Skip to main content
Erschienen in: Pediatric Rheumatology 1/2023

Open Access 01.12.2023 | Research Article

Juvenile dermatomyositis: association between nail fold capillary end row loop– area under the curve– and disease damage indicators

verfasst von: Amer Khojah, Gabrielle Morgan, Marisa S. Klein-Gitelman, Lauren M. Pachman

Erschienen in: Pediatric Rheumatology | Ausgabe 1/2023

Abstract

Background

Juvenile Dermatomyositis (JDM) is a rare autoimmune disease characterized by skin and muscle inflammation. The loss of nail fold capillary end row loops (ERL) is evidence of small vessel involvement in JDM. This study aimed to examine the specific association of ERL over the disease course with evidence of JDM disease damage.

Methods

We analyzed data from 68 initially treatment-naïve JDM children who had been observed for at least five years with multiple ERL density assessments. The JDM disease course were categorized into monocyclic short, monocyclic long, polycyclic, and chronic. The ERL capillary count was cumulatively evaluated using the area under the curve (AUC) method.

Results

The mean ERL density for the treatment-naive JDM was significantly lower than that of their healthy age-matched controls (4.8 ± 1.6 /mm vs. 7.9 ± 0.9 /mm; p < 0.0001). The ERL AUC was significantly lower in children with a chronic disease course compared to those with a monocyclic short (p = 0.001) or monocyclic long disease course (p = 0.013). JDM patients with lipodystrophy had lower ERL AUC than those without lipodystrophy (p = 0.04). There was no association between ERL AUC and calcifications or fractures.

Conclusion

Persistently decreased ERL capillary density, reflected by low ERL AUC, is associated with a chronic disease course and lipodystrophy in JDM. Despite medical therapy, the mean ERL count remained below normal even after five years, particularly in polycyclic and chronic cases. It is not clear that restoring normal capillary density is currently feasible in children with JDM.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12969-023-00919-3.

Publisher’s Note

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

Introduction

Juvenile Dermatomyositis (JDM) is a systemic pediatric autoimmune disease characterized by skin and muscle inflammation [1]. Although it is the most common pediatric inflammatory myopathy, with an incidence rate of approximately 3.2 cases per million children in the United States, JDM remains a rare disease [2]. Its etiology is not entirely understood, but it is believed to involve a combination of specific genetic predisposition and environmental factors, such as viral infections and exposure to ultraviolet rays [3, 4].
One of the key features of JDM is the loss of nailfold capillary end row loops (ERL), which can be evaluated at the bedside using capillaroscopy [57]. The loss of capillary ERL indicates small vessel vascular injury, which is further supported by elevated levels of von Willebrand factor antigen (vWF antigen) in the treatment-naïve JDM patients [8]. The reduction in ERL is linked to lower bioavailability of oral prednisone compared to IV methylprednisolone making the oral route of medical therapy less effective [9]. Inadequate treatment of JDM can lead to a higher risk of complications such as calcinosis, deposition of insoluble calcium salts in the skin and subcutaneous tissue, and lipodystrophy [10, 11]. Therefore, defining the association between ERL density and disease progression is important for patient care.
This study aims to examine the association between ERL density over time (5 years) using the area under the curve (AUC) method to define the range of various diseases courses (monocyclic short, monocyclic long, polycyclic, and chronic) in addition to indicators of disease damage (lipodystrophy and calcification).

Methods

Subjects

This retrospective chart review study (IRB# 2012–14,858) was conducted at Ann & Robert H. Lurie Children’s Hospital of Chicago. We included all subjects with the JDM diagnosis based on Bohan and Peter criteria [12, 13] who had at least five years of follow-up data and had at least four ERL assessments at a prespecified time point (0,6,12,24,36,48, and 60 months from initiation of medical therapy). Patients who received medical therapy before the initial ERL assessments, those lacking a five-year follow-up, or those with overlap syndrome were excluded from the analysis. The JDM disease activity was evaluated using standardized scoring systems—the Disease Activity Score (DAS) [14] and Childhood Myositis Assessment Scale (CMAS) [15]. The demographic data of the JDM children are presented in Table 1.
Table 1
Demographic and disease characteristics of the JDM cohort
 
Frequency (n)
Percentage
Sample size
68
 
Sex
  
 Female
57
16.2%
 Male
11
83.8%
Race/Ethnicity
  
 White
51
75.0%
 Hispanic
13
19.1%
 African American
2
2.9%
 Others
2
2.9%
Myositis specific antibodies
  
 P155/140
28
41.2%
 MJ
2
2.9%
 Mi2
5
7.4%
 MDA5
2
2.9%
 Others or multiple MSAs
5
7.4%
 Negative
21
30.8%
 Not done
5
7.4%
Treatment
  
 Oral steroid
68
100%
 Intravenous steroid
64
94.1%
 Methotrexate
63
92.6%
 Intravenous immunoglobulin
8
11.8%
 Hydroxychloroquine
41
60.3%
 Cyclosporine
12
17.6%
 Mycophenolate
26
38.2%
Disease course
  
 Monocyclic short
12
17.6%
 Monocyclic long
29
42.6%
 Polycyclic
14
20.6%
 Chronic
13
19.1%
The study included 77 healthy children as a control group after providing appropriate written consent (IRB# 2001–11,715). These healthy controls did not have any autoimmune disease or active infection at time of their enrollment. In appreciation for their participation in nailfold capillaroscopy, they received a $25 gift card. Demographic details of healthy controls are available in the supplementary materials (Supplemental Table 1).

Disease course

Children with JDM were categorized into four distinct disease courses according to their treatment response: (A) monocyclic short: if the child completed therapy within the first 36 months without a subsequent disease flare; (B) monocyclic long: if the child completed therapy after 36 months without a subsequent disease flare; (C) polycyclic: if the child had completed treatment but had a subsequent relapse of disease requiring re-initiation of medication; (D) chronic: no clinical resolution within 60 months.

Nailfold capillary ERL studies

Standardized images of the nailfold area were obtained using a Nikon Coolpix p6000 digital camera equipped with a Dermlite2 ProHR (18x). The analysis of the nailfold images was performed by a single experienced observer (GM) using Photoshop. The number of ERL per 3 mm section on each of the eight fingers (excluding thumbs) was counted and subsequently divided by three. Each patient’s mean ERL/mm was calculated by averaging the ERL/mm of the eight fingers [5, 16].

ERL area under the curve calculation

GraphPad Prism was used to calculate the AUC to measure the ERL cumulatively across the study duration. First, the curve was created by plotting the ERL data over time. Then, Prism divides AUC into multiple small trapezoid areas, which are measured individually, using the trapezoid rule [area = ½ (base a + base b) x height], and added up to get the total AUC (Fig. 1).

Statistical analysis

The Person’s correlation coefficient was utilized to assess the relationship between ERL at diagnosis and ERL AUC. The student t-test was used to compare the mean ERL AUC of subjects with and without signs of disease damage. Statistical analyses were conducted using IBM SPSS Statistics® and GraphPad Prism® version 9.4.1 was utilized to generate the figures.

Results

The study included 68 treatment-naive JDM children, the majority of whom were female (84%). The racial and ethnic distribution was as follows: 75% Caucasian, 19% Hispanic, 3% African American and 3% Others. Their MSAs (Myositis-specific antibodies) were as follows: 41% P155/140+, 3% MJ+, 7.5% Mi-2+, 3% MDA-5+, 7.5% multiple MSAs, and 31% MSA negative (Table 1). The mean age of onset for JDM was 6 ± 3.1 years, and the mean duration of untreated disease was 9.6 ± 10.2 months (Table 2). The initial disease activity scores were 11.0 ± 3.6 for DAS total, 5.9 ± 1.5 for DAS skin, and 5.1 ± 1.5 for DAS muscle, and CMAS score was 37 ± 10.3 (Table 2). The disease course distribution was as follows: 17.6% monocyclic short, 42.6% monocyclic long, 20.6% polycyclic, and 19.1% chronic (Table 1).
Table 2
Baseline (before treatment) disease activity assessment of the JDM cohort
 
Reference Range
Mean ± SD
Median (range)
Age (years)
 
6 ± 3.1
5.4 (1.9–16.4)
Duration of untreated disease (months)
 
9.6 ± 10.2
6.5 (1–73)
Clinical disease activity indicator
   
 DAS-total
0
11 ± 3.6
11.5 (3–19)
 DAS-skin
0
5.9 ± 1.5
6 (2–9)
 DAS-muscle
0
5.1 ± 1.5
5 (0–10)
 CMAS
52
37 ± 10.3
38 (12–52)
 ERL (#/mm)
> 7
5 ± 3.1
4.9 (2.3–10.3)
Laboratory disease activity indicators
   
 Neopterin (nmol/L)
< 10
19.4 ± 10.6
18.5 (2.4–49.3)
ESR (mm/hr)
< 20
19 ± 13.4
15 (3–65)
 vWF Antigen
 
156 ± 75
140 (52–374)
Muscle enzymes
   
 CK (IU/L)
26–27
1700 ± 5291
140 (57-35471)
 AST (IU/L)
17–96
107 ± 162.5
46 (22–890)
 LDH (IU/L)
147–463
455 ± 392.6
342 (166–2259)
 Aldolase (U/L)
3.4–8.6
21.6 ± 41.6
9.9 (2.8–237)
Flow cytometry
   
 Total T cells (CD3+)
 
64 ± 8.3
64 (41–86)
 T helper cells (CD3 + CD4+)
 
44.4 ± 8
45 (26–64)
 T cytotoxic cells (CD3 + CD8+)
 
18.6 ± 4.4
19 (6–31)
 B cells (CD19+)
 
29 ± 8.3
29 (12–52)
 NK cells (CD16+/CD56+)
 
6.3 ± 3.6
5 (1–15)
The mean ERL count for treatment-naive JDM was 4.8 ± 1.6/mm, which is significantly lower than the healthy control, 7.9 ± 0.9/mm (p < 0.0001). Despite the improvement in mean ERL count over time, it still remained below the expected normal level obtained in healthy controls (6.1–9.7/mm), even after five years of medical therapy (Fig. 2). The rate of improvement varied depending on the different disease courses, with the monocyclic short disease course showing more change than the other groups (4.8 ± 1.5/mm at baseline vs. 6.7 ± 1.5/mm at 12 months, p = 0.038 paired T-test).
To evaluate the accumulative effects of chronic ERL capillary loss, the ERL AUC was calculated for each patient (Fig. 1). Although there was a positive correlation between the initial ERL count and ERL AUC, the correlation was not strong (r2 = 0.18, p = 0.001). There was a significant difference between the ERL AUC for monocyclic short vs. chronic (389 ± 46.46 vs. 313 ± 46.69, p = 0.001) and monocyclic long vs. chronic (359 ± 44.53 vs. 313 ± 46.69, p = 0.013) (Fig. 3a). Next, the relationship between ERL AUC and indicators of disease damage (lipodystrophy and calcification) was evaluated. JDM patients with lipodystrophy exhibited a lower ERL AUC than those without lipodystrophy (335.7 ± 52.52 vs. 363.0 ± 47.13, p = 0.04) (Fig. 3b). However, the ERL AUC had no significant association with calcifications (Fig. 3c). Lastly, the relationship between ERL AUC and fractures was assessed, revealing no significant correlation (Fig. 3d).
Next, we evaluated the effect of medical treatment on ERL AUC. There was a negative correlation between the duration of oral steroid use and ERL AUC (r2 = 0.12, p = 0.004). Patients who required multiple immunosuppressive medications (additional immunosuppression more than steroid, hydroxychloroquine, and methotrexate) tended to have a lower ERL AUC (339.9 ± 46.8 vs. 371.0 ± 51.0, p = 0.01) (Fig. 4).

Discussion

This study provides insight into the association between endothelial dysfunction demonstrated by decreased ERL count and disease progression in children with JDM. Consistent with previous studies, we observed a significantly lower mean ERL density in untreated JDM children compared to healthy controls [6, 7]. This finding supports the concept of endothelial involvement in JDM pathophysiology [3]. Furthermore, the reduction in ERL count often correlates with the severity of skin disease and/or muscle weakness [6, 17], suggesting its possible role as a potential physical exam indicator ofr disease activity. Few studies have evaluated the changes in capillary density and disease activity longitudinally over the disease course [17, 18]. We observed variations in the rate of improvement in ERL count in the different JDM disease courses. Monocyclic disease courses show improvement of the ERL capillary count at a faster rate than that of chronic disease. This finding suggests that the rate of improvement in ERL count may be a more critical factor than the initial ERL count in predicting disease course and outcome.
Despite medical therapy, the mean ERL count in JDM patients remained below normal levels even after five years of treatment, particularly in the polyphasic and chronic disease courses. This implies that the restoration of capillary density might be challenging to achieve and may require additional novel therapeutic strategies to target endothelial dysfunction effectively [19].
Additionally, we utilized the AUC method to evaluate the cumulative change in ERL capillary density over the study period. Our results demonstrated a correlation between ERL AUC and a more chronic disease course, as well as the presence of complications such as lipodystrophy. Of note, we found a weakly positive correlation between the initial ERL count and ERL AUC, which suggests that factors other than the initial capillary density, including MSAs, may contribute to the cumulative capillary loss. For example, it has been shown JDM with anti-P155/140 antibody tend to have lower ERL capillary count and are less likely to have a monophasic disease course [5]. However, our study was not powered enough to investigate the impact of the type or the duration of different MSAs on ERL AUC.
Circulating endothelial cells and markers of endothelial injury, such as vWF antigen, and thrombomodulin are elevated in JDM, providing further evidence of endothelial involvement in the disease pathophysiology [8, 19, 20]. B cell activation and expansion as well as the formation of anti-endothelial cell antibodies have been demonstrated in JDM [2124], suggesting other potential mechanisms for endothelial cell injury. Soluble adhesion molecule markers, such as ICAM-1, ICAM-3, and VCAM1, and inflammatory cytokine and markers like neopterin have been used as possible biomarkers of vasculopathy in JDM [2527]. These findings highlight the complex interplay between immune dysregulation and endothelial dysfunction in JDM, requiring further investigation to elucidate the underlying mechanisms. Furthermore, it is important to recognize the clinical implications of microvascular injury in JDM as it can affect the gastrointestinal system [28]. The reduced nailfold capillary density observed in JDM has been associated with impaired absorption of oral prednisone, potentially leading to suboptimal drug levels and ineffective treatment [9]. Therefore, administering medications by the intravenous or subcutaneous routes might be preferred in patients with persistently low ERL counts.
The study has several limitations. First, the sample size is relatively small, particularly when considering the potential heterogeneity within the JDM population. Second, the diagnosis of lipodystrophy was based on physician assessment, which can introduce some subjectivity. Lastly, the study was conducted at a single center, potentially limiting the generalizability, especially in geographic regions where P155/140 autoantibodies are not the predominant autoantibody.

Conclusions

Persistently decreased ERL capillary density documented by low ERL AUC is associated with both a chronic disease course and lipodystrophy in JDM. Despite medical therapy, the mean ERL count remained below normal, even after five years, particularly in polycyclic and chronic cases. Therefore, restoring normal ERL capillary density might be challenging and require novel therapeutic strategies targeting endothelial dysfunction.

Acknowledgements

The authors acknowledge and appreciate the contributions of the many Pachman Lab members and the Division of Pediatric Rheumatology who enabled the data collection: Dr. Klein-Gitelman cared for the JDM patients over the past 6 years. Drs. Megan Curran, Kaveh Ardalan and Christopher Costin were also involved. Dr. Amer Khojah started the data analysis while he was on the staff of Ann & Robert H. Lurie Children’s Hospital of Chicago, and completed his work while at Umm Al-Qura University.

Declarations

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of Ann & Robert H. Lurie Children’s Hospital of Chicago (IRB# 2012–14,858). Signed informed consent was obtained from all subjects involved in the study.
Consent for publication was obtained from all study subjects.

Competing interests

No potential conflict of interest is real or perceived by any of the authors.
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 Pachman LM, Nolan BE, DeRanieri D, Khojah AM. Juvenile Dermatomyositis: New clues to diagnosis and therapy. Curr Treatm Opt Rheumatol. 2021;7(1):39–62.CrossRefPubMedPubMedCentral Pachman LM, Nolan BE, DeRanieri D, Khojah AM. Juvenile Dermatomyositis: New clues to diagnosis and therapy. Curr Treatm Opt Rheumatol. 2021;7(1):39–62.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Mendez EP, Lipton R, Ramsey-Goldman R, Roettcher P, Bowyer S, Dyer A, et al. US incidence of juvenile dermatomyositis, 1995–1998: results from the National Institute of Arthritis and Musculoskeletal and Skin Diseases Registry. Arthritis Rheum. 2003;49(3):300–5.CrossRefPubMed Mendez EP, Lipton R, Ramsey-Goldman R, Roettcher P, Bowyer S, Dyer A, et al. US incidence of juvenile dermatomyositis, 1995–1998: results from the National Institute of Arthritis and Musculoskeletal and Skin Diseases Registry. Arthritis Rheum. 2003;49(3):300–5.CrossRefPubMed
3.
Zurück zum Zitat Pachman LM, Khojah AM. Advances in Juvenile Dermatomyositis: Myositis specific antibodies aid in understanding Disease Heterogeneity. J Pediatr. 2018;195:16–27.CrossRefPubMedPubMedCentral Pachman LM, Khojah AM. Advances in Juvenile Dermatomyositis: Myositis specific antibodies aid in understanding Disease Heterogeneity. J Pediatr. 2018;195:16–27.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Costin C, Morgan G, Khojah A, Klein-Gitelman M, Pachman LM. Lower NK cell numbers in children with untreated juvenile Dermatomyositis during the COVID-19 pandemic. Clin Immunol Commun. 2023. Costin C, Morgan G, Khojah A, Klein-Gitelman M, Pachman LM. Lower NK cell numbers in children with untreated juvenile Dermatomyositis during the COVID-19 pandemic. Clin Immunol Commun. 2023.
5.
Zurück zum Zitat Khojah A, Liu V, Savani SI, Morgan G, Shore R, Bellm J, et al. Association of p155/140 Autoantibody with loss of Nailfold Capillaries but not generalized Lipodystrophy: a study of ninety-six children with Juvenile Dermatomyositis. Arthritis Care Res (Hoboken). 2022;74(7):1065–9.CrossRefPubMed Khojah A, Liu V, Savani SI, Morgan G, Shore R, Bellm J, et al. Association of p155/140 Autoantibody with loss of Nailfold Capillaries but not generalized Lipodystrophy: a study of ninety-six children with Juvenile Dermatomyositis. Arthritis Care Res (Hoboken). 2022;74(7):1065–9.CrossRefPubMed
6.
Zurück zum Zitat Smith RL, Sundberg J, Shamiyah E, Dyer A, Pachman LM. Skin involvement in juvenile dermatomyositis is associated with loss of end row nailfold capillary loops. J Rheumatol. 2004;31(8):1644–9.PubMed Smith RL, Sundberg J, Shamiyah E, Dyer A, Pachman LM. Skin involvement in juvenile dermatomyositis is associated with loss of end row nailfold capillary loops. J Rheumatol. 2004;31(8):1644–9.PubMed
7.
Zurück zum Zitat Christen-Zaech S, Seshadri R, Sundberg J, Paller AS, Pachman LM. Persistent association of nailfold capillaroscopy changes and skin involvement over thirty-six months with duration of untreated Disease in patients with juvenile dermatomyositis. Arthritis Rheum. 2008;58(2):571–6.CrossRefPubMedPubMedCentral Christen-Zaech S, Seshadri R, Sundberg J, Paller AS, Pachman LM. Persistent association of nailfold capillaroscopy changes and skin involvement over thirty-six months with duration of untreated Disease in patients with juvenile dermatomyositis. Arthritis Rheum. 2008;58(2):571–6.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Gibbs EKA, Morgan G, Ehwerhemuepha L, Pachman LM. The Von Willebrand Factor Antigen reflects the Juvenile Dermatomyositis Disease activity score. Biomedicines. 2023;11(2):552.CrossRefPubMedPubMedCentral Gibbs EKA, Morgan G, Ehwerhemuepha L, Pachman LM. The Von Willebrand Factor Antigen reflects the Juvenile Dermatomyositis Disease activity score. Biomedicines. 2023;11(2):552.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Rouster-Stevens KA, Gursahaney A, Ngai KL, Daru JA, Pachman LM. Pharmacokinetic study of oral prednisolone compared with intravenous methylprednisolone in patients with juvenile dermatomyositis. Arthritis Rheum. 2008;59(2):222–6.CrossRefPubMedPubMedCentral Rouster-Stevens KA, Gursahaney A, Ngai KL, Daru JA, Pachman LM. Pharmacokinetic study of oral prednisolone compared with intravenous methylprednisolone in patients with juvenile dermatomyositis. Arthritis Rheum. 2008;59(2):222–6.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Saini I, Kalaivani M, Kabra SK. Calcinosis in juvenile dermatomyositis: frequency, risk factors and outcome. Rheumatol Int. 2016;36(7):961–5.CrossRef Saini I, Kalaivani M, Kabra SK. Calcinosis in juvenile dermatomyositis: frequency, risk factors and outcome. Rheumatol Int. 2016;36(7):961–5.CrossRef
11.
Zurück zum Zitat Bingham A, Mamyrova G, Rother KI, Oral E, Cochran E, Premkumar A, et al. Predictors of acquired lipodystrophy in juvenile-onset dermatomyositis and a gradient of severity. Med (Baltim). 2008;87(2):70–86.CrossRef Bingham A, Mamyrova G, Rother KI, Oral E, Cochran E, Premkumar A, et al. Predictors of acquired lipodystrophy in juvenile-onset dermatomyositis and a gradient of severity. Med (Baltim). 2008;87(2):70–86.CrossRef
12.
Zurück zum Zitat Bohan A, Peter JB. Polymyositis and dermatomyositis (second of two parts). N Engl J Med. 1975;292(8):403–7.CrossRef Bohan A, Peter JB. Polymyositis and dermatomyositis (second of two parts). N Engl J Med. 1975;292(8):403–7.CrossRef
13.
Zurück zum Zitat Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med. 1975;292(7):344–7.CrossRefPubMed Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med. 1975;292(7):344–7.CrossRefPubMed
14.
Zurück zum Zitat Bode RK, Klein-Gitelman MS, Miller ML, Lechman TS, Pachman LM. Disease activity score for children with juvenile dermatomyositis: reliability and validity evidence. Arthritis Rheum. 2003;49(1):7–15.CrossRefPubMed Bode RK, Klein-Gitelman MS, Miller ML, Lechman TS, Pachman LM. Disease activity score for children with juvenile dermatomyositis: reliability and validity evidence. Arthritis Rheum. 2003;49(1):7–15.CrossRefPubMed
15.
Zurück zum Zitat Rider LG, Werth VP, Huber AM, Alexanderson H, Rao AP, Ruperto N, et al. Measures of adult and juvenile dermatomyositis, polymyositis, and inclusion body myositis: physician and Patient/Parent global activity, manual muscle testing (MMT), Health Assessment Questionnaire (HAQ)/Childhood Health Assessment Questionnaire (C-HAQ), Childhood Myositis Assessment Scale (CMAS), Myositis Disease Activity Assessment Tool (MDAAT), Disease Activity score (DAS), short form 36 (SF-36), Child Health Questionnaire (CHQ), physician global damage, myositis damage index (MDI), quantitative muscle testing (QMT), Myositis Functional Index-2 (FI-2), Myositis activities Profile (MAP), inclusion body myositis functional rating scale (IBMFRS), cutaneous Dermatomyositis Disease Area and Severity Index (CDASI), cutaneous Assessment Tool (CAT), Dermatomyositis skin Severity Index (DSSI), Skindex, and Dermatology Life Quality Index (DLQI). Arthritis Care Res (Hoboken). 2011;63(0 11):118–57. Rider LG, Werth VP, Huber AM, Alexanderson H, Rao AP, Ruperto N, et al. Measures of adult and juvenile dermatomyositis, polymyositis, and inclusion body myositis: physician and Patient/Parent global activity, manual muscle testing (MMT), Health Assessment Questionnaire (HAQ)/Childhood Health Assessment Questionnaire (C-HAQ), Childhood Myositis Assessment Scale (CMAS), Myositis Disease Activity Assessment Tool (MDAAT), Disease Activity score (DAS), short form 36 (SF-36), Child Health Questionnaire (CHQ), physician global damage, myositis damage index (MDI), quantitative muscle testing (QMT), Myositis Functional Index-2 (FI-2), Myositis activities Profile (MAP), inclusion body myositis functional rating scale (IBMFRS), cutaneous Dermatomyositis Disease Area and Severity Index (CDASI), cutaneous Assessment Tool (CAT), Dermatomyositis skin Severity Index (DSSI), Skindex, and Dermatology Life Quality Index (DLQI). Arthritis Care Res (Hoboken). 2011;63(0 11):118–57.
16.
Zurück zum Zitat Pachman LM, Morgan G, Klein-Gitelman MS, Ahsan N, Khojah A. Nailfold capillary density in 140 untreated children with juvenile dermatomyositis: an indicator of Disease activity. Pediatr Rheumatol. 2023;21(1):118.CrossRef Pachman LM, Morgan G, Klein-Gitelman MS, Ahsan N, Khojah A. Nailfold capillary density in 140 untreated children with juvenile dermatomyositis: an indicator of Disease activity. Pediatr Rheumatol. 2023;21(1):118.CrossRef
17.
Zurück zum Zitat Schmeling H, Stephens S, Goia C, Manlhiot C, Schneider R, Luthra S, et al. Nailfold capillary density is importantly associated over time with muscle and Skin Disease activity in juvenile dermatomyositis. Rheumatology (Oxford). 2011;50(5):885–93.CrossRef Schmeling H, Stephens S, Goia C, Manlhiot C, Schneider R, Luthra S, et al. Nailfold capillary density is importantly associated over time with muscle and Skin Disease activity in juvenile dermatomyositis. Rheumatology (Oxford). 2011;50(5):885–93.CrossRef
18.
Zurück zum Zitat Lim LS, Pullenayegum E, Moineddin R, Gladman DD, Silverman ED, Feldman BM. Methods for analyzing observational longitudinal prognosis studies for rheumatic Diseases: a review & worked example using a clinic-based cohort of juvenile dermatomyositis patients. Pediatr Rheumatol Online J. 2017;15(1):18.CrossRefPubMedCentral Lim LS, Pullenayegum E, Moineddin R, Gladman DD, Silverman ED, Feldman BM. Methods for analyzing observational longitudinal prognosis studies for rheumatic Diseases: a review & worked example using a clinic-based cohort of juvenile dermatomyositis patients. Pediatr Rheumatol Online J. 2017;15(1):18.CrossRefPubMedCentral
19.
Zurück zum Zitat Papadopoulou C, Chew C, Wilkinson MGL, McCann L, Wedderburn LR. Juvenile idiopathic inflammatory myositis: an update on pathophysiology and clinical care. Nat Rev Rheumatol. 2023;19(6):343–62.CrossRefPubMedPubMedCentral Papadopoulou C, Chew C, Wilkinson MGL, McCann L, Wedderburn LR. Juvenile idiopathic inflammatory myositis: an update on pathophysiology and clinical care. Nat Rev Rheumatol. 2023;19(6):343–62.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Kishi T, Chipman J, Evereklian M, Nghiem K, Stetler-Stevenson M, Rick ME, et al. Endothelial activation markers as Disease activity and damage measures in Juvenile Dermatomyositis. J Rheumatol. 2020;47(7):1011–8.CrossRefPubMed Kishi T, Chipman J, Evereklian M, Nghiem K, Stetler-Stevenson M, Rick ME, et al. Endothelial activation markers as Disease activity and damage measures in Juvenile Dermatomyositis. J Rheumatol. 2020;47(7):1011–8.CrossRefPubMed
21.
Zurück zum Zitat Karasawa R, Tamaki M, Sato T, Tanaka M, Nawa M, Yudoh K, et al. Multiple target autoantigens on endothelial cells identified in juvenile dermatomyositis using proteomics. Rheumatology (Oxford). 2018;57(4):671–6.CrossRefPubMed Karasawa R, Tamaki M, Sato T, Tanaka M, Nawa M, Yudoh K, et al. Multiple target autoantigens on endothelial cells identified in juvenile dermatomyositis using proteomics. Rheumatology (Oxford). 2018;57(4):671–6.CrossRefPubMed
22.
Zurück zum Zitat Yu HH, Chang HM, Chiu CJ, Yang YH, Lee JH, Wang LC, et al. Detection of anti-p155/140, anti-p140, and antiendothelial cells autoantibodies in patients with juvenile dermatomyositis. J Microbiol Immunol Infect. 2016;49(2):264–70.CrossRefPubMed Yu HH, Chang HM, Chiu CJ, Yang YH, Lee JH, Wang LC, et al. Detection of anti-p155/140, anti-p140, and antiendothelial cells autoantibodies in patients with juvenile dermatomyositis. J Microbiol Immunol Infect. 2016;49(2):264–70.CrossRefPubMed
23.
Zurück zum Zitat Bukhari A, Khojah A, Marin W, Khramtsov A, Khramtsova G, Costin C, et al. Increased otoferlin expression in B cells is Associated with muscle weakness in untreated juvenile dermatomyositis: a pilot study. Int J Mol Sci. 2023;24(13):10553.CrossRefPubMedPubMedCentral Bukhari A, Khojah A, Marin W, Khramtsov A, Khramtsova G, Costin C, et al. Increased otoferlin expression in B cells is Associated with muscle weakness in untreated juvenile dermatomyositis: a pilot study. Int J Mol Sci. 2023;24(13):10553.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Ochfeld E, Hans V, Marin W, Ahsan N, Morgan G, Pachman LM, et al. Coding joint: kappa-deleting recombination excision circle ratio and B cell activating factor level: predicting juvenile dermatomyositis rituximab response, a proof-of-concept study. BMC Rheumatol. 2022;6(1):36.CrossRefPubMedPubMedCentral Ochfeld E, Hans V, Marin W, Ahsan N, Morgan G, Pachman LM, et al. Coding joint: kappa-deleting recombination excision circle ratio and B cell activating factor level: predicting juvenile dermatomyositis rituximab response, a proof-of-concept study. BMC Rheumatol. 2022;6(1):36.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Khojah A, Morgan G, Pachman LM. Clues to Disease Activity in Juvenile Dermatomyositis: neopterin and other biomarkers. Diagnostics (Basel). 2021;12(1). Khojah A, Morgan G, Pachman LM. Clues to Disease Activity in Juvenile Dermatomyositis: neopterin and other biomarkers. Diagnostics (Basel). 2021;12(1).
26.
Zurück zum Zitat McLellan K, Papadopoulou C. Update on biomarkers of Vasculopathy in Juvenile and Adult Myositis. Curr Rheumatol Rep. 2022;24(7):227–37.CrossRefPubMed McLellan K, Papadopoulou C. Update on biomarkers of Vasculopathy in Juvenile and Adult Myositis. Curr Rheumatol Rep. 2022;24(7):227–37.CrossRefPubMed
27.
Zurück zum Zitat Bloom BJ, Miller LC, Blier PR. Soluble adhesion molecules in pediatric rheumatic Diseases. J Rheumatol. 2002;29(4):832–6.PubMed Bloom BJ, Miller LC, Blier PR. Soluble adhesion molecules in pediatric rheumatic Diseases. J Rheumatol. 2002;29(4):832–6.PubMed
28.
Zurück zum Zitat Wang A, Khojah A, Morgan G, Pachman LM. Nailfold capillary dropout precedes the presentation of Pneumatosis Intestinalis and micro-perforation in juvenile dermatomyositis. Clin Immunol Commun. 2023;3:74–6.CrossRef Wang A, Khojah A, Morgan G, Pachman LM. Nailfold capillary dropout precedes the presentation of Pneumatosis Intestinalis and micro-perforation in juvenile dermatomyositis. Clin Immunol Commun. 2023;3:74–6.CrossRef
Metadaten
Titel
Juvenile dermatomyositis: association between nail fold capillary end row loop– area under the curve– and disease damage indicators
verfasst von
Amer Khojah
Gabrielle Morgan
Marisa S. Klein-Gitelman
Lauren M. Pachman
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Pediatric Rheumatology / Ausgabe 1/2023
Elektronische ISSN: 1546-0096
DOI
https://doi.org/10.1186/s12969-023-00919-3

Weitere Artikel der Ausgabe 1/2023

Pediatric Rheumatology 1/2023 Zur Ausgabe

Kinder mit anhaltender Sinusitis profitieren häufig von Antibiotika

30.04.2024 Rhinitis und Sinusitis Nachrichten

Persistieren Sinusitisbeschwerden bei Kindern länger als zehn Tage, ist eine Antibiotikatherapie häufig gut wirksam: Ein Therapieversagen ist damit zu über 40% seltener zu beobachten als unter Placebo.

Neuer Typ-1-Diabetes bei Kindern am Wochenende eher übersehen

23.04.2024 Typ-1-Diabetes Nachrichten

Wenn Kinder an Werktagen zum Arzt gehen, werden neu auftretender Typ-1-Diabetes und diabetische Ketoazidosen häufiger erkannt als bei Arztbesuchen an Wochenenden oder Feiertagen.

Neue Studienergebnisse zur Myopiekontrolle mit Atropin

22.04.2024 Fehlsichtigkeit Nachrichten

Augentropfen mit niedrig dosiertem Atropin können helfen, das Fortschreiten einer Kurzsichtigkeit bei Kindern zumindest zu verlangsamen, wie die Ergebnisse einer aktuellen Studie mit verschiedenen Dosierungen zeigen.

Spinale Muskelatrophie: Neugeborenen-Screening lohnt sich

18.04.2024 Spinale Muskelatrophien Nachrichten

Seit 2021 ist die Untersuchung auf spinale Muskelatrophie Teil des Neugeborenen-Screenings in Deutschland. Eine Studie liefert weitere Evidenz für den Nutzen der Maßnahme.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.