Serum ferritin is a candidate biomarker of disease aggravation in amyotrophic lateral sclerosis

  • Authors:
    • Jixu Yu
    • Nian Wang
    • Faying Qi
    • Xianjun Wang
    • Qiyi Zhu
    • Yucheng Lu
    • Huiling Zhang
    • Fengyuan Che
    • Wei Li
  • View Affiliations

  • Published online on: August 2, 2018     https://doi.org/10.3892/br.2018.1138
  • Pages: 333-338
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease. The mechanism that defines the loss of neurons in ALS is still not clearly understood, and there is no effective therapy to block its progression. Previous studies indicate that a disorder of iron homeostasis exists in ALS and based on this, the change of serum iron and ferritin and the association between iron metabolism and clinical features in Chinese Han patients with ALS was further investigated in the present study, in order to define its pathogenesis. Two cohorts were established: An ALS group consisting of 24 patients and a control group consisting of 38 healthy volunteers. Venous blood samples were collected for serum iron and ferritin analysis. The results indicated that the levels of serum iron were significantly decreased in patients with ALS (P<0.05), while there was no significant difference in serum ferritin between the ALS and control groups. However, the levels of serum ferritin were increased significantly in ALS patients with bulbar-onset (vs. limb-onset in females), dysphagia (vs. without dysphagia), longer disease duration (>12 months vs. ≤12 months in males) and lower ALS Functional Rating Scale-Revised score (<33 vs. ≥33; P<0.05). These results suggested that there was dysregulation of iron metabolism in Chinese Han patients with ALS and that serum ferritin may be a candidate biomarker of aggravation in these patients.

Introduction

Amyotrophic lateral sclerosis (ALS) is a progressive, lethal disease characterized by degeneration of motor neurons in the cortex, brainstem and spinal cord (1,2). ALS is among the most common types of neurodegenerative disorder, with an incidence of approximately 2–3/100,000 individuals in Europe (3,4). The majority of cases of ALS are sporadic, while 5–10% are familial (5,6). With the current therapy (riluzole, 100 mg/day), the typical survival time for patients is 3–5 years following diagnosis (7).

Previous studies have indicated that the loss of neurons in ALS results from a complex interplay among oxidative injury, excitotoxic stimulation, aggregation and/or dysfunction of critical proteins, and genetic factors (812). However, to the best of our knowledge, the mechanism that defines the loss of neurons in ALS is not clearly understood. Further studies have demonstrated the following: A disorder of iron homeostasis exists in ALS (1317); there is increased iron in the spinal cord of patients with ALS (18,19); the iron level in the cerebrospinal fluid of patients with ALS was higher than in control subjects (20,21); and serum ferritin levels were increased in patients with ALS (2226). However, it remains unconfirmed whether a high level of ferritin is specific to ALS. A number of studies have reported that patients with elevated serum ferritin have a shorter survival time (25); however, another study concluded that ferritin levels are unrelated to the survival of patients with ALS (26).

In the present study, the levels of serum iron and ferritin in Chinese Han patients with ALS and control subjects were measured, and the association between iron metabolism and clinical features was studied in order to implicate iron metabolism in Chinese Han patients with ALS.

Materials and methods

Subjects

Two cohorts were established as follows: An ALS group consisting of 24 patients, 14 males and 10 females (42–69 years old; mean, 51.96±7.57 years), all diagnosed with ALS according to El Escorial criteria (27); and a control group consisting of 38 healthy volunteers, 28 males and 10 females (50–62 years old; mean, 53.32±3.28 years), recruited from the local community. Participants who had iron deficiency anemia, acute inflammation, a history of chronic kidney disease, neurologic or psychiatric diseases, or neurologic sequelae induced by brain trauma were excluded. All subjects were of Chinese Han ethnicity. All blood samples were collected for analysis of serum iron and ferritin, and then serum iron and ferritin was compared between those patients with limb-onset and bulbar-onset, with dysphagia and without dysphagia, disease duration ≤12 months and >12 months, and ALS Functional Rating Scale-Revised (ALSFRS-R) score <33 points and ≥33 points. The ALSFRS-R is among the most frequently used scales in the neurological and functional assessment of patients with ALS, and survival time is reportedly shorter in ALS patients with lower ALSFRS-R scores (28). The range of ALSFRS-R scores was 20–42 with a mean 33.04±6.89 in the present study, and therefore patients with an ALSFRS-R score <33 points were separated from those with a score ≥33 points for analysis, as in previous studies (28,29).

Each subject that agreed to enroll in the study reviewed and signed an informed consent form prior to their participation

The study protocol and consent forms were approved by the Medical Ethics Committee of Linyi People's Hospital, Linyi, China.

Measurement of serum iron

Venous blood samples (5 ml) were collected at Linyi People's Hospital (Linyi, China) between 2014 and 2016, and placed upright for 2 h for clotting at room temperature. Then, the samples were centrifuged at 3,000 × g for 15 min, and the supernatant serum was collected for analysis of serum iron and ferritin. Subjects did not have a fever or other signs of inflammation when the blood samples were taken. Iron reagent kit (catalogue no. 467910) was purchased from Beckman Coulter, Inc. and the iron assays were performed on a UniCel DxC 800 Synchron Clinical System (both from Beckman Coulter, Inc., Brea, CA, USA). Iron concentration was measured by an end-point method. In brief, iron is released from transferrin by decomposition of acetic acid and then reduced to ferrous ion by hydroxylamine and sulfhydryl acetate, which combines with zinc-iron by complexation. The proper quantities of samples and reagents were automatically distributed in proportion to a reaction cup by the Synchron System. The proportion of samples to reagents was 1:8. The absorbance at 560 nm was then measured; the change of this absorption is directly proportional to the iron concentration of the samples. Subsequently, the system calculated the iron concentration. All tests were conducted according to the manufacturer's instructions and the standard operating procedures at the Biochemistry Department of Linyi People's Hospital. The reference range of serum iron was 8.1–32.6 µmol/l.

Measurement of serum ferritin

Ferritin detection kit (catalogue no.: 04491785) was purchased from Roche Diagnostics GmbH (Mannheim, Germany). Ferritin was measured on a Roche Cobas E601 Immunology analyzer (Roche Diagnostics GmbH). Serum samples (10 µl) were reacted with the biotin monoclonal specific antibody of ferritin, and the specific monoclonal antibody of ferritin was tagged with the ruthenium complex to form sandwich complexes. Following the addition of particles coated with streptavidin, the complexes combined with them to form a solid phase through interaction between biotin and streptavidin. The reaction mixture was placed in a measuring cell, and particles were adsorbed onto the surface of the electrode by magnetism. Procell/Procell M (catalogue no. 04880340190; Roche Diagnostics GmbH) was used to remove the uncombined substances. Chemiluminescence was produced by applying voltage to the electrode, which was then measured by a photomultiplier. Results were calculated using a calibration curve, which was produced by a two-point calibration and master curve (provided by the reagent barcode). All tests were conducted according to the manufacturer's instructions and standard operating procedures at the Immunology Department of Linyi People's Hospital. The reference ranges of serum ferritin were as follows: 30–400 ng/ml for males and 13–150 ng/ml for females.

Statistical analysis

Results are expressed as the mean ± standard deviation. Statistical analyses were performed using the unpaired t-test and Wilcoxon rank-sum test, depending on the normality of the data. The rank-sum test was used when comparing serum ferritin between male control and ALS patients, and the unpaired Student's t-test was used to analyze all other datasets. Difference was considered to be significant at P<0.05. Statistical analysis was performed using the SAS System for Windows v8 (SAS Institute Inc., Cary, NC, USA).

Results

Patient characteristics

The patients with ALS in the current study included 10 definite ALS and 14 probable ALS diagnoses. A total of 16 patients exhibited signs of limb-onset ALS and 8 patients signs of bulbar-onset ALS. Dysphagia symptoms were apparent in 17 patients (71%) when collecting blood samples. All patients had sporadic ALS. Table I presents the clinical characteristics of the included patients. No significant difference in age was identified between the ALS patients and control subjects (P=0.4129).

Table I.

Clinical characteristics of included ALS patients.

Table I.

Clinical characteristics of included ALS patients.

PatientSexAge, yearsDiagnosisOnset siteDisease duration, monthsALSFRS-R score
  1Male52DefiniteBulbar-onset3625
  2Male47ProbableLimb-onset1237
  3Male69ProbableLimb-onset3023
  4Male45DefiniteLimb-onset2431
  5Male50ProbableBulbar-onset1639
  6Male42ProbableLimb-onset2422
  7Male47ProbableLimb-onset2435
  8Male62DefiniteLimb-onset1241
  9Male48DefiniteLimb-onset1840
10Male55ProbableLimb-onset1236
11Male48ProbableLimb-onset2436
12Male43ProbableLimb-onset742
13Male52ProbableBulbar-onset637
14Male68DefiniteBulbar-onset1239
15Female48ProbableLimb-onset3031
16Female64DefiniteLimb-onset1020
17Female46DefiniteLimb-onset1225
18Female49ProbableBulbar-onset3022
19Female46DefiniteBulbar-onset2430
20Female53DefiniteBulbar-onset2431
21Female56ProbableLimb-onset2835
22Female45ProbableLimb-onset1236
23Female54ProbableBulbar-onset1239
24Female58DefiniteLimb-onset1741

[i] ALS, amyotrophic lateral sclerosis; ALSFRS-R, Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised.

Serum iron and ferritin in patients with ALS and the control group

As illustrated in Table II, the serum iron of the ALS group was significantly lower than that of the control group (P=0.0074), while there was no significant difference in serum ferritin between the groups (male, P=0.3504; female, P=0.1068; Table III). However, the mean levels of serum ferritin in the ALS group were higher than those in the control group for both male and female patients; therefore, it was indicated that serum ferritin was increased in patients with ALS.

Table II.

Serum iron of ALS patients and controls.

Table II.

Serum iron of ALS patients and controls.

GroupNumber of subjectsSerum iron, µmol/lP-value
Control3819.44±6.470.0074
ALS patients2414.95±5.77

[i] ALS, amyotrophic lateral sclerosis.

Table III.

Serum ferritin of ALS patients and controls.

Table III.

Serum ferritin of ALS patients and controls.

GroupNumber of subjectsSerum ferritin, ng/mlP-value
Male control28171.35±73.640.3504
Male ALS patients14221.95±138.20
Female control1093.67±65.880.1068
Female ALS patients10157.86±99.76

[i] ALS, amyotrophic lateral sclerosis.

Serum iron and ferritin in patients with limb-onset and bulbar-onset ALS

When comparing the serum iron and ferritin levels between patients with limb-onset and bulbar-onset ALS, no significant difference in serum iron was identified (P=0.0586; Table IV). There was also no significant difference in the levels of serum ferritin between male patients with limb-onset and bulbar-onset ALS (P=0.2257; Table V); however, the serum ferritin levels of female patients with bulbar-onset ALS were significantly higher than those of female patients with limb-onset ALS (P=0.008; Table VI).

Table IV.

Serum iron of ALS patients.

Table IV.

Serum iron of ALS patients.

Disease characteristicNumber of subjectsSerum iron, µmol/lP-value
Limb-onset1613.39±4.910.0586
Bulbar-onset818.08±6.40
Without dysphagia714.39±4.300.7659
With dysphagia1715.18±6.38
Disease duration ≤12 months1016.40±8.340.3852
Disease duration >12 months1413.91±2.81
ALSFRS-R score <331011.69±4.110.0155
ALSFRS-R score ≥331417.28±5.76

[i] ALS, amyotrophic lateral sclerosis; ALSFRS-R, Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised.

Table V.

Serum ferritin of male ALS patients.

Table V.

Serum ferritin of male ALS patients.

Disease characteristicNumber of subjectsSerum ferritin, ng/mlP-value
Limb-onset10251.08±148.710.2257
Bulbar-onset4149.12±80.75
Without dysphagia4125.68±33.960.0195
With dysphagia10260.46±146.41
Disease duration ≤12 months6142.46±58.430.0422
Disease duration >12 months8281.57±153.47
ALSFRS-R score <334344.70±152.140.0287
ALSFRS-R score ≥3310172.85±102.45

[i] ALS, amyotrophic lateral sclerosis; ALSFRS-R, Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised.

Table VI.

Serum ferritin of female ALS patients.

Table VI.

Serum ferritin of female ALS patients.

Disease characteristicNumber of subjectsSerum ferritin, ng/mlP-value
Limb-onset697.71±59.420.0080
Bulbar-onset4248.08±76.69
Without dysphagia361.63±43.890.0357
With dysphagia7199.10±87.59
Disease duration ≤12 months4113.40±73.930.2739
Disease duration >12 months6187.50±109.54
ALSFRS-R score <336207.11±93.090.0474
ALSFRS-R score ≥33483.98±57.28

[i] ALS, amyotrophic lateral sclerosis; ALSFRS-R, Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised.

Serum iron and ferritin in ALS patients with and without dysphagia

On comparing the serum iron and ferritin between ALS patients with and without dysphagia, no significant difference in serum iron was observed (P=0.7659; Table IV). However, the serum ferritin of ALS patients with dysphagia was significantly higher than in those without dysphagia (male, P=0.0195; female, P=0.0357; Tables V and VI).

Serum iron and ferritin in patients with ALS of different disease duration

The serum iron and ferritin in patients with ALS of different disease durations (≤12 months and >12 months) were subsequently compared. No significant difference in serum iron in ALS cases of different disease duration was determined (P=0.3852; Table IV). Serum ferritin was increased significantly in male ALS cases with longer disease duration (P=0.0422; Table V); however, there was no significant difference in serum ferritin in female cases of ALS of different disease duration (P=0.2739; Table VI).

Serum iron and ferritin in ALS patients with different ALSFRS-R scores

By comparing the serum iron and ferritin of ALS patients with different ALSFRS-R scores, it was identified that the levels of serum iron in ALS patients with ALSFRS-R scores <33 points were lower compared with in those patients with ALSFRS-R scores ≥33 points (P=0.0155; Table IV). Additionally, the levels of serum ferritin were increased significantly in ALS patients with ALSFRS-R scores <33 points compared with in those patients with ALSFRS-R scores ≥33 points (male, P=0.0287; female, P=0.0474, Tables V and VI).

Discussion

Iron is established to potentiate the toxic effects of reactive oxygen species by catalyzing the formation of highly reactive hydroxyl radicals from hydrogen peroxide through Fenton chemistry (30,31). Thus, while iron is essential for normal physiology (32), it is also implicated in many pathological processes, including neuron degenerative disorders (33,34). Iron accelerates β-amyloid aggregation and promotes oxidative damage in Alzheimer's disease (3537). A number of studies have indicated that iron is increased in both models of ALS and humans with ALS (3841). Even though it is unclear whether oxidative stress is a primary or secondary cause of neurodegeneration in ALS, previous study has suggested that it is a major contributory factor that leads to chronic motor neuron death (42). Increased iron levels are possibly involved in oxidative damage through the Fenton reaction in ALS, and iron chelation therapy may have potential in the prevention and treatment of ALS (43,44).

Symptoms of dysphagia, difficulty with self-feeding and fatigue with meals often afflict patients with ALS, making them susceptible to a high risk of malnutrition (45). Dysphagia affects up to 80% of patients with ALS (46), and thus enteral nutrition (EN) and parenteral nutrition (PN) are important for patients with ALS (45). The recommended daily intake of iron for healthy individuals is 10 mg/day, but the iron content of polymeric enteral formulas that are usually employed to meet the nutritional and metabolic needs of patients with ALS ranges from 13 to 24 mg/l (47). Many patients with ALS receive EN, which may exacerbate iron overload. Although previous studies do not report many patients receiving EN, iron overload exacerbated by EN may serve a role in the reported results (26).

In the present study, no significant difference between the ALS and control subjects was identified regarding serum ferritin, which is in contrast to the results of previous studies (2226); however, the mean levels of serum ferritin in the ALS group were higher than those in the control group. The levels of serum iron in patients with ALS were significantly lower than those in the control group. The reason for this may be attributed to a number of external factors. Environmental factors, including diet, can affect iron homeostasis in the body. The majority of patients with ALS that were enrolled in the present study lived in their own home and could not get help from health workers, or EN or PN treatment, which could result in malnutrition that further influenced their levels of serum iron. By contrast, the Caucasian patients assessed previously lived in developed countries (26), and likely had greater access to treatments and adequate nutrition compared with patients in the present study, which may explain the significant increase in serum ferritin reported (26). Our previous study indicated that iron levels are increased in the motor cortex of patients with ALS by using susceptibility weighted imaging (48). The majority of the patients with ALS in the present study also participated in our aforementioned previous study. Therefore, it may be concluded that the decrease in serum iron levels does not affect the iron levels of the motor cortex in Chinese Han patients with ALS.

Further investigations were conducted in the present study to clarify the effect of onset site, dysphagia, disease duration and ALSFRS-R scores. The results showed that onset site, dysphagia and disease duration did not affect serum iron in patients with ALS. The levels of serum ferritin were increased significantly in male ALS patients with dysphagia, longer disease duration (>12 months) and lower ALSFRS-R scores (<33 points). Furthermore, the levels of serum ferritin were increased significantly in female ALS patients with bulbar-onset, dysphagia and lower ALSFRS-R scores. Intracellular ferritin may be upregulated in response to oxidative stress, and serum ferritin may be increased in patients with ALS due to oxidative stress (25). The results of the present study indicate that serum ferritin is a marker of aggravation in ALS. However, a limitation of this study was the relatively small sample group, and thus data from more patients should be collected for further studies; a further aim is to assess other molecules related to ALS in future study.

In conclusion, serum iron levels of patients with ALS were decreased significantly in the present study, while serum ferritin exhibited an increasing tendency. Furthermore, the levels of serum ferritin were increased significantly in ALS patients with bulbar-onset, dysphagia, longer disease duration and lower ALSFRS-R scores. Collectively the results indicate that dysregulation of iron metabolism exists in Chinese Han patients with ALS, and that serum ferritin may be a candidate biomarker of aggravation in ALS.

Acknowledgements

Not applicable.

Funding

The present study was supported in part by grants from the Shandong Provincial Natural Science Foundation, China (grant no. ZR2010HM041) and the Shandong Provincial Postdoctoral Innovation Foundation, China (grant no. 201102004).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JY contributed to study design, to analysis and interpretation of the data, and to writing and revision of the manuscript. NW, YL and HZ performed the tests of serum iron and ferritin. FQ, XW and QZ prepared the blood samples and aided with the testing of serum iron and ferritin. FC and WL analyzed and interpreted the data, and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study protocol was approved by the Medical Ethics Committee of Linyi People's Hospital (Linyi, China). Each subject that agreed to enroll in the study reviewed and signed an informed consent form prior to their participation.

Consent for publication

Written informed consent obtained from all subjects permitted the publication of their data for research purposes following anonymization.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kiernan MC, Vucic S, Cheah BC, Turner MR, Eisen A, Hardiman O, Burrell JR and Zoing MC: Amyotrophic lateral sclerosis. Lancet. 377:942–955. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Hardiman O, van den Berg LH and Kiernan MC: Clinical diagnosis and management of amyotrophic lateral sclerosis. Nat Rev Neurol. 7:639–649. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Logroscino G, Traynor BJ, Hardiman O, Chiò A, Mitchell D, Swingler RJ, Millul A, Benn E and Beghi E: EURALS: Incidence of amyotrophic lateral sclerosis in Europe. J Neurol Neurosurg Psychiatry. 81:385–390. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Couratier P, Corcia P, Lautrette G, Nicol M, Preux PM and Marin B: Epidemiology of amyotrophic lateral sclerosis: A review of literature. Rev Neurol (Paris). 172:37–45. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Byrne S, Walsh C, Lynch C, Bede P, Elamin M, Kenna K, McLaughlin R and Hardiman O: Rate of familial amyotrophic lateral sclerosis: A systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 82:623–627. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, Pagani W, Lodin D, Orozco G and Chinea A: A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int. 6:1712015. View Article : Google Scholar : PubMed/NCBI

7 

Holecek V and Rokyta R: Possible etiology and treatment of amyotrophic lateral sclerosis. Neuro Endocrinol Lett. 38:528–531. 2018.PubMed/NCBI

8 

Redler RL and Dokholyan NV: The complex molecular biology of amyotrophic lateral sclerosis (ALS). Prog Mol Biol Transl Sci. 107:215–262. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Cookson MR and Shaw PJ: Oxidative stress and motor neurone disease. Brain Pathol. 9:165–186. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Kim K, Lee SG, Kegelman TP, Su ZZ, Das SK, Dash R, Dasgupta S, Barral PM, Hedvat M, Diaz P, et al: Role of excitatory amino acid transporter-2 (EAAT2) and glutamate in neurodegeneration: Opportunities for developing novel therapeutics. J Cell Physiol. 226:2484–2493. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Taylor JP, Hardy J and Fischbeck KH: Toxic proteins in neurodegenerative disease. Science. 296:1991–1995. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O'Regan JP, Deng HX, et al: Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 362:59–62. 1993. View Article : Google Scholar : PubMed/NCBI

13 

Jeong SY, Rathore KI, Schulz K, Ponka P, Arosio P and David S: Dysregulation of iron homeostasis in the CNS contributes to disease progression in a mouse model of amyotrophic lateral sclerosis. J Neurosci. 29:610–619. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Oshiro S, Morioka MS and Kikuchi M: Dysregulation of iron metabolism in Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Adv Pharmacol Sci. 2011:3782782011.PubMed/NCBI

15 

Hadzhieva M, Kirches E, Wilisch-Neumann A, Pachow D, Wallesch M, Schoenfeld P, Paege I, Vielhaber S, Petri S, Keilhoff G, et al: Dysregulation of iron protein expression in the G93A model of amyotrophic lateral sclerosis. Neuroscience. 230:94–101. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Chen P, Miah MR and Aschner M: Metals and Neurodegeneration. F1000Res. Mar 17–2016.doi: 10.12688/f1000research.7431.1. View Article : Google Scholar

17 

Veyrat-Durebex C, Corcia P, Mucha A, Benzimra S, Mallet C, Gendrot C, Moreau C, Devos D, Piver E, Pagès JC, et al: Iron metabolism disturbance in a French cohort of ALS patients. BioMed Res Int. 2014:4857232014. View Article : Google Scholar : PubMed/NCBI

18 

Kasarskis EJ, Tandon L, Lovell MA and Ehmann WD: Aluminum, calcium, and iron in the spinal cord of patients with sporadic amyotrophic lateral sclerosis using laser microprobe mass spectroscopy: A preliminary study. J Neurol Sci. 130:203–208. 1995. View Article : Google Scholar : PubMed/NCBI

19 

Ince PG, Shaw PJ, Candy JM, Mantle D, Tandon L, Ehmann WD and Markesbery WR: Iron, selenium and glutathione peroxidase activity are elevated in sporadic motor neuron disease. Neurosci Lett. 182:87–90. 1994. View Article : Google Scholar : PubMed/NCBI

20 

Hozumi I, Hasegawa T, Honda A, Ozawa K, Hayashi Y, Hashimoto K, Yamada M, Koumura A, Sakurai T, Kimura A, et al: Patterns of levels of biological metals in CSF differ among neurodegenerative diseases. J Neurol Sci. 303:95–99. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Zheng Y, Gao L, Wang D and Zang D: Elevated levels of ferritin in the cerebrospinal fluid of amyotrophic lateral sclerosis patients. Acta Neurol Scand. 136:145–150. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Qureshi M, Brown RH Jr, Rogers JT and Cudkowicz ME: Serum ferritin and metal levels as risk factors for amyotrophic lateral sclerosis. Open Neurol J. 2:51–54. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Goodall EF, Haque MS and Morrison KE: Increased serum ferritin levels in amyotrophic lateral sclerosis (ALS) patients. J Neurol. 255:1652–1656. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Ikeda K, Hirayama T, Takazawa T, Kawabe K and Iwasaki Y: Relationships between disease progression and serum levels of lipid, urate, creatinine and ferritin in Japanese patients with amyotrophic lateral sclerosis: A cross-sectional study. Intern Med. 51:1501–1508. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Nadjar Y, Gordon P, Corcia P, Bensimon G, Pieroni L, Meininger V and Salachas F: Elevated serum ferritin is associated with reduced survival in amyotrophic lateral sclerosis. PLoS One. 7:e450342012. View Article : Google Scholar : PubMed/NCBI

26 

Su XW, Clardy SL, Stephens HE, Simmons Z and Connor JR: Serum ferritin is elevated in amyotrophic lateral sclerosis patients. Amyotroph Lateral Scler Frontotemporal Degener. 16:102–107. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Brooks BR: El Escorial World Federation of Neurology criteria for the diagnosis of amyotrophic lateral sclerosis. Subcommittee on Motor Neuron Diseases/Amyotrophic Lateral Sclerosis of the World Federation of Neurology Research Group on Neuromuscular Diseases and the El Escorial ‘Clinical limits of amyotrophic lateral sclerosis’ workshop contributors. J Neurol Sci. 124:96–107. 1994. View Article : Google Scholar : PubMed/NCBI

28 

Kollewe K, Mauss U, Krampfl K, Petri S, Dengler R and Mohammadi B: ALSFRS-R score and its ratio: A useful predictor for ALS-progression. J Neurol Sci. 275:69–73. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Lechtzin N, Maragakis NJ, Kimball R, Busse A, Hoffman V and Clawson L: Accurate ALSFRS-R scores can be generated from retrospective review of clinic notes. Amyotroph Lateral Scler. 10:244–247. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Contestabile A: Amyotrophic lateral sclerosis: From research to therapeutic attempts and therapeutic perspectives. Curr Med Chem. 18:5655–5665. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Jomova K, Vondrakova D, Lawson M and Valko M: Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem. 345:91–104. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Boldt DH: New perspectives on iron: An introduction. Am J Med Sci. 318:207–212. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Qian ZM and Wang Q: Expression of iron transport proteins and excessive iron accumulation in the brain in neurodegenerative disorders. Brain Res Brain Res Rev. 27:257–267. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Rivera-Mancía S, Pérez-Neri I, Ríos C, Tristán-López L, Rivera-Espinosa L and Montes S: The transition metals copper and iron in neurodegenerative diseases. Chem Biol Interact. 186:184–199. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Casadesus G, Smith MA, Zhu X, Aliev G, Cash AD, Honda K, Petersen RB and Perry G: Alzheimer disease: Evidence for a central pathogenic role of iron-mediated reactive oxygen species. J Alzheimers Dis. 6:165–169. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Castellani RJ, Moreira PI, Liu G, Dobson J, Perry G, Smith MA and Zhu X: Iron: The Redox-active center of oxidative stress in Alzheimer disease. Neurochem Res. 32:1640–1645. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Smith MA, Harris PL, Sayre LM and Perry G: Iron accumulation in Alzheimer disease is a source of redox-generated free radicals. Proc Natl Acad Sci USA. 94:9866–9868. 1997. View Article : Google Scholar : PubMed/NCBI

38 

Olsen MK, Roberds SL, Ellerbrock BR, Fleck TJ, McKinley DK and Gurney ME: Disease mechanisms revealed by transcription profiling in SOD1-G93A transgenic mouse spinal cord. Ann Neurol. 50:730–740. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Yu J, Guo Y, Sun M, Li B, Zhang Y and Li C: Iron is a potential key mediator of glutamate excitotoxicity in spinal cord motor neurons. Brain Res. 1257:102–107. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Langkammer C, Enzinger C, Quasthoff S, Grafenauer P, Soellinger M, Fazekas F and Ropele S: Mapping of iron deposition in conjunction with assessment of nerve fiber tract integrity in amyotrophic lateral sclerosis. J Magn Reson Imaging. 31:1339–1345. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Mitchell RM, Simmons Z, Beard JL, Stephens HE and Connor JR: Plasma biomarkers associated with ALS and their relationship to iron homeostasis. Muscle Nerve. 42:95–103. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Petri S, Körner S and Kiaei M: Nrf2/ARE signaling pathway: Key mediator in oxidative stress and potential therapeutic target in ALS. Neurol Res Int. 2012:8780302012. View Article : Google Scholar : PubMed/NCBI

43 

Wang Q, Zhang X, Chen S, Zhang X, Zhang S, Youdium M and Le W: Prevention of motor neuron degeneration by novel iron chelators in SOD1(G93A) transgenic mice of amyotrophic lateral sclerosis. Neurodegener Dis. 8:310–321. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Golko-Perez S, Mandel S, Amit T, Kupershmidt L, Youdim MB and Weinreb O: Additive Neuroprotective effects of the multifunctional iron chelator M30 with enriched diet in a mouse model of amyotrophic lateral sclerosis. Neurotox Res. 29:208–217. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Greenwood DI: Nutrition management of amyotrophic lateral sclerosis. Nutr Clin Pract. 28:392–399. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Traynor BJ, Codd MB, Corr B, Forde C, Frost E and Hardiman OM: Clinical features of amyotrophic lateral sclerosis according to the El Escorial and Airlie House diagnostic criteria: A population-based study. Arch Neurol. 57:1171–1176. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Molfino A, Kushta I, Tommasi V, Fanelli Rossi F and Muscaritoli M: Amyotrophic lateral sclerosis, enteral nutrition and the risk of iron overload. J Neurol. 256:1015–1016. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Yu J, Qi F, Wang N, Gao P, Dai S, Lu Y, Su Q, Du Y and Che F: Increased iron level in motor cortex of amyotrophic lateral sclerosis patients: An in vivo MR study. Amyotroph Lateral Scler Frontotemporal Degener. 15:357–361. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 9 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu J, Wang N, Qi F, Wang X, Zhu Q, Lu Y, Zhang H, Che F and Li W: Serum ferritin is a candidate biomarker of disease aggravation in amyotrophic lateral sclerosis. Biomed Rep 9: 333-338, 2018
APA
Yu, J., Wang, N., Qi, F., Wang, X., Zhu, Q., Lu, Y. ... Li, W. (2018). Serum ferritin is a candidate biomarker of disease aggravation in amyotrophic lateral sclerosis. Biomedical Reports, 9, 333-338. https://doi.org/10.3892/br.2018.1138
MLA
Yu, J., Wang, N., Qi, F., Wang, X., Zhu, Q., Lu, Y., Zhang, H., Che, F., Li, W."Serum ferritin is a candidate biomarker of disease aggravation in amyotrophic lateral sclerosis". Biomedical Reports 9.4 (2018): 333-338.
Chicago
Yu, J., Wang, N., Qi, F., Wang, X., Zhu, Q., Lu, Y., Zhang, H., Che, F., Li, W."Serum ferritin is a candidate biomarker of disease aggravation in amyotrophic lateral sclerosis". Biomedical Reports 9, no. 4 (2018): 333-338. https://doi.org/10.3892/br.2018.1138