediators of neuroinflammation in the pathogenesis of Parkinson’s disease. Mediators of inflammation. 2013;2013. More SV, Kumar H, Kim IS, Song S-Y, Choi D-K. Cellular and molecular mediators of neuroinflammation in the pathogenesis of Parkinson’s disease. Mediators of inflammation. 2013;2013.
59.
Zurück zum Zitat Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2018;4:575–90. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2018;4:575–90.
60.
Zurück zum Zitat Lyon MS, Wosiski-Kuhn M, Gillespie R, Caress J, Milligan C, Inflammation. Immunity, and amyotrophic lateral sclerosis: I. etiology and pathology. Muscle Nerve. 2019;59(1):10–22.PubMed Lyon MS, Wosiski-Kuhn M, Gillespie R, Caress J, Milligan C, Inflammation. Immunity, and amyotrophic lateral sclerosis: I. etiology and pathology. Muscle Nerve. 2019;59(1):10–22.PubMed
61.
Zurück zum Zitat Lu C-H, Allen K, Oei F, Leoni E, Kuhle J, Tree T et al. Systemic inflammatory response and neuromuscular involvement in amyotrophic lateral sclerosis. Neurology-Neuroimmunology Neuroinflammation. 2016;3(4). Lu C-H, Allen K, Oei F, Leoni E, Kuhle J, Tree T et al. Systemic inflammatory response and neuromuscular involvement in amyotrophic lateral sclerosis. Neurology-Neuroimmunology Neuroinflammation. 2016;3(4).
62.
Zurück zum Zitat Vivekanantham S, Shah S, Dewji R, Dewji A, Khatri C, Ologunde R. Neuroinflammation in Parkinson’s disease: role in neurodegeneration and tissue repair. Int J Neurosci. 2015;125(10):717–25.PubMed Vivekanantham S, Shah S, Dewji R, Dewji A, Khatri C, Ologunde R. Neuroinflammation in Parkinson’s disease: role in neurodegeneration and tissue repair. Int J Neurosci. 2015;125(10):717–25.PubMed
63.
Zurück zum Zitat Troncoso-Escudero P, Parra A, Nassif M, Vidal RL. Outside in: unraveling the role of Neuroinflammation in the progression of Parkinson’s Disease. Front Neurol. 2018;9:860.PubMedPubMedCentral Troncoso-Escudero P, Parra A, Nassif M, Vidal RL. Outside in: unraveling the role of Neuroinflammation in the progression of Parkinson’s Disease. Front Neurol. 2018;9:860.PubMedPubMedCentral
64.
Zurück zum Zitat Boyd RJ, Avramopoulos D, Jantzie LL, McCallion AS. Neuroinflammation represents a common theme amongst genetic and environmental risk factors for Alzheimer and Parkinson diseases. J Neuroinflamm. 2022;19(1):1–20. Boyd RJ, Avramopoulos D, Jantzie LL, McCallion AS. Neuroinflammation represents a common theme amongst genetic and environmental risk factors for Alzheimer and Parkinson diseases. J Neuroinflamm. 2022;19(1):1–20.
Metadaten
Titel
Neutrophil to lymphocyte ratio in parkinson’s disease: a systematic review and meta-analysis
verfasst von
Samaneh Hosseini
Nasim Shafiabadi
Monireh Khanzadeh
Arshin Ghaedi
Raziyeh Ghorbanzadeh
Amir Azarhomayoun
Aida Bazrgar
Jalil Pezeshki
Hanieh Bazrafshan
Shokoufeh Khanzadeh
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Neurology / Ausgabe 1/2023
Elektronische ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-023-03380-7

Weitere Artikel der Ausgabe 1/2023

BMC Neurology 1/2023 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Delir bei kritisch Kranken – Antipsychotika versus Placebo

16.05.2024 Delir nicht substanzbedingt Nachrichten

Um die Langzeitfolgen eines Delirs bei kritisch Kranken zu mildern, wird vielerorts auf eine Akuttherapie mit Antipsychotika gesetzt. Eine US-amerikanische Forschungsgruppe äußert jetzt erhebliche Vorbehalte gegen dieses Vorgehen. Denn es gibt neue Daten zum Langzeiteffekt von Haloperidol bzw. Ziprasidon versus Placebo.

Schwindelursache: Massagepistole lässt Otholiten tanzen

14.05.2024 Benigner Lagerungsschwindel Nachrichten

Wenn jüngere Menschen über ständig rezidivierenden Lagerungsschwindel klagen, könnte eine Massagepistole der Auslöser sein. In JAMA Otolaryngology warnt ein Team vor der Anwendung hochpotenter Geräte im Bereich des Nackens.

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.