1
|
Liu X, Zhang X, Cen M. Dysregulation of miR-106a-5p/PTEN axis associated with progression and diagnostic of postmenopausal osteoporosis. J Orthop Surg Res 2025; 20:456. [PMID: 40355896 PMCID: PMC12070524 DOI: 10.1186/s13018-025-05872-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE Postmenopausal osteoporosis (PMOP) is a bone disorder in postmenopausal women and a significant risk factor for fragility fractures. This study aims to explore the role of miR-106a-5p in the pathogenesis of PMOP and its potential as a diagnostic biomarker. METHODS 220 postmenopausal women were recruited. The levels of miR-106a-5p, PTEN, and osteogenic-related genes were quantified using qRT-PCR. The relative protein of PTEN was detected using Western blotting. ROC curve and Pearson correlation were employed to evaluate the diagnostic value and relationships between variables. To model iron accumulation, hFOB1.19 osteoblasts were treated with ferric ammonium citrate (FAC). Cell proliferation and apoptosis were assessed using the CCK-8 and flow cytometry. The target relationship was verified using dual-luciferase assays. RESULTS miR-106a-5p levels were reduced, while PTEN levels were increased in PMOP. miR-106a-5p was positively correlated with bone mineral density and negatively correlated with ferritin. In the FAC-treated cells, miR-106a-5p decreased, and PTEN increased. Dual-luciferase assays confirmed that miR-106a-5p targets PTEN. Successful transfection was confirmed by observing the corresponding changes in miR-106a-5p and PTEN expression. Up-regulated miR-106a-5p increased the PTEN protein level, mRNA expression of RUNX2, OPN, and OCN, promoted cell proliferation, and decreased cell apoptosis under iron accumulation conditions. These effects were reversed by the upregulation of PTEN. CONCLUSION miR-106a-5p has the potential to diagnose osteoporosis in postmenopausal women and is linked to ferritin levels. miR-106a-5p plays a protective role in PMOP by regulating PTEN under conditions of iron accumulation, suggesting its potential as a promising biomarker for PMOP.
Collapse
Affiliation(s)
- Xiangjie Liu
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Xiaogang Zhang
- Department of Orthopedics, Hebei Yanda Hospital, Sanhe, 065201, China
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
- Guangxi Key Laboratory for Preclinical and Translational Research on Bone and Joint Degenerative Diseases, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, China.
| |
Collapse
|
2
|
Yu WL, Ma FZ, Huang H, Xiao BH, Li XM, Wáng YXJ. Age and gender differences of normative values of spleen diffusion MRI parameters. ROFO-FORTSCHR RONTG 2025; 197:535-545. [PMID: 39079680 DOI: 10.1055/a-2357-9741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
This study investigates age and gender differences of normative values of spleen diffusion MRI parameters.We recruited 124 volunteers with MRI conducted at 1.5T. Diffusion imaging had b-values of 0, 2, 4, 7, 10, 15, 20, 30, 46, 60, 72, 100, 150, 200, 400, 600 s/mm2. ADC, IVIM-Dslow, IVIM-PF, IVIM-Dfast, and DDVD (diffusion-derived vessel density) were computed. DDVD is the signal difference between the b=0 s/mm2 image and b=2, 4 s/mm2 image. Only images without apparent artifacts and with good curving fitting were included in the analysis. Finally, 34 females (age: 20-71 years) and 69 males (22-70 years) were measured with ADC; 20 females (20-71 years) and 48 males (22-67 years) were measured with IVIM; 32 females (20-71 years) and 65 males (22-70 years) were measured with DDVD parameter.An age-related decrease in ADC was noted for females, while such a trend was not noted for males. A very high level of heterogeneity was noted for the data for the males, with the highest ADC value being 1.710 × 10-3mm 2/s and the lowest ADC value being 0.705 × 10-3 mm2/s when b=0 and 600 s/mm 2 were used for ADC calculation. A male-female data comparison did not show a statistically significant difference between the ADC median value. However, ADCs > 1.3 × 10-3 mm2/s were only seen among males. A very high level of heterogeneity was also noted for males' Dslow, with the highest value being 1.468 × 10-3 mm2/s and the lowest value being 0.600 × 10-3 mm2/s. Both PF and Dfast demonstrated a trend of age-related increase for older subjects. PF values were higher among males than females. However, no difference was noted for Dfast between males and females. DDVD did not show an age-related trend both for females and males. No difference was noted in DDVD values between males and females.Interpreting normal spleen diffusion MRI parameters should consider age and gender factors. · An age-related decrease in spleen ADC and IVIM-Dslow was seen for healthy females.. · There is a high level of heterogeneity for spleen ADC and IVIM-Dslow data for healthy males.. · IVIM modelled perfusion fraction and Dfast demonstrate an artificial trend of age-related increase for older subjects.. · Vessel density measured on diffusion imaging does not show an age-related trend.. · Yu W, Ma FZ, Huang H et al. Age and gender differences of normative values of spleen diffusion MRI parameters. Rofo 2025; 197: 535-545.
Collapse
Affiliation(s)
- Wei-Ling Yu
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong
| | - Fu-Zhai Ma
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong
| | - Hua Huang
- Department of Radiology, The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Ben-Heng Xiao
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong
| | - Xin-Ming Li
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yì Xiáng J Wáng
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong
| |
Collapse
|
3
|
Barnett AL, Wenger MJ, Miles P, Wu D, Isingizwe ZR, Benbrook DM, Yuan H. Cognitive Performance in Relation to Systemic and Brain Iron at Perimenopause. Nutrients 2025; 17:745. [PMID: 40077615 PMCID: PMC11901746 DOI: 10.3390/nu17050745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The literature on the relationships among blood iron levels, cognitive performance, and brain iron levels specific to women at the menopausal transition is ambiguous at best. The need to better understand these potential relationships in women for whom monthly blood loss (and thus iron loss) is ceasing is highlighted by iron's accumulation in brain tissue over time, thought to be a factor in the development of neurodegenerative disease. METHODS Non-anemic women who were either low in iron or had normal iron levels for their age and race/ethnicity provided blood samples, underwent MRI scans to estimate brain iron levels, and performed a set of cognitive tasks with concurrent EEG. RESULTS Cognitive performance and brain dynamics were positively related to iron levels, including measures associated with oxygen transport. There were no relationships between any of the blood measures of iron and brain iron. CONCLUSIONS Higher iron status was associated with better cognitive performance in a sample of women who were neither iron deficient nor anemic, without there being any indication that higher levels of systemic iron were related to higher levels of brain iron. Consequently, addressing low iron levels at the menopausal transition may be a candidate approach for alleviating the "brain fog" commonly experienced at menopause.
Collapse
Affiliation(s)
- Amy L. Barnett
- Psychology and Cellular and Behavioral Neurobiology, The University of Oklahoma, Norman, OK 73019, USA;
| | - Michael J. Wenger
- Psychology and Cellular and Behavioral Neurobiology, The University of Oklahoma, Norman, OK 73019, USA;
| | - Pamela Miles
- Obstetrics and Gynecology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Dee Wu
- Radiological Sciences, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Zitha Redempta Isingizwe
- Gynecological Oncology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Z.R.I.); (D.M.B.)
| | - Doris M. Benbrook
- Gynecological Oncology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Z.R.I.); (D.M.B.)
| | - Han Yuan
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, USA;
| |
Collapse
|
4
|
Barad A, Xu Y, Bender E, Kang W, Xu R, Gu Z, Pressman EK, O'Brien KO. Characterization of iron status biomarkers and hematological indices among young adults of East Asian or Northern European ancestry: A cross-sectional analysis from the Iron Genes in East Asian and Northern European Adults Study (FeGenes). Am J Clin Nutr 2025; 121:394-405. [PMID: 39909709 PMCID: PMC11863323 DOI: 10.1016/j.ajcnut.2024.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Excess body iron (Fe) accrual is linked to chronic diseases. East Asian (EA) adults (median age 50 y) were reported to have higher Fe stores compared to other populations despite lacking the mutation that causes Fe overload in Northern European (NE) adults. It is unknown if these differences are evident in a healthy population under 50 y of age. OBJECTIVES This cross-sectional study aims to compare Fe-related markers in young adults of EA and NE ancestry and identify determinants of Fe status. METHODS Participants were healthy United States males and premenopausal/nonpregnant females of genetically confirmed EA (n = 251) or NE (n = 253) ancestry, aged 18-50 y and without obesity. A complete blood count was obtained. Serum ferritin (SF; μg/L), c-reactive protein, and interleukin-6 were measured by immunoassay, and serum soluble transferrin receptor (mg/L) and transferrin by quantitative immunoturbidimetry. Total body Fe (mg/kg) was calculated. Elevated Fe stores were defined as SF >200 (females) or >300 (males) and c-reactive protein <5 mg/L. Results are shown as the geometric mean 95% confidence interval (CI) or mean ± standard deviation. RESULTS The mean age of the population was (26.3 y; 25.6, 26.9 y), with 69.2% of participants aged under 30 y. SF was higher in EA (172; 152, 194) compared with NE (85.3; 76.8, 94.8) males (P < 0.001), and in EA (42.6; 36.7, 49.5) compared with NE (31.9; 27.8, 36.5) females (P = 0.004). The prevalence of elevated Fe stores was 16.7% in EA compared with 0.8% in NE males (P < 0.001) and 1.6% in EA compared with 0% in NE females (P = 0.47). Total body Fe was higher in EA (11.7 ± 2.7) compared with NE (9.1 ± 2.6) males (P < 0.001) and in EA (6.7 ± 3.6) compared with NE (5.6 ± 3.4) females (P = 0.01). All differences persisted after adjustment for confounders (all P < 0.05). CONCLUSIONS Individuals of EA ancestry had a significantly greater body Fe burden compared to NE individuals. Of concern, these differences were evident in a cohort primarily consisting of young individuals aged 18-29 y. This trial was registered at clinicaltrials.gov as NCT04198545.
Collapse
Affiliation(s)
- Alexa Barad
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Yaqin Xu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Erica Bender
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Wanhui Kang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Ruihan Xu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Eva K Pressman
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, United States
| | - Kimberly O O'Brien
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
5
|
Barnett AL, Wenger MJ, Miles P, Wu D, Isingizwe ZR, Benbrook DM, Yuan H. Cognitive Performance in Relation to Systemic and Brain Iron at Perimenopause. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.28.25321122. [PMID: 39974002 PMCID: PMC11838962 DOI: 10.1101/2025.01.28.25321122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background The literature on the relationships among blood iron levels, cognitive performance, and brain iron levels specific to women at the menopausal transition is ambiguous at best. The need to better to understand these potential relationships in women for whom monthly blood loss (and thus iron loss) is ceasing is highlighted by the fact that iron accumulates in brain tissue over time and that accumulation is thought to be a factor in the development of neurodegenerative disease. Methods Non-anemic women who were either low in iron or had normal iron levels for their age and race/ethnicity provided blood samples, underwent MRI scans to estimate brain iron levels, and performed a set of cognitive tasks with concurrent EEG. Results: Cognitive performance as well as brain dynamics were positively related to iron levels, including measures associated with oxygen transport. There were no relationships between any of the blood measures of iron and brain iron. Conclusions Higher iron status was associated with better cognitive performance in a sample of women who were neither iron deficient nor anemic, without there being any indication that higher levels of systemic iron were related to higher levels of brain. Consequently, addressing low iron levels at the menopausal transition may be a candidate approach for alleviating the "brain fog" commonly experienced at menopause.
Collapse
|
6
|
Boonyagars L, Thinpangnga P, Munsakul W. Incidence and Risk Factors of Zidovudine-Induced Anemia in Patients With HIV Infection Receiving Zidovudine-Containing Antiretroviral Therapy. J Int Assoc Provid AIDS Care 2025; 24:23259582251321577. [PMID: 39967242 PMCID: PMC11863231 DOI: 10.1177/23259582251321577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Anemia is among the frequently observed conditions among people living with HIV (PLWH). Zidovudine (AZT) is the most common ART that causes anemia. This study aimed to identify the incidence and risk factors of AZT-induced anemia in PLWH receiving AZT-containing regimens. METHODS A retrospective cohort study was conducted on nonpregnant PLWH aged >18 years who received AZT-containing regimens for >6 months and had normal baseline hemoglobin levels. Data on medical history, clinical characteristics, and laboratory examination were collected. Cox proportional hazard regression analysis was performed to determine the risk factors of AZT-induced anemia. RESULTS A total of 401 individuals were included in the study. In total, 71 individuals presented with AZT-induced anemia. The incidence rate of anemia was 1.98 per 100 person-years of observations. Female sex, low-normal baseline hemoglobin level, low recent and low baseline CD4 cell count were independently associated with an increased risk of anemia. However, a higher body mass index was independently associated with a decreased risk of anemia. CONCLUSION Routine screening, early detection, and treatment of anemia should be considered in PLWH receiving AZT-containing regimens.
Collapse
Affiliation(s)
- Lakkana Boonyagars
- Division of Infectious Diseases, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Peerawit Thinpangnga
- Research Facilitation Division, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Warangkana Munsakul
- Division of Infectious Diseases, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| |
Collapse
|
7
|
Conway RB, Pratte KA, Bowler RP, Young KA, Kinney GL, Austin E, Li Y, McClain D, Hokanson J, Crapo JD. Plasma Proteomic Markers of Iron and Risk of Diabetes in a Cohort of African American and White American Current and Former Smokers. Diabetes Metab Syndr Obes 2024; 17:4767-4776. [PMID: 39678225 PMCID: PMC11646377 DOI: 10.2147/dmso.s492124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024] Open
Abstract
Background Little information is available on iron with diabetes risk among African Americans, a population where both anemia and elevated ferritin are common. We tested whether plasma proteomic measurements of ferritin and transferrin were associated with increased diabetes risk in a cohort of current and former African American (NHB) and Non-Hispanic White (NHW) smokers. Methods NHB and NHW participants from the COPDGene study who were free of diabetes (n = 4693) at baseline were followed for incident diabetes. The SomaScan was used to determine the relative amounts of natural log-transformed ferritin, transferrin, and hepcidin. Findings During an average of 5.6 years of follow-up, diabetes incidence was 7.9%. Ferritin at follow-up was higher in NHB than NHW participants (p = <0.0001). Ferritin at follow-up was associated with increased diabetes risk (OR = 1.36, 95% CI = 1.08-1.70), while transferrin was associated with decreased risk (OR = 0.25, 95% CI = 0.08-0.77) controlling for age, sex, BMI, smoking pack-years, hepcidin, CRP, and Il-6. Race-specifically, increased risk associated with higher ferritin levels among NHB (OR = 1.56, 95% CI = 1.13-2.16) but not NHW (OR = 1.22, 95% CI = 0.89-1.68) participants. Sex-specifically, ferritin's relationship was similar among NHB men and women and NHW women (ORs ranging from 1.41-1.59); but not NHW men (OR = 0.98, 95% CI = 0.64-1.49). Similarly, transferrin ORs non-significantly ranged from 0.19-0.30 for NHB men and women and NHW women, but was significant for NHW men (OR = 0.07, 95% CI = 0.01-0.63). Interpretation Higher body iron stores is associated with increased diabetes risk among both NHB and NHW people. Unsuspected elevated iron stores may increase diabetes risk in NHB patients and should be monitored.
Collapse
Affiliation(s)
- Rebecca Baqiyyah Conway
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Katherine A Pratte
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, CO, 80206, USA
| | - Russell Paul Bowler
- Department of Genomic Sciences, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
| | - Kendra A Young
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Gregory l Kinney
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Erin Austin
- Department of Mathematical and Statistical Sciences, Denver, University of Colorado, Denver, CO, 80204, USA
| | - Yisha Li
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Donald McClain
- Section of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John Hokanson
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - James D Crapo
- Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| |
Collapse
|
8
|
Wang D, Wang J, Yu Z, Yao R, Zhang J, Zhao X. Quercetin Alleviates Perimenopausal Depression Induced by Ovariectomy Combined with Chronic Unpredictable Mild Stress Through Regulating Serum Elements and Inhibiting Ferroptosis in Prefrontal Cortex of Rats. Biol Trace Elem Res 2024; 202:5596-5611. [PMID: 38388751 DOI: 10.1007/s12011-024-04106-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
This study investigated the effects of quercetin on the alterations of serum elements in perimenopausal depression rat model induced by ovariectomy combined with chronic unpredictable mild stress (OVX-CUMS) and possible mechanisms. According to the results of the sucrose preference test, the rats were randomly assigned to four groups: sham, OVX-CUMS, OVX-CUMS + 17β-estradiol (17β-estradiol: 0.27 mg/kg.bw), and OVX-CUMS + Quercetin (Quercetin: 50 mg/kg.bw). At the end of experiment, serum and prefrontal cortex of rats were collected. The inductively coupled plasma mass spectrometry (ICP-MS) analysis showed that levels of calcium (Ca), magnesium (Mg), selenium (Se), cobalt (Co) and zinc (Zn) decreased, and levels of iron (Fe) and copper (Cu) increased in serum and prefrontal cortex of OVX-CUMS rats compared with sham group (p < 0.01). Meanwhile, the levels of the above elements in prefrontal cortex had correlation with behavioral characteristics in OVX-CUMS rats (p < 0.05 or p < 0.01). The abnormal elements in serum may cross blood-brain-barrier into the brain and induce oxidative stress, leading to ferroptosis. Furtherly, the expressions of ferroptosis-related protein including GPX4 and SLC7A11 were decreased in prefrontal cortex of OVX-CUMS rats (p < 0.01), which confirmed the above results. Quercetin treatment restored the above abnormal indicators (p < 0.05 or p < 0.01) induced by OVX-CUMS in rats. Our study suggested that quercetin regulated variation of elements in serum and prefrontal cortex, further inhibiting ferroptosis in prefrontal cortex through alleviating oxidative stress in OVX-CUMS rats.
Collapse
Affiliation(s)
- Dan Wang
- Department of Nutrition and Food Hygiene, Key Laboratory of Precision Nutrition and Health, Ministry of Education, School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Jing Wang
- Department of Nutrition and Food Hygiene, Key Laboratory of Precision Nutrition and Health, Ministry of Education, School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Ziran Yu
- Department of Nutrition and Food Hygiene, Key Laboratory of Precision Nutrition and Health, Ministry of Education, School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Ranqi Yao
- Department of Nutrition and Food Hygiene, Key Laboratory of Precision Nutrition and Health, Ministry of Education, School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Jingnan Zhang
- Department of Nutrition and Food Hygiene, Key Laboratory of Precision Nutrition and Health, Ministry of Education, School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Xiujuan Zhao
- Department of Nutrition and Food Hygiene, Key Laboratory of Precision Nutrition and Health, Ministry of Education, School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
9
|
Oh SJ, Shin YY, Ahn JS, Park HJ, Kang MJ, Shin TH, Lee BC, Kim WK, Oh JM, Lee D, Kim YH, Kim JM, Sung ES, Lee EW, Jeong JH, Lee BJ, Seo Y, Kim HS. TGFβ2-Driven Ferritin Degradation and Subsequent Ferroptosis Underlie Salivary Gland Dysfunction in Postmenopausal Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400660. [PMID: 39481440 DOI: 10.1002/advs.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/05/2024] [Indexed: 11/02/2024]
Abstract
Despite the high incidence of dry mouth in postmenopausal women, its underlying mechanisms and therapeutic interventions remain underexplored. Using ovariectomized (OVX) mouse models, here this study identifies ferroptosis, an iron-dependent regulated cell death, as a central mechanism driving postmenopausal salivary gland (SG) dysfunction. In the OVX-SGs, TGFβ signaling pathway is enhanced with the aberrant TGFβ2 expression in SG mesenchymal cells. Intriguingly, TGFβ2 treatment reduces iron-storing ferritin levels, leading to lipid peroxidation and ferroptotic death in SG epithelial organoids (SGOs). Mechanistically, TGFβ2 promotes the autophagy-mediated ferritin degradation, so-called ferritinophagy. A notable overexpression of the type III TGFβ receptor (TβRIII) is found in the OVX-SGs and TGFβ2-treated SGOs, while the silencing of TβRIII mitigates the ferroptosis-mediated deleterious effects of TGFβ2 on SGOs. Finally, administration of ferroptosis inhibitor, Liproxstatin-1 (Lip-1), improves saliva secretion in OVX mice. Present findings collectively suggest a link between TGFβ signaling, ferroptosis, and SG injury, offering new therapeutic avenues for postmenopausal xerostomia.
Collapse
Affiliation(s)
- Su-Jeong Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ye Young Shin
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Seoul, 08590, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Tae-Hoon Shin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju-si, 63243, Republic of Korea
| | - Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Won Kyu Kim
- Natural Product Research Center, Korea Institute of Science andTechnology (KIST), Gangneung, 25451, Republic of Korea
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
- Division of Natural Products Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jung-Min Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Ji Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Republic of Korea
| | - Eui-Suk Sung
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Pusan National University School of Medicine, Yangsan Pusan National University Hospital, Yangsan, 50612, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Byung-Joo Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| |
Collapse
|
10
|
Thompson RB, Sherrington R, Beaulieu C, Kirkham A, Paterson DI, Seres P, Grenier J. Reference Values for Water-Specific T1 of the Liver at 3 T: T2*-Compensation and the Confounding Effects of Fat. J Magn Reson Imaging 2024; 60:2063-2075. [PMID: 38305588 DOI: 10.1002/jmri.29262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND T1 mapping of the liver is confounded by the presence of fat. Multiparametric T1 mapping combines fat-water separation with T1-weighting to enable imaging of water-specific T1 (T1Water), proton density fat fraction (PDFF), and T2* values. However, normative T1Water values in the liver and its dependence on age/sex is unknown. PURPOSE Determine normative values for T1Water in the liver with comparison to MOLLI and evaluate a T2*-compensation approach to reduce T1 variability. STUDY TYPE Prospective observational; phantoms. POPULATIONS One hundred twenty-four controls (56 male, 18-75 years), 50 patients at-risk for liver disease (18 male, 30-76 years). FIELD STRENGTH/SEQUENCE 2.89 T; Saturation-recovery chemical-shift encoded T1 Mapping (SR-CSE); MOLLI. ASSESSMENT SR-CSE provided T1Water measurements, PDFF and T2* values in the liver across three slices in 6 seconds. These were compared with MOLLI T1 values. A new T2*-compensation approach to reduce T1 variability was evaluated test/re-test reproducibility. STATISTICAL TESTS Linear regression, ANCOVA, t-test, Bland and Altman, intraclass correlation coefficient (ICC). P < 0.05 was considered statistically significant. RESULTS Liver T1 values were significantly higher in healthy females (F) than males (M) for both SR-CSE (F-973 ± 78 msec, M-930 ± 72 msec) and MOLLI (F-802 ± 55 msec, M-759 ± 69 msec). T1 values were negatively correlated with age, with similar sex- and age-dependencies observed in T2*. The T2*-compensation model reduced the variability of T1 values by half and removed sex- and age-differences (SR-CSE: F-946 ± 36 msec, M-941 ± 43 msec; MOLLI: F-775 ± 35 msec, M-770 ± 35 msec). At-risk participants had elevated PDFF and T1 values, which became more distinct from the healthy cohort after T2*-compensation. MOLLI systematically underestimated liver T1 values by ~170 msec with an additional positive T1-bias from fat content (~11 msec/1% in PDFF). Reproducibility ICC values were ≥0.96 for all parameters. DATA CONCLUSION Liver T1Water values were lower in males and decreased with age, as observed for SR-CSE and MOLLI acquisitions. MOLLI underestimated liver T1 with an additional large positive fat-modulated T1 bias. T2*-compensation removed sex- and age-dependence in liver T1, reduced the range of healthy values and increased T1 group differences between healthy and at-risk groups. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Richard B Thompson
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Rachel Sherrington
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Christian Beaulieu
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Amy Kirkham
- Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - David I Paterson
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Peter Seres
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Justin Grenier
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Wylenzek F, Bühling KJ, Laakmann E. A systematic review on the impact of nutrition and possible supplementation on the deficiency of vitamin complexes, iron, omega-3-fatty acids, and lycopene in relation to increased morbidity in women after menopause. Arch Gynecol Obstet 2024; 310:2235-2245. [PMID: 38935105 PMCID: PMC11393286 DOI: 10.1007/s00404-024-07555-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 05/14/2024] [Indexed: 06/28/2024]
Abstract
A balanced and healthy diet during the menopausal transition and after menopause is crucial for women to reduce the risk for morbidities and chronic diseases due to deficiency of essential nutrients. PURPOSE The objective of this study was to conduct a systematic review of studies that analyzed the impact of vitamin and nutrient deficiencies in postmenopausal women in relation to increased morbidities and chronic conditions. METHODS Observational studies were searched in the databases PubMed, UpToDate, and Google Scholar. RESULTS We searched 122 studies, of which 90 were included in our analysis. The meta-analysis of the data could not be performed because of the heterogeneity of the statistical methods in the included studies. In our study, we focused on the aspects of vitamin B6, vitamin B12, vitamin D, iron, omega-3-fatty acids, and lycopene, belonging to the family of carotenoids. Postmenopausal women with deficiencies of these nutrients are more vulnerable to comorbidities such as cardiovascular and cerebrovascular events, metabolic diseases, osteoporosis, obesity, cancer and neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, depression, cognitive decline, dementia, and stroke. We concluded that women after menopause tend to have a greater probability of suffering from deficiencies in various vitamins and nutrients, and consequently have an increased risk of developing morbidities and chronic diseases. CONCLUSION In conclusion, maintaining optimum serum levels of nutrients and vitamins, either through a balanced and healthy diet consuming fresh fruits, vegetables, and fats or by taking appropriate supplementation, is essential in maintaining optimal health-related quality of life and reducing the risk for women during the menopausal transition and after menopause. Nevertheless, more recent studies need to be assessed to formulate adequate recommendations to achieve positive clinical outcomes.
Collapse
Affiliation(s)
- Friederike Wylenzek
- Department of Gynecological Endocrinology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Kai J Bühling
- Department of Gynecological Endocrinology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Elena Laakmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
12
|
Wu Y, Xiao M, Chen J, Tao Y, Chen A, Lin H, Xu Y, Li L, Jia H, Xue Y, Jia Y, Zheng Z. Association of dietary iron intake with diabetic kidney disease among individuals with diabetes. Endocrine 2024; 85:1154-1161. [PMID: 38758293 DOI: 10.1007/s12020-024-03819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/02/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE The current study investigated the correlation between dietary iron intake and diabetic kidney disease among diabetic adults. METHODS This cross-sectional study enrolled 8118 participants who suffered from diabetes from the National Health and Nutrition Examination Survey (NHANES) 1999-2018. Dietary iron intake was obtained from 24 h recall interviews, and diabetic kidney disease was defined as eGFR < 60 mL/min per 1.73 m2 or albumin creatinine ratio (ACR) ≥ 30 mg/g. Three weighted logistic regression models were utilized to investigate odd ratio (OR) and 95% CIs for diabetic kidney disease. Stratified analyses were performed by gender, age, BMI, HbA1c, hypertension status, and smoking status, and diabetes types. RESULTS Among 8118 participants (51.6% male, mean age 61.3 years), 40.7% of participants suffered from diabetic kidney disease. With the adjustment of potential covariates, we found that ≥ 12.59 mg of dietary iron was related to a lower risk of diabetic kidney disease (OR = 0.78, 95% CI: 0.63 to 0.96; OR = 0.79, 95% CI: 0.63 to 0.98). In stratified analyses, higher iron intake was negatively related to diabetic kidney disease, especially among those who were male, < 60 years, those with hypertension, those with HbA1c < 7.0%, and those who were ex-smokers. The result remained robust in sensitivity analyses. CONCLUSION We found that ≥ 12.59 mg of dietary iron is associated with a lower risk of diabetic kidney disease, especially in those who were male, younger, heavier weight, have better blood sugar control, and those who were ex-smokers.
Collapse
Affiliation(s)
- Yichuan Wu
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Manlu Xiao
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaqi Chen
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Tao
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Aomiao Chen
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Huanjia Lin
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Ying Xu
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Linna Li
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongxia Jia
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaoming Xue
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yijie Jia
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- De Feng Academy, Southern Medical University, Guangzhou, China.
| | - Zongji Zheng
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- De Feng Academy, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Barton JC, Wiener HW, Barton JC, Acton RT. Prevalence of Iron Deficiency Using 3 Definitions Among Women in the US and Canada. JAMA Netw Open 2024; 7:e2413967. [PMID: 38848068 PMCID: PMC11161847 DOI: 10.1001/jamanetworkopen.2024.13967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/26/2024] [Indexed: 06/10/2024] Open
Abstract
Importance The prevalence of iron deficiency varies widely according to how it is defined. Objective To compare the prevalence of iron deficiency among women using 3 different definitions. Design, Setting, and Participants The cross-sectional Hemochromatosis and Iron Overload Screening Study (HEIRS; 2000-2006) evaluated the prevalence, determinants, and outcomes of hemochromatosis and other iron-related disorders. Multiethnic, primary care-based screening (2001-2003) was performed at 5 field centers (4 in the US and 1 in Canada). Volunteer women aged 25 years and older were recruited at primary care venues associated with the field centers. Data were analyzed from June to December 2023. Main Outcomes and Measures Measures included transferrin saturation, serum ferritin level, and self-reported age, pregnancy, and race and ethnicity. Three iron deficiency definitions were studied: (1) combined transferrin saturation less than 10% and serum ferritin less than 15 ng/mL (HEIRS), (2) serum ferritin less than 15 ng/mL (World Health Organization [WHO]), and (3) serum ferritin less than 25 ng/mL (a threshold for iron-deficient erythropoiesis [IDE]). Results Among 62 685 women (mean [SD] age, 49.58 [14.27] years), 1957 women (3.12%) had iron deficiency according to the HEIRS definition, 4659 women (7.43%) had iron deficiency according to the WHO definition, and 9611 women (15.33%) had iron deficiency according to the IDE definition. Among 40 381 women aged 25 to 54 years, 1801 women (4.46%) had iron deficiency according to HEIRS, 4267 women (10.57%) had iron deficiency according to WHO, and 8573 women (21.23%) had iron deficiency according to IDE. Prevalence rates of iron deficiency among 2039 women aged 25 to 44 years who reported pregnancy were 5.44% (111 women) according to HEIRS, 18.05% (368 women) according to WHO, and 36.10% (736 women) according to IDE. Iron deficiency prevalence by the 3 respective definitions increased significantly in each racial and ethnic group and was significantly higher among Black and Hispanic participants than Asian and White participants. The relative iron deficiency prevalence among the 62 685 women increased 2.4-fold (95% CI, 2.3-2.5; P < .001) using the WHO definition and increased 4.9-fold (95% CI, 4.7-5.2; P < .001) using the IDE definition. Conclusions and Relevance Three definitions of iron deficiency were associated with significantly different prevalence of iron deficiency in women, regardless of self-reported age, pregnancy, or race and ethnicity. Using higher serum ferritin thresholds to define iron deficiency could lead to diagnosis and treatment of more women with iron deficiency and greater reduction of related morbidity.
Collapse
Affiliation(s)
- James C. Barton
- Department of Medicine, The University of Alabama at Birmingham, Birmingham
- Southern Iron Disorders Center, Birmingham, Alabama
| | - Howard W. Wiener
- Department of Epidemiology, School of Public Health, The University of Alabama at Birmingham, Birmingham
| | | | - Ronald T. Acton
- Southern Iron Disorders Center, Birmingham, Alabama
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham
| |
Collapse
|
14
|
Ahanchi NS, Khatami F, Llanaj E, Quezada-Pinedo HG, Dizdari H, Bano A, Glisic M, Eisenga MF, Vidal PM, Muka T. The complementary roles of iron and estrogen in menopausal differences in cardiometabolic outcomes. Clin Nutr 2024; 43:1136-1150. [PMID: 38593499 DOI: 10.1016/j.clnu.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/25/2024] [Accepted: 03/24/2024] [Indexed: 04/11/2024]
Abstract
Biological hormonal changes are frequently cited as an explanatory factor of sex and menopause differences in cardiometabolic diseases (CMD) and its associated risk factors. However, iron metabolism which varies between sexes and among women of different reproductive stages could also play a role. Recent evidence suggest that iron may contribute to CMD risk by modulating oxidative stress pathways and inflammatory responses, offering insights into the mechanistic interplay between iron and CMD development. In the current review, we provide a critical appraisal of the existing evidence on sex and menopausal differences in CMD, discuss the pitfall of current estrogen hypothesis as sole explanation, and the emerging role of iron in CMD as complementary pathway. Prior to menopause, body iron stores are lower in females as compared to males, but the increase during and after menopause, is tandem with an increased CMD risk. Importantly, basic science experiments show that an increased iron status is related to the development of type 2 diabetes (T2D), and different cardiovascular diseases (CVD). While epidemiological studies have consistently reported associations between heme iron intake and some iron biomarkers such as ferritin and transferrin saturation with the risk of T2D, the evidence regarding their connection to CVD remains controversial. We delve into the factors contributing to this inconsistency, and the limitation of relying on observational evidence, as it does not necessarily imply causation. In conclusion, we provide recommendations for future studies on evaluating the potential role of iron in elucidating the sex and menopausal differences observed in CMD.
Collapse
Affiliation(s)
- Noushin Sadat Ahanchi
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland; Graduate School for Health Sciences, University of Bern, Bern, Switzerland; Department of Internal Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Farnaz Khatami
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland; Graduate School for Health Sciences, University of Bern, Bern, Switzerland; Community Medicine Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Erand Llanaj
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hugo G Quezada-Pinedo
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland; The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics Erasmus MC-Sophia Children's Hospital University, Rotterdam, the Netherlands
| | - Helga Dizdari
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Arjola Bano
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland; Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marija Glisic
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland; Swiss Paraplegic Research, Nottwil, Switzerland
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| | - Pedro-Marques Vidal
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
15
|
Kastrati L, Groothof D, Quezada-Pinedo HG, Raeisi-Dehkordi H, Bally L, De Borst MH, Bakker SJL, Vidal PM, Eisenga MF, Muka T. Utility of iron biomarkers in differentiating menopausal status: Findings from CoLaus and PREVEND. Maturitas 2024; 179:107872. [PMID: 37952488 DOI: 10.1016/j.maturitas.2023.107872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/21/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023]
Abstract
AIM To examine the association of iron biomarkers with menopausal status and assess whether these biomarkers can help differentiate menopausal status beyond age. METHODS In this cross-sectional study we included 1679 women from the CoLaus and 2133 from the PREVEND cohorts, with CoLaus used as primary cohort and PREVEND for replication. Ferritin, transferrin, iron, and transferrin saturation (TSAT) were used to assess iron status. Hepcidin and soluble transferrin receptor were assessed only in PREVEND. Menopausal status was self-reported and defined as menopausal or non-menopausal. Logistic regressions were used to explore the association of these iron biomarkers with menopause status. Sensitivity, specificity, area under the receiver operating characteristic curves (AUC), positive and negative predictive values as well as cut-off points for the iron biomarkers were calculated. The model with the highest AUC was defined as the best. RESULTS In the CoLaus and PREVEND cohorts, respectively, 513 (30.6 %) and 988 (46.3 %) women were postmenopausal. Ferritin (OR, 2.20; 95 % CI 1.72-2.90), transferrin (OR, 0.03; 95 % CI 0.01-0.10), and TSAT (OR, 1.28; 95 % CI 1.06-1.54) were significantly associated with menopausal status in CoLaus, with the findings replicated in PREVEND. AUC of age alone was 0.971. The best model resulted from combining age, ferritin, and transferrin, with an AUC of 0.976, and sensitivity and specificity of 87.1 % and 96.5 %, respectively. Adding transferrin and ferritin to a model with age improved menopause classification by up to 7.5 %. In PREVEND, a model with age and hepcidin outperformed a model with age, ferritin, and transferrin. CONCLUSION Iron biomarkers were consistently associated with menopausal status in both cohorts, and modestly improved a model with age alone for differentiating menopause status. Our findings on hepcidin need replication.
Collapse
Affiliation(s)
- Lum Kastrati
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland; Graduate School for Health Sciences, University of Bern, Bern, Switzerland; Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism UDEM, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Dion Groothof
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713, GZ, Groningen, the Netherlands
| | - Hugo G Quezada-Pinedo
- The Generation R Study Group, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hamidreza Raeisi-Dehkordi
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lia Bally
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism UDEM, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Martin H De Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713, GZ, Groningen, the Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713, GZ, Groningen, the Netherlands
| | - Pedro-Marques Vidal
- Department of Medicine, internal medicine, Lausanne university hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713, GZ, Groningen, the Netherlands
| | - Taulant Muka
- Epistudia, 3011 Bern, Switzerland; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA.
| |
Collapse
|
16
|
Wu C, Xiao Y, Jiang Y. Associations of blood trace elements with bone mineral density: a population-based study in US adults. J Orthop Surg Res 2023; 18:827. [PMID: 37924110 PMCID: PMC10623864 DOI: 10.1186/s13018-023-04329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/29/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND This study aimed to investigate the association between blood trace elements and bone mineral density (BMD) and to determine the association between blood trace elements and the risk of low BMD/osteoporosis among US adults. METHODS We performed a cross-sectional study using data from National Health and Nutrition Examination Survey (NHANES, 2011-2016). Multivariable linear regression models were employed to assess the associations of BMD in lumbar spine (LS-BMD), pelvic (PV-BMD) and total femur (TF-BMD) with blood trace elements, including Fe, Zn, Cu, Se, Mn, Cd, Pb, Hg. Additionally, the associations of low BMD/osteoporosis with blood trace elements were also evaluated using multivariable logistic regression. RESULTS Higher blood Pb levels were found associated with decreased LS-BMD (p for trend < 0.001), PV-BMD (p for trend = 0.007), and TF-BMD (p for trend = 0.003) in female, while higher blood Se levels were associated with increased PV-BMD in female (p for trend = 0.042); no linear association between BMD and other blood trace element was observed. Also, significant associations were found between Pb levels and the prevalence of low BMD (p for trend = 0.030) and the prevalence of osteoporosis (p for trend = 0.036), while association between other blood trace elements and low BMD/osteoporosis was not observed. CONCLUSION This study provides comprehensive insight into the association between blood trace elements and BMD and supports a detrimental effect of blood Pb levels on bone mass in women. Considering our analysis from a representative US general population, further study is warranted for the extreme levels of blood trace elements on bone metabolism.
Collapse
Affiliation(s)
- Chunli Wu
- Xiangya School of Nursing, Central South University, Changsha, 410008, Hunan, China
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang, 550004, China
| | - Yao Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yuexia Jiang
- Xiangya School of Nursing, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
17
|
Zhu J, Wang Y, Rivett A, Yang G. H 2S regulation of iron homeostasis by IRP1 improves vascular smooth muscle cell functions. Cell Signal 2023; 110:110826. [PMID: 37487913 DOI: 10.1016/j.cellsig.2023.110826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
Either H2S or iron is essential for cellular processes. Abnormal metabolism of H2S and iron has increased risk for cardiovascular diseases. The aim of the present study is to examine the mutual interplay of iron and H2S signals in regulation of vascular smooth muscle cell (SMC) functions. Here we found that deficiency of cystathionine gamma-lyase (CSE, a major H2S-producing enzyme in vascular system) induced but NaHS (a H2S donor) administration attenuated iron accumulation in aortic tissues from angiotensin II-infused mice. In vitro, iron overload induced labile iron levels, promoted cell proliferation, disrupted F-actin filaments, and inhibited protein expressions of SMC-specific markers (αSMA and calponin) more significantly in SMCs from CSE knockout mice (KO-SMCs) than the cells from wild-type mice (WT-SMCs), which could be reversed by exogenously applied NaHS. In contrast, KO-SMCs were more vulnerable to iron starvation-induced cell death. Either iron overload or NaHS did not affect elastin level and gelatinolytic activity. We further found that H2S induced more aconitase activity of iron regulatory protein 1 (IRP1) but inhibited its RNA binding activity accompanied with increased protein levels of ferritin and ferriportin, which would contribute to the lower level of labile iron level inside the cells. In addition, iron was able to suppress CSE-derived H2S generation, while iron also non-enzymatically induced H2S release from cysteine. This study reveals the mutual interaction between iron and H2S signals in regulating SMC phenotypes and functions; CSE/H2S system would be a target for preventing iron metabolic disorder-related vascular diseases.
Collapse
Affiliation(s)
- Jiechun Zhu
- School of Natural Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Yuehong Wang
- School of Natural Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Alexis Rivett
- School of Natural Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Guangdong Yang
- School of Natural Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.
| |
Collapse
|
18
|
Han H, Chen Y, Zhang S, Ji X, Zhu M, Ma W, Ge H, Chu H. Association between serum ferritin and liver stiffness in adults aged ≥20 years: A cross-sectional study based on NHANES. Medicine (Baltimore) 2023; 102:e34838. [PMID: 37657022 PMCID: PMC10476712 DOI: 10.1097/md.0000000000034838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/28/2023] [Indexed: 09/03/2023] Open
Abstract
The importance of serum ferritin has been demonstrated in many liver diseases, but its relationship with liver stiffness remains unclear. The objective of this study was to investigate the association between serum ferritin levels and participants' liver stiffness measurement (LSM) in the United States population. We conducted a screening of participants from National Health and Nutrition Examination Survey (NHANES) 2017.1 to 2020.3 to ensure that participants included in this study had complete serum ferritin and LSM information. Association between the independent variable (serum ferritin) and the dependent variable (LSM) was investigated by multiple linear regression and subgroup analysis was performed to identify sensitive individuals, and we subsequently assessed whether there was a non-linear relationship between the 2 using smoothed curve fitting and threshold effect models. The final 7143 participants were included in this study. There was a positive association between participants' serum ferritin concentration and LSM, with an effect value of (β = 0.0007, 95% confidence interval (CI): 0.0002-0.0011) in the all-adjusted model. The smoothing curve and threshold effect models indicated a non-linear positive correlation between serum ferritin and LSM, which was more pronounced when serum ferritin concentration exceeded 440 ng/mL. Subsequent subgroup analysis showed that this positive correlation was more pronounced in males (β = 0.0007, 95% CI: 0.0001-0.0012), age >60 years (β = 0.00015, 95% CI: 0.0007-0.0023), black participants (β = 0.00018, 95% CI: 0.0009-0.0026), and participants with body mass index (BMI) <25 kg/m2 (β = 0.00012, 95% CI: 0.0005-0.0020). In U.S. adults, there was a positive correlation between serum ferritin levels and liver stiffness, which was more pronounced when serum ferritin exceeded 440 ng/mL. Our study suggested that regular serum ferritin testing would be beneficial in monitoring changes in liver stiffness. Male, age >60 years, black participants, and those with a BMI < 25 kg/m2 should be of greater consideration.
Collapse
Affiliation(s)
- Hao Han
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| | - Yan Chen
- Department of General Practice, Wuhu City Second People`s Hospital, Wuhu City, Anhui Province, People’s Republic of China
| | - Siqi Zhang
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| | - Xiaojuan Ji
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| | - Mingli Zhu
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| | - Wanyu Ma
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| | - Hongfeng Ge
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| | - Hailiang Chu
- Department of Hematology, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou City, Anhui Province, People’s Republic of China
| |
Collapse
|
19
|
Merlo F, Groothof D, Khatami F, Ahanchi NS, Wehrli F, Bakker SJL, Eisenga MF, Muka T. Changes in Iron Status Biomarkers with Advancing Age According to Sex and Menopause: A Population-Based Study. J Clin Med 2023; 12:5338. [PMID: 37629382 PMCID: PMC10455248 DOI: 10.3390/jcm12165338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The risk of chronic diseases increases markedly with age and after menopause. An increase in bodily iron following menopause could contribute to this phenomenon of increased risk of chronic diseases. We aimed to investigate how various iron biomarkers change with advancing age, according to sex and menopausal status. METHODS We enrolled community-dwelling individuals with available information on ferritin, transferrin, iron, hepcidin, and soluble transferrin receptor levels from the Prevention of Renal and Vascular Endstage Disease study. The association of the iron biomarkers with age, sex, and menopausal status was investigated with linear regression models. RESULTS Mean (SD) age of the 5222 individuals (2680 women [51.3%], among whom 907 [33.8%] were premenopausal, 529 [19.7%] perimenopausal, and 785 [29.3%] postmenopausal), was 53.4 (12.0) years. Iron biomarkers showed a constant increase in women throughout their life course, in some cases at older ages surpassing values in men who, in turn, showed consistently higher levels of iron status compared to women in most age categories. Ferritin, hepcidin, and transferrin saturation levels were 3.03, 2.92, and 1.08-fold (all p < 0.001) higher in postmenopausal women compared to premenopausal. CONCLUSIONS We found that iron accumulates differently depending on sex, age, and menopausal status. An increased iron status was identified in women, especially during and after menopause.
Collapse
Affiliation(s)
- Francesco Merlo
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland; (F.M.); (F.K.); (N.S.A.)
| | - Dion Groothof
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (D.G.); (S.J.L.B.); (M.F.E.)
| | - Farnaz Khatami
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland; (F.M.); (F.K.); (N.S.A.)
- Graduate School for Health Sciences, University of Bern, 3012 Bern, Switzerland
- Community Medicine Department, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Noushin Sadat Ahanchi
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland; (F.M.); (F.K.); (N.S.A.)
- Graduate School for Health Sciences, University of Bern, 3012 Bern, Switzerland
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Rue de Bugnon 21, 1005 Lausanne, Switzerland
| | - Faina Wehrli
- Dr. Risch, Lagerstrasse 30, 9470 Buchs, Switzerland;
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (D.G.); (S.J.L.B.); (M.F.E.)
| | - Michele F. Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (D.G.); (S.J.L.B.); (M.F.E.)
| | - Taulant Muka
- Epistudia, Schanzenstrasse 4a, 3008 Bern, Switzerland
| |
Collapse
|
20
|
Li GF, Gao Y, Weinberg ED, Huang X, Xu YJ. Role of Iron Accumulation in Osteoporosis and the Underlying Mechanisms. Curr Med Sci 2023; 43:647-654. [PMID: 37326889 DOI: 10.1007/s11596-023-2764-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/09/2021] [Indexed: 06/17/2023]
Abstract
Osteoporosis is prevalent in postmenopausal women. The underlying reason is mainly estrogen deficiency, but recent studies have indicated that osteoporosis is also associated with iron accumulation after menopause. It has been confirmed that some methods of decreasing iron accumulation can improve the abnormal bone metabolism associated with postmenopausal osteoporosis. However, the mechanism of iron accumulation-induced osteoporosis is still unclear. Iron accumulation may inhibit the canonical Wnt/β-catenin pathway via oxidative stress, leading to osteoporosis by decreasing bone formation and increasing bone resorption via the osteoprotegerin (OPG)/receptor activator of nuclear factor kappa-B ligand (RANKL)/receptor activator of nuclear factor kappa-B (RANK) system. In addition to oxidative stress, iron accumulation also has been reported to inhibit either osteoblastogenesis or osteoblastic function as well as to stimulate either osteoclastogenesis or osteoclastic function directly. Furthermore, serum ferritin has been widely used for the prediction of bone status, and nontraumatic measurement of iron content by magnetic resonance imaging may be a promising early indicator of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Guang-Fei Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 2015004, China
- Osteoporosis Institute of Soochow University, Suzhou, 215004, China
| | - Yan Gao
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 2015004, China
- Osteoporosis Institute of Soochow University, Suzhou, 215004, China
| | - E D Weinberg
- Department of Biology & Program in Medical Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Xi Huang
- Department of Environmental Medicine, New York University, School of Medicine, New York, NY, 10016, USA
| | - You-Jia Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 2015004, China.
- Osteoporosis Institute of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
21
|
Yehia A, Sousa RAL, Abulseoud OA. Sex difference in the association between blood alcohol concentration and serum ferritin. Front Psychiatry 2023; 14:1230406. [PMID: 37547205 PMCID: PMC10401063 DOI: 10.3389/fpsyt.2023.1230406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction The sex difference in alcohol use disorder (AUD) is ingrained in distinctive neurobiological responses between men and women, which necessitates further investigation for a more tailored management. Methods Minding the findings of iron dysregulation in AUD and the sex difference in iron homeostasis in multiple physiological and pathological settings, we examined the sex difference in the association between serum ferritin and blood alcohol concentration (BAC) in intoxicated males (n = 125) and females (n = 59). We included patients with both serum ferritin tested of any value and a BAC above the level of detection during the same hospital admission period. We investigated sex difference in the relationship between BAC, serum ferritin and liver enzymes in intoxicated critically ill and noncritically ill patients. Results We found a negative association between serum ferritin and BAC in critically ill, intoxicated females [R2 = 0.44, F(1,14) = 11.02, p = 0.005], with much attenuated serum ferritin in females compared to their male counterparts (194.5 ± 280.4 vs. 806.3 ± 3405.7 ng/L, p = 0.002). We found a positive association between serum ferritin and liver enzymes [alanine transaminase (ALT) and aspartate transferase (AST)] in critically ill intoxicated females [ALT: R2 = 0.48, F(1,10) = 9.1, p = 0.013; AST: R2 = 0.68, F(1,10) = 21.2, p = 0.001] and in noncritically ill intoxicated males [ALT: R2 = 0.1, F(1,83) = 9.4, p = 0.003; AST: R2 = 0.1, F(1,78) = 10.5, p = 0.002]. The effect of BAC on serum ferritin was not mediated by ALT [indirect effect: (B = 0.13, p = 0.1)]. We also found a significant effect of sex, anemia, intensive care unit (ICU) admission and mortality on serum ferritin. Discussion Our results suggest that high BAC in intoxicated female patients is associated with attenuated serum ferritin levels, questioning the role of low serum ferritin in female vulnerability to alcohol.
Collapse
Affiliation(s)
- Asmaa Yehia
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ricardo A L Sousa
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Osama A Abulseoud
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| |
Collapse
|
22
|
Toljić B, Milašin J, De Luka SR, Dragović G, Jevtović D, Maslać A, Ristić-Djurović JL, Trbovich AM. HIV-Infected Patients as a Model of Aging. Microbiol Spectr 2023; 11:e0053223. [PMID: 37093018 PMCID: PMC10269491 DOI: 10.1128/spectrum.00532-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/01/2023] [Indexed: 04/25/2023] Open
Abstract
We appraised the relationship between the biological and the chronological age and estimated the rate of biological aging in HIV-infected patients. Two independent biomarkers, the relative telomere length and iron metabolism parameters, were analyzed in younger (<35) and older (>50) HIV-infected and uninfected patients (control group). In our control group, telomeres of younger patients were significantly longer than telomeres of older ones. However, in HIV-infected participants, the difference in the length of telomeres was lost. By combining the length of telomeres with serum iron, ferritin, and transferrin iron-binding capacity, a new formula for determination of the aging process was developed. The life expectancy of the healthy population was related to their biological age, and HIV-infected patients were biologically older. The effect of antiretroviral HIV drug therapies varied with respect to the biological aging process. IMPORTANCE This article is focused on the dynamics of human aging. Moreover, its interdisciplinary approach is applicable to various systems that are aging.
Collapse
Affiliation(s)
- Boško Toljić
- School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Milašin
- School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | | | |
Collapse
|
23
|
Bao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, et alBao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, Wang S, Wang X, Wang X, Wang YJ, Wang Y, Wong CCL, Xiang AP, Xiao Y, Xie Z, Xu D, Ye J, Yue R, Zhang C, Zhang H, Zhang L, Zhang W, Zhang Y, Zhang YW, Zhang Z, Zhao T, Zhao Y, Zhu D, Zou W, Pei G, Liu GH. Biomarkers of aging. SCIENCE CHINA. LIFE SCIENCES 2023; 66:893-1066. [PMID: 37076725 PMCID: PMC10115486 DOI: 10.1007/s11427-023-2305-0] [Show More Authors] [Citation(s) in RCA: 163] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023]
Abstract
Aging biomarkers are a combination of biological parameters to (i) assess age-related changes, (ii) track the physiological aging process, and (iii) predict the transition into a pathological status. Although a broad spectrum of aging biomarkers has been developed, their potential uses and limitations remain poorly characterized. An immediate goal of biomarkers is to help us answer the following three fundamental questions in aging research: How old are we? Why do we get old? And how can we age slower? This review aims to address this need. Here, we summarize our current knowledge of biomarkers developed for cellular, organ, and organismal levels of aging, comprising six pillars: physiological characteristics, medical imaging, histological features, cellular alterations, molecular changes, and secretory factors. To fulfill all these requisites, we propose that aging biomarkers should qualify for being specific, systemic, and clinically relevant.
Collapse
Affiliation(s)
- Hainan Bao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Jiani Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Min Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yingjie Ding
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junlin Feng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mengmeng Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Chuting He
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Yujuan Jia
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Haiping Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Ying Jing
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyi Li
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Qinhao Liang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jianwei Lv
- School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jingyi Ma
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kehang Mao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China
| | - Jiawei Nie
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Siyuan Wang
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xuan Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuhan Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Rimo Wu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Kai Xia
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fu-Hui Xiao
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Xu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Haoteng Yan
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuanxin Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Le Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiwei Zhang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China
| | - Wenwan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qingchen Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China.
| | - Zhongwei Cao
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou, 510000, China.
| | - Hou-Zao Chen
- Department of Biochemistryand Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China.
| | - Weimin Ci
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Feng Gao
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| | - Qing-Peng Kong
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Xin Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Baohua Liu
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518060, China.
| | - Feng Liu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South Unversity, Changsha, 410011, China.
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China.
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, 300000, China.
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Institute of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
| | - Yong Liu
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Shuai Ma
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jing Nie
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yaojin Peng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Center for Aging and Cancer, Hainan Medical University, Haikou, 571199, China.
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China.
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China.
| | - Si Wang
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Xia Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Xiaoning Wang
- Institute of Geriatrics, The second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Yunfang Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Catherine C L Wong
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China.
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, 100101, China.
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Cuntai Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China.
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Liang Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yong Zhang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Zhuohua Zhang
- Key Laboratory of Molecular Precision Medicine of Hunan Province and Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Dahai Zhu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Gang Pei
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, 200070, China.
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
24
|
Zheng H, Yang F, Deng K, Wei J, Liu Z, Zheng YC, Xu H. Relationship between iron overload caused by abnormal hepcidin expression and liver disease: A review. Medicine (Baltimore) 2023; 102:e33225. [PMID: 36930080 PMCID: PMC10019217 DOI: 10.1097/md.0000000000033225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/16/2023] [Indexed: 03/18/2023] Open
Abstract
Iron is essential to organisms, the liver plays a vital role in its storage. Under pathological conditions, iron uptake by the intestine or hepatocytes increases, allowing excess iron to accumulate in liver cells. When the expression of hepcidin is abnormal, iron homeostasis in humans cannot be regulated, and resulting in iron overload. Hepcidin also regulates the release of iron from siderophores, thereby regulating the concentration of iron in plasma. Important factors related to hepcidin and systemic iron homeostasis include plasma iron concentration, body iron storage, infection, inflammation, and erythropoietin. This review summarizes the mechanism and regulation of iron overload caused by hepcidin, as well as related liver diseases caused by iron overload and treatment.
Collapse
Affiliation(s)
- Haoran Zheng
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Fan Yang
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Kaige Deng
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaxin Wei
- Department of Emergency, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhenting Liu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Yong-Chang Zheng
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Haifeng Xu
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Övermöhle C, Waniek S, Rimbach G, Weber KS, Lieb W. Plasma Ferritin Concentrations in the General Population: A Cross-Sectional Analysis of Anthropometric, Metabolic, and Dietary Correlates. J Nutr 2023; 153:1524-1533. [PMID: 36906150 DOI: 10.1016/j.tjnut.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Elevated concentrations of ferritin seem to be detrimental to human health while being quite common in the elderly. Data on dietary, anthropometric, and metabolic correlates of circulating ferritin levels in the elderly are scant. OBJECTIVES We aimed to identify a dietary pattern, anthropometric, and metabolic traits associated with plasma ferritin status in an elderly cohort (n = 460, 57% male, age: 66 ± 12 y) from Northern Germany. METHODS Plasma ferritin levels were measured by immunoturbidimetry. Reduced rank regression (RRR) yielded a dietary pattern explaining 13% of the variation in circulating ferritin concentrations. Cross-sectional associations of anthropometric and metabolic traits with plasma ferritin concentrations were assessed using multivariable-adjusted linear regression analysis. Restricted cubic spline regression was used to identify nonlinear associations. RESULTS The RRR pattern was characterized by a high intake of potatoes, certain vegetables, beef, pork, processed meat, fats (frying and animal fat), and beer and a low intake of snacks, representing elements of the traditional German diet. BMI, waist circumference, and CRP were directly, HDL cholesterol inversely, and age nonlinearly associated with plasma ferritin concentrations (all P < 0.05). After additional adjustment for CRP, only the association of ferritin with age remained statistically significant. CONCLUSION Higher plasma ferritin concentrations were associated with a traditional German dietary pattern. The associations of ferritin with unfavorable anthropometric traits and low HDL cholesterol were rendered statistically nonsignificant upon additional adjustment for chronic systemic inflammation (CRP), suggesting that these associations were largely driven by the proinflammatory role of ferritin (an acute-phase reactant). J Nutr 20xx;x:xx.
Collapse
Affiliation(s)
- Cara Övermöhle
- Institute of Epidemiology, Kiel University, Kiel, Germany
| | - Sabina Waniek
- Institute of Epidemiology, Kiel University, Kiel, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | | | - Wolfgang Lieb
- Institute of Epidemiology, Kiel University, Kiel, Germany
| |
Collapse
|
26
|
Chang VC, Cotterchio M, Kotsopoulos J, Bondy SJ. Iron Status and Associated Factors among Canadian Women: Results from the Canadian Health Measures Survey. J Nutr 2023; 153:781-797. [PMID: 36788041 DOI: 10.1016/j.tjnut.2022.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/25/2022] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Iron deficiency and overload may negatively impact women's health. There has been limited assessment of iron status and its associated factors among Canadian women. OBJECTIVES This study investigated associations of various sociodemographic, lifestyle, medication, and dietary factors with body iron stores among pre- and postmenopausal women in Canada. METHODS Analyses were conducted using cross-sectional, nationally representative survey and biomarker data from women aged 20-79 y (n = 6362) in the Canadian Health Measures Survey (2009-2017). Body iron stores were assessed by measuring serum concentrations of ferritin (SF). Information on potential correlates was collected during an in-home interview. Multivariable linear regression analyses were performed to evaluate associations with SF concentration, and logistic regression was used to estimate associations with iron deficiency (SF <15 μg/L) or elevated iron stores (SF >150 μg/L). RESULTS Geometric mean SF concentrations were significantly higher in postmenopausal than in premenopausal women (73.2 versus 33.8 μg/L; P < 0.001). The prevalence of iron deficiency among pre- and postmenopausal women was 16.0% and 4.0%, respectively, whereas that of elevated iron stores was 2.7% and 21.0%, respectively. After simultaneous adjustment for multiple factors, including high-sensitivity CRP (inflammation marker), we found that age, East/Southeast Asian (versus White) race/ethnicity, alcohol, and red meat consumption were positively associated with SF concentration among pre- and postmenopausal women. In addition, aspirin use and dairy consumption were inversely associated with SF concentration among postmenopausal women only. Similar patterns were observed for associations with elevated iron stores among postmenopausal women, whereas higher grain consumption was associated with an increased prevalence of iron deficiency among premenopausal women. CONCLUSIONS Sociodemographic, lifestyle, medication, and dietary factors are correlated with iron status determined by SF concentration among Canadian women. The findings may have implications for intervention strategies aimed at optimizing body iron stores in pre- and postmenopausal women.
Collapse
Affiliation(s)
- Vicky C Chang
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Prevention and Cancer Control, Ontario Health (Cancer Care Ontario), Toronto, ON, Canada.
| | - Michelle Cotterchio
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Prevention and Cancer Control, Ontario Health (Cancer Care Ontario), Toronto, ON, Canada
| | - Joanne Kotsopoulos
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| | - Susan J Bondy
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Alfaro-Magallanes VM, Romero-Parra N, Barba-Moreno L, Rael B, Benito PJ, Díaz ÁE, Cupeiro R, Peinado AB. Serum iron availability, but not iron stores, is lower in naturally menstruating than in oral contraceptive athletes. Eur J Sport Sci 2023; 23:231-240. [PMID: 34904534 DOI: 10.1080/17461391.2021.2018503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This study measured serum markers of iron status in naturally menstruating and oral contraceptive (OC) athletes during the main hormonal milieus of these two profiles to identify potential differences confounding the diagnosis of iron deficiency in female athletes. Resting blood samples were collected from 36 naturally menstruating athletes during the early-follicular phase (EFP), mid- late-follicular phase (MLFP) and mid-luteal phase (MLP) of the menstrual cycle. Simultaneously, blood samples were collected from 24 OC athletes during the withdrawal and active-pill phase of the OC cycle. Serum iron, ferritin, transferrin, transferrin saturation (TSAT), C-reactive protein (CRP), interleukin-6 and sex hormones were analyzed. Naturally menstruating athletes showed lower levels of TSAT, iron and transferrin than OC athletes when comparing the bleeding phase of both profiles (p<0.05) as well as when comparing all analyzed phases of the menstrual cycle to the active pill phase of the OC cycle (p<0.05). Interestingly, only lower transferrin was found during MLFP and MLP compared to the withdrawal phase of the OC cycle (p>0.05), with all other iron markers showing no differences (p>0.05). Intracycle variations were also found within both types of cycle, presenting reduced TSAT and iron during menstrual bleeding phases (p<0.05). In conclusion, in OC athletes, serum iron availability, but not serum ferritin, seems higher than in naturally menstruating ones. However, such differences are lost when comparing the MLFP and MLP of the menstrual cycle with the withdrawal phase of the OC cycle. This should be considered in the assessment of iron status in female athletes.Highlights Naturally menstruating athletes present lower TSAT, iron and transferrin in all analyzed phases of the menstrual cycle compared to OC athletes during their active pill phase. However, both the mid-late follicular and mid-luteal phases of the menstrual cycle do not differ from the withdrawal phase of the oral contraceptive cycle.Intracycle variations are found for TSAT and iron in both naturally menstruating and oral contraceptive athletes, which are mainly driven by a reduction in TSAT and iron during menstrual bleeding phases.As serum iron availability changes significantly as a function of the athlete's hormonal status, it should be considered in the assessment of the athlete's iron status as well as standardise the phase of the menstrual cycle in which to assess iron markers to avoid misdiagnosis or misleading results.In contrast, the assessment of iron stores through serum ferritin is substantially stable and the athlete's hormonal status does not seem to be of relevance for this purpose.
Collapse
Affiliation(s)
- Víctor M Alfaro-Magallanes
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Nuria Romero-Parra
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Laura Barba-Moreno
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Beatriz Rael
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Pedro J Benito
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Ángel E Díaz
- Clinical laboratory. National Center of Sport Medicine. Health and Sports Department, AEPSAD, Madrid, Spain
| | - Rocío Cupeiro
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Ana B Peinado
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | -
- LFE Research Group. Department of Health and Human Performance. Faculty of Physical Activity and Sport Sciences (INEF), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| |
Collapse
|
28
|
Li Z, Li D, Chen R, Gao S, Xu Z, Li N. Cell death regulation: A new way for natural products to treat osteoporosis. Pharmacol Res 2023; 187:106635. [PMID: 36581167 DOI: 10.1016/j.phrs.2022.106635] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/11/2022] [Accepted: 12/24/2022] [Indexed: 12/27/2022]
Abstract
Osteoporosis is a common metabolic bone disease that results from the imbalance of homeostasis within the bone. Intra-bone homeostasis is dependent on a precise dynamic balance between bone resorption by osteoclasts and bone formation by mesenchymal lineage osteoblasts, which comprises a series of complex and highly standardized steps. Programmed cell death (PCD) (e.g., apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis) is a cell death process that involves a cascade of gene expression events with tight structures. These events play a certain role in regulating bone metabolism by determining the fate of bone cells. Moreover, existing research has suggested that natural products derived from a wide variety of dietary components and medicinal plants modulate the PCDs based on different mechanisms, which show great potential for the prevention and treatment of osteoporosis, thus revealing the emergence of more acceptable complementary and alternative drugs with lower costs, fewer side effects and more long-term application. Accordingly, this review summarizes the common types of PCDs in the field of osteoporosis. Moreover, from the perspective of targeting PCDs, this review also discussed the roles of currently reported natural products in the treatment of osteoporosis and the involved mechanisms. Based on this, this review provides more insights into new molecular mechanisms of osteoporosis and provides a reference for developing more natural anti-osteoporosis drugs in the future.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Renchang Chen
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Shang Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
29
|
Khatami F, Muka T, Groothof D, de Borst MH, Buttia C, van Hassel G, Baumgartner I, Kremer D, Bakker SJL, Bano A, Eisenga MF. Sex and N-terminal pro B-type natriuretic peptide: The potential mediating role of iron biomarkers. Front Cardiovasc Med 2022; 9:897148. [PMID: 36451923 PMCID: PMC9703058 DOI: 10.3389/fcvm.2022.897148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/24/2022] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Levels of N-terminal pro B-type natriuretic peptide (NT-proBNP), a marker of heart failure and cardiovascular risk, are generally higher in women than men. We explored whether iron biomarkers mediate sex differences in NT-proBNP levels. METHODS We included 5,343 community-dwelling individuals from the Prevention of Renal and Vascular Endstage Disease study. With linear regression analyses, we investigated the association of sex and iron biomarkers with NT-proBNP levels, independent of adjustment for potential confounders. The assessed iron biomarkers included ferritin, transferrin saturation (TSAT), hepcidin, and soluble transferrin receptor (sTfR). Next, we performed mediation analyses to investigate to which extent iron biomarkers influence the association between sex and NT-proBNP. RESULTS Of the included 5,343 participants, the mean standard deviation age was 52.2 ± 11.6 years and 52% were females. After adjustment for potential confounders, women compared to men, had higher NT-proBNP (β = 0.31; 95%CI = 0.29, 0.34), but lower ferritin (β = -0.37; 95%CI = -0.39, -0.35), hepcidin (β = -0.22, 95%CI = -0.24, -0.20), and TSAT (β = -0.07, 95% CI = -0.08, -0.06). Lower ferritin (β = -0.05, 95%CI = -0.08, -0.02), lower hepcidin (β = -0.04, 95%CI = -0.07, -0.006), and higher TSAT (β = 0.07; 95%CI = 0.01, 0.13) were associated with higher NT-proBNP. In mediation analyses, ferritin and hepcidin explained 6.5 and 3.1% of the association between sex and NT-proBNP, respectively, while TSAT minimally suppressed (1.9%) this association. CONCLUSION Our findings suggest that iron biomarkers marginally explain sex differences in levels of NT-proBNP. Future studies are needed to explore causality and potential mechanisms underlying these pathways.
Collapse
Affiliation(s)
- Farnaz Khatami
- Institute of Social and Preventive Medicine (ISPM), Graduate School of Health Sciences, University of Bern, Bern, Switzerland
- Department of Community Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Taulant Muka
- Institute of Social and Preventive Medicine (ISPM), Graduate School of Health Sciences, University of Bern, Bern, Switzerland
- Epistudia, Bern, Switzerland
| | - Dion Groothof
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| | - Martin H. de Borst
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| | - Chepkoech Buttia
- Institute of Social and Preventive Medicine (ISPM), Graduate School of Health Sciences, University of Bern, Bern, Switzerland
| | - Gaston van Hassel
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| | - Iris Baumgartner
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| | - Arjola Bano
- Institute of Social and Preventive Medicine (ISPM), Graduate School of Health Sciences, University of Bern, Bern, Switzerland
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Michele F. Eisenga
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, Netherlands
| |
Collapse
|
30
|
Sze SCW, Zhang L, Zhang S, Lin K, Ng TB, Ng ML, Lee KF, Lam JKW, Zhang Z, Yung KKL. Aberrant Transferrin and Ferritin Upregulation Elicits Iron Accumulation and Oxidative Inflammaging Causing Ferroptosis and Undermines Estradiol Biosynthesis in Aging Rat Ovaries by Upregulating NF-Κb-Activated Inducible Nitric Oxide Synthase: First Demonstration of an Intricate Mechanism. Int J Mol Sci 2022; 23:ijms232012689. [PMID: 36293552 PMCID: PMC9604315 DOI: 10.3390/ijms232012689] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
We report herein a novel mechanism, unraveled by proteomics and validated by in vitro and in vivo studies, of the aberrant aging-associated upregulation of ovarian transferrin and ferritin in rat ovaries. The ovarian mass and serum estradiol titer plummeted while the ovarian labile ferrous iron and total iron levels escalated with age in rats. Oxidative stress markers, such as nitrite/nitrate, 3-nitrotyrosine, and 4-hydroxy-2-nonenal, accumulated in the aging ovaries due to an aberrant upregulation of the ovarian transferrin, ferritin light/heavy chains, and iron regulatory protein 2(IRP2)-mediated transferrin receptor 1 (TfR1). Ferritin inhibited estradiol biosynthesis in ovarian granulosa cells in vitro via the upregulation of a nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and p65/p50-induced oxidative and inflammatory factor inducible nitric oxide synthase (iNOS). An in vivo study demonstrated how the age-associated activation of NF-κB induced the upregulation of iNOS and the tumor necrosis factor α (TNFα). The downregulation of the keap1-mediated nuclear factor erythroid 2-related factor 2 (Nrf2), that induced a decrease in glutathione peroxidase 4 (GPX4), was observed. The aberrant transferrin and ferritin upregulation triggered an iron accumulation via the upregulation of an IRP2-induced TfR1. This culminates in NF-κB-iNOS-mediated ovarian oxi-inflamm-aging and serum estradiol decrement in naturally aging rats. The iron accumulation and the effect on ferroptosis-related proteins including the GPX4, TfR1, Nrf2, Keap1, and ferritin heavy chain, as in testicular ferroptosis, indicated the triggering of ferroptosis. In young rats, an intraovarian injection of an adenovirus, which expressed iron regulatory proteins, upregulated the ovarian NF-κB/iNOS and downregulated the GPX4. These novel findings have contributed to a prompt translational research on the ovarian aging-associated iron metabolism and aging-associated ovarian diseases.
Collapse
Affiliation(s)
- Stephen Cho Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Correspondence: (S.C.W.S.); (K.K.L.Y.); Tel.: +852-34112318 (S.C.W.S.); Tel.: +852-34117060 (K.K.L.Y.)
| | - Liang Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China
| | - Shiqing Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou 999077, China
| | - Kaili Lin
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- School of Public Health, Guangzhou Medical University, Guangzhou 999077, China
| | - Tzi Bun Ng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR 999077, China
| | - Man Ling Ng
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, HKU, Pokfulam, Hong Kong SAR 999077, China
| | - Jenny Ka Wing Lam
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
- Department of Pharmacology & Pharmacy, LKS Faculty of Medicine, HKU, Pokfulam, Hong Kong SAR 999077, China
| | - Zhang Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Correspondence: (S.C.W.S.); (K.K.L.Y.); Tel.: +852-34112318 (S.C.W.S.); Tel.: +852-34117060 (K.K.L.Y.)
| |
Collapse
|
31
|
Abstract
In this review, we discuss the effects on androgens on the haemopoietic system, focussing largely on the effects of testosterone on erythropoiesis. Stimulation of erythropoiesis is one of the most consistent effects of testosterone treatment observed in clinical trials. In men with anaemia this effect can be beneficial. Conversely, erythrocytosis is one of the most common adverse effects of testosterone treatment with a relative risk of 8.14 (95% CI: 1.87-35.40) estimated by a recent meta-analysis of randomised placebo controlled clinical trials. A reduction in haemoglobin is commonly seen in men receiving androgen deprivation therapy for prostate cancer, and in transwomen receiving gender affirming therapy to reduce serum testosterone. While mechanisms by which androgens regulate erythropoiesis are not fully understood, it is likely that effects on erythropoietic progenitor cells and erythropoietin are involved, with secondary effects on iron metabolism. In contrast, whether androgens exert clinically relevant effects on white blood cells and on platelets requires further study.
Collapse
Affiliation(s)
- Annabelle M Warren
- Department of Endocrinology, Austin Health and University of Melbourne, Australia.
| | - Mathis Grossmann
- Department of Endocrinology, Austin Health and University of Melbourne, Australia.
| |
Collapse
|
32
|
Sex Differences in Cardiomyopathy. CURRENT CARDIOVASCULAR RISK REPORTS 2022. [DOI: 10.1007/s12170-022-00700-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Liu M, Yang C, Chu Q, Wang J, Wang Q, Kong F, Sun G. High serum levels of ferritin may predict poor survival and walking ability for patients with hip fractures: a propensity score matching study. Biomark Med 2022; 16:857-866. [PMID: 35704298 DOI: 10.2217/bmm-2022-0160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To identify the relationship between ferritin and outcomes for patients with hip fractures. Patients & methods: All patients with hip fractures presenting between May 2017 and January 2021 were included. Univariate and multivariate analyses were performed to determine the risk factors for 1-year survival. Propensity score matching (PSM) was performed for groups divided by ferritin levels. Results: A total of 165 patients were included of whom 28 died during the first year after surgery. Ferritin levels differed significantly between groups divided by 1-year survival. High ferritin (≥308.5 ng/ml) was related to poor 1-year survival and 6-month and 1-year independent walking rate. Conclusion: High ferritin (≥308.5 ng/ml) may predict poor survival and free-walking abilities after surgery for patients with hip fractures.
Collapse
Affiliation(s)
- Mingchong Liu
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Chensong Yang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qining Chu
- Emergency Trauma Center, Nanyang Second General Hospital, No 66, East Jianshe Road, Nanyang, 473000, China
| | - Jiansong Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qidong Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fanyu Kong
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Guixin Sun
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
34
|
Lymperaki E, Stalika E, Tzavelas G, Tormpantoni E, Samara D, Vagdatli E, Tsamesidis I. The Clinical Utility of ABO and RHD Systems as Potential Indicators of Health Status, a Preliminary Study in Greek Population. Clin Pract 2022; 12:406-418. [PMID: 35735664 PMCID: PMC9221977 DOI: 10.3390/clinpract12030045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/16/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
Objective: The objective of this study is to further highlight the differences between different ABO blood groups and Rhesus types with health biomarkers. Methods: In total 150 active healthy blood donors participated in our study comprising of 80 males from 19–61 years and 70 females aged from 21 to 64. Participants carrying blood group A were 55 individuals, blood group B 32, blood group O 51, and blood group AB 12, RHD+ 132, and RHD- 18. All the volunteer regular blood donors were selected recognizing them as a healthy population excluding drug and supplements intake. Their blood samples were analyzed just before blood donation for biochemical, hematological, and antioxidant markers. Statistical computations were performed using the SPSS tool, specifically, the one-way ANOVA test, Chi-square statistics, and logistic regression were used as statistical models. Results: O blood donors presented better iron absorption and the worst lipid profile. Indeed, a significant trend of high atheromatic index values revealed an increased risk for hyperlipidemia, in contrast with blood group A presenting a better lipid profile with lower atheromatic index values. There was also a gender related association for blood group A compared with O that was further highlighted using binary logistic regression. Conclusion: In this study, a significant difference was observed among the ABO blood groups in several of the examined biochemical and hematological biomarkers. O blood group appeared different behavior in comparison to all the tested blood groups and furthermore the RHD-group presented a better lipid profile in comparison to the RHD+ group. In order to obtain a more comprehensive view of the correlation between the ABO blood group and biochemical markers, further studies are required.
Collapse
Affiliation(s)
- Evgenia Lymperaki
- Department of Biomedical Sciences, International Hellenic University, 57001 Thessaloniki, Greece;
| | - Evangelia Stalika
- Lab of Computing and Medical Informatics, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - George Tzavelas
- Department of Statistics and Insurance Science, University of Piraeus, 18534 Piraeus, Greece;
| | - Efthymia Tormpantoni
- Blood Bank Section, Naousa General Hospital, 59200 Naousa, Greece; (E.T.); (D.S.)
| | - Diana Samara
- Blood Bank Section, Naousa General Hospital, 59200 Naousa, Greece; (E.T.); (D.S.)
| | - Eleni Vagdatli
- Laboratory of Biopathology, Hippokratio General Hospital, 54642 Thessaloniki, Greece;
| | - Ioannis Tsamesidis
- Department of Biomedical Sciences, International Hellenic University, 57001 Thessaloniki, Greece;
- Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-699-631-12-60
| |
Collapse
|
35
|
Cai C, Hu W, Chu T. Interplay Between Iron Overload and Osteoarthritis: Clinical Significance and Cellular Mechanisms. Front Cell Dev Biol 2022; 9:817104. [PMID: 35096841 PMCID: PMC8795893 DOI: 10.3389/fcell.2021.817104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/28/2021] [Indexed: 01/15/2023] Open
Abstract
There are multiple diseases or conditions such as hereditary hemochromatosis, hemophilia, thalassemia, sickle cell disease, aging, and estrogen deficiency that can cause iron overload in the human body. These diseases or conditions are frequently associated with osteoarthritic phenotypes, such as progressive cartilage degradation, alterations in the microarchitecture and biomechanics of the subchondral bone, persistent joint inflammation, proliferative synovitis, and synovial pannus. Growing evidences suggest that the conditions of pathological iron overload are associated with these osteoarthritic phenotypes. Osteoarthritis (OA) is an important complication in patients suffering from iron overload-related diseases and conditions. This review aims to summarize the findings and observations made in the field of iron overload-related OA while conducting clinical and basic research works. OA is a whole-joint disease that affects the articular cartilage lining surfaces of bones, subchondral bones, and synovial tissues in the joint cavity. Chondrocytes, osteoclasts, osteoblasts, and synovial-derived cells are involved in the disease. In this review, we will elucidate the cellular and molecular mechanisms associated with iron overload and the negative influence that iron overload has on joint homeostasis. The promising value of interrupting the pathologic effects of iron overload is also well discussed for the development of improved therapeutics that can be used in the field of OA.
Collapse
Affiliation(s)
- Chenhui Cai
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tongwei Chu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
36
|
Burton LH, Afzali MF, Radakovich LB, Campbell MA, Culver LA, Olver CS, Santangelo KS. Systemic administration of a pharmacologic iron chelator reduces cartilage lesion development in the Dunkin-Hartley model of primary osteoarthritis. Free Radic Biol Med 2022; 179:47-58. [PMID: 34923104 PMCID: PMC8760171 DOI: 10.1016/j.freeradbiomed.2021.12.257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Iron has been emerging as a key contributor to aging-associated, chronic disorders due to the propensity for generating reactive oxygen species. To date, there are a limited number of publications exploring the role of iron in the pathogenesis of primary/age-related osteoarthritis (OA). The objective of this study was to determine whether reduced iron via pharmacologic iron chelation with deferoxamine (DFO) affected the development and/or severity of cartilage lesions in a primary OA model. At 12-weeks-of-age, 15 male Dunkin-Hartley guinea pigs received either 46 mg/kg DFO (n = 8) or vehicle control (n = 7) injected subcutaneously twice daily for five days each week. Movement changes, captured via overhead enclosure monitoring, were also determined. Termination occurred at 30-weeks-of-age. Iron was quantified in serum, urine, liver, and femoral head articular cartilage. Left knees were evaluated for: structural changes using histopathology guidelines; and immunohistochemistry. Gene expression analysis was conducted on right knee articular cartilage. DFO reduced iron levels in femoral head articular cartilage (p = 0.0006) and liver (p = 0.02), and increased iron within urine (p = 0.04) and serum (p = 0.0009). Mobility of control animals declined, while the DFO group maintained activity levels similar to the first month of treatment (p = 0.05). OA-associated cartilage lesions were reduced in knees of DFO animals (p = 0.0001), with chondrocyte hypocellularity a key histologic difference between groups (p < 0.0001). DFO-receiving animals had increased immunostaining for phosphorylated adenosine monophosphate activated protein kinase alpha within knee articular cartilage; lower transcript counts of several proapoptotic genes (p = 0.04-0.0004) and matrix-degrading enzymes (p = 0.02-<0.0001), and increased expression of the anti-apoptotic gene Bcl-2 (p < 0.0001) and a tissue inhibitor of matrix-metalloproteinases (p = 0.03) were also observed. These results suggest that iron chelation delayed the progression of primary OA in an animal model and could hold potential as a translational intervention. These findings provide expanded insight into factors that may contribute to the pathogenesis of primary OA.
Collapse
Affiliation(s)
- Lindsey H Burton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Maryam F Afzali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Lauren B Radakovich
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Margaret A Campbell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Lauren A Culver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Christine S Olver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Kelly S Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
37
|
Yu YC, Luu HN, Wang R, Thomas CE, Glynn NW, Youk AO, Behari J, Yuan JM. Serum Biomarkers of Iron Status and Risk of Hepatocellular Carcinoma Development in Patients with Nonalcoholic Fatty Liver Disease. Cancer Epidemiol Biomarkers Prev 2022; 31:230-235. [PMID: 34649958 PMCID: PMC9204666 DOI: 10.1158/1055-9965.epi-21-0754] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/27/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become a major contributor to the rising incidence of hepatocellular carcinoma (HCC) in the United States and other developed countries. Iron, an essential metal primarily stored in hepatocytes, may play a role in the development of NAFLD-related HCC. Epidemiologic data on iron overload without hemochromatosis in relation to HCC are sparse. This study aimed to examine the associations between serum biomarkers of iron and the risk of HCC in patients with NAFLD. METHODS We identified 18,569 patients with NAFLD using the University of Pittsburgh Medical Center electronic health records from 2004 through 2018. After an average 4.34 years of follow-up, 244 patients developed HCC. Cox proportional hazard regression was used to calculate hazard ratios (HR) and 95% confidence intervals (CI) of HCC incidence associated with elevated levels of iron biomarkers with adjustment for age, sex, race, body mass index, history of diabetes, and tobacco smoking. RESULTS The HRs (95% CIs) of HCC for clinically defined elevation of serum iron and transferrin saturation were 2.91 (1.34-6.30) and 2.02 (1.22-3.32), respectively, compared with their respective normal range. No statistically significant association was observed for total iron-binding capacity or serum ferritin with HCC risk. CONCLUSIONS Elevated levels of serum iron and transferrin saturation were significantly associated with increased risk of HCC among patients with NAFLD without hemochromatosis or other major underlying causes of chronic liver diseases. IMPACT Clinical surveillance of serum iron level may be a potential strategy to identify patients with NAFLD who are at high risk for HCC.
Collapse
Affiliation(s)
- Yi-Chuan Yu
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hung N Luu
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Renwei Wang
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Claire E Thomas
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nancy W Glynn
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ada O Youk
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jaideep Behari
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
38
|
Association of Habitual Dietary Intake with Liver Iron-A Population-Based Imaging Study. Nutrients 2021; 14:nu14010132. [PMID: 35011009 PMCID: PMC8746950 DOI: 10.3390/nu14010132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 01/04/2023] Open
Abstract
Iron-related disorders of the liver can result in serious health conditions, such as liver cirrhosis. Evidence on the role of modifiable lifestyle factors like nutrition in liver iron storage is lacking. Thus, we aimed to assess the association of habitual diet with liver iron content (LIC). We investigated 303 participants from the population-based KORA-MRI study who underwent whole-body magnetic resonance imaging (MRI). Dietary habits were evaluated using repeated 24 h food lists and a food frequency questionnaire. Sex-stratified multiple linear regression models were applied to quantify the association between nutrition variables of interest and LIC, adjusting for liver fat content (LFC), energy intake, and age. Mean age of participants was 56.4 ± 9.0 years and 44.2% were female. Mean LIC was 1.23 ± 0.12 mg/g dry weight, with higher values in men than in women (1.26 ± 0.13 and 1.20 ± 0.10 mg/g, p < 0.001). Alcohol intake was positively associated with LIC (men: β = 1.94; women: β = 4.98, p-values < 0.03). Significant negative associations with LIC were found for fiber (β = −5.61, p < 0.001) and potassium (β = −0.058, p = 0.034) for female participants only. Furthermore, LIC was highly correlated with liver fat content in both sexes. Our findings suggests that there are sex-specific associations of habitual dietary intake and LIC. Alcohol, fiber, and potassium may play a considerable role in liver iron metabolism.
Collapse
|
39
|
Costa SA, Ribeiro CCC, Moreira ARO, Carvalho Souza SDF. High serum iron markers are associated with periodontitis in post-menopausal women: A population-based study (NHANES III). J Clin Periodontol 2021; 49:221-229. [PMID: 34879443 DOI: 10.1111/jcpe.13580] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/11/2021] [Accepted: 11/25/2021] [Indexed: 11/26/2022]
Abstract
AIM To investigate the association between increased serum markers of iron (ferritin and transferrin saturation) and the severity and extent of periodontitis in post-menopausal (PM) women. MATERIALS AND METHODS Data from 982 PM women participating in NHANES III were analysed. Exposures were high ferritin (≥300 μg/ml) and transferrin saturation (≥45%). The primary outcome was moderate/severe periodontitis defined according to Centers for Disease Control and Prevention and the American Academy of Periodontology. The extent of periodontitis was also assessed as outcome: proportion of sites affected by clinical attachment loss ≥4 mm and probing depth ≥4 mm. Crude and adjusted prevalence ratio (PR) and mean ratio (MR) were estimated using Poisson regression. RESULTS The prevalence of moderate/severe periodontitis was 27.56%. High ferritin was associated with moderate/severe periodontitis in the crude (PR 1.55, p = .018) and in the final adjusted model (PR 1.53, p = .008). High ferritin and transferrin saturation levels were associated with a higher proportion of sites with clinical attachment loss ≥4 mm (p < .05). CONCLUSIONS The increasing serum iron markers seem to contribute to periodontitis severity and extent in PM women.
Collapse
|
40
|
Faramarzi M, Shirmohammadi A, Khorramdel A, Sadighi M, Bargahi E. Effect of non-surgical periodontal therapy on serum ferritin levels in postmenopausal women with chronic periodontitis. J Dent Res Dent Clin Dent Prospects 2021; 15:178-182. [PMID: 34712408 PMCID: PMC8538142 DOI: 10.34172/joddd.2021.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/11/2021] [Indexed: 11/13/2022] Open
Abstract
Background. Ferritin is a positive acute phase protein (APP) in inflammation and chronic infections, including chronic periodontitis. Two key factors that can regulate ferritin expression are iron and pro-inflammatory cytokines. Serum ferritin levels increase after menopause, affecting women’s health. This study aimed to evaluate serum ferritin levels in postmenopausal women upon undertaking non-surgical periodontal treatment.
Methods. In this cross-sectional study, blood samples of 38 postmenopausal women with chronic periodontitis were collected before any treatment. The serum ferritin levels and periodontal parameters, probing depth (PD), clinical attachment level (CAL), and gingival index (GI) were recorded at baseline and three months after non-surgical periodontal therapy. Wilcoxon test was used to compare serum ferritin levels before and after treatment. T-test was used for comparison of periodontal parameters, with a P value of ≤0.05 considered significant.
Results. A decrease was observed in the serum ferritin level (from 108.55 mcg/L to 98.28 mcg/L) after treatment compared to baseline (P < 0.001). Also, significant improvements in periodontal parameters were observed compared to the baseline (P < 0.001).
Conclusion. Based on the results, it can be concluded that non-surgical periodontal treatment significantly reduces serum ferritin levels in postmenopausal women with chronic periodontitis.
Collapse
Affiliation(s)
- Masoumeh Faramarzi
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Science, Tabriz, Iran
| | - Adileh Shirmohammadi
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Science, Tabriz, Iran
| | - Azin Khorramdel
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Science, Tabriz, Iran
| | - Mehrnoosh Sadighi
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Science, Tabriz, Iran
| | | |
Collapse
|
41
|
Youssef SR, Hassan EH, Morad CS, Elazab Elged AA, El-Gamal RA. Erythroferrone Expression in Anemic Rheumatoid Arthritis Patients: Is It Disordered Iron Trafficking or Disease Activity? J Inflamm Res 2021; 14:4445-4455. [PMID: 34522114 PMCID: PMC8434928 DOI: 10.2147/jir.s327465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Erythroferrone (ERFE) is well acknowledged for its inhibitory function on hepcidin synthesis in the liver during stress erythropoiesis, thereby ensuring sufficient iron supply to bone marrow erythroblasts. Hepcidin plays an indispensable role in the pathogenesis of anemia of chronic disease (ACD). Thus, ERFE was suggested to protect against ACD in various diseases. Rheumatoid arthritis (RA) is commonly involved with ACD and high hepcidin levels, with a further increase of the latter in active states. The present study is a case-control study that aimed to determine the pattern of ERFE expression in RA patients with concomitant ACD and study its relationship with hepcidin, erythropoietin (EPO) and disease activity. Patients and Methods Fifty-five RA patients with ACD were categorized into active and inactive RA using the disease activity score (DAS28); 15 healthy subjects were included as control subjects. ERFE was measured for patients and control subjects using quantitative real-time polymerase chain reaction, in addition to testing for CBC, ESR, CRP, iron profile parameters and hepcidin. EPO was assessed for patients of both active and inactive RA groups. Results ERFE and hepcidin showed the highest levels in active RA; ERFE values were similar in control subjects and inactive RA patients, while hepcidin was significantly higher in inactive RA than control subjects. Patients with high ERFE levels had higher RBC, Hct, MCV, hepcidin and EPO levels. Stepwise regression analysis has identified DAS28 and disease duration as the best predictors of ERFE values, whereas ERFE and hepcidin were independent predictors of disease activity. Conclusion We introduce ERFE as a novel marker of RA activity. Although the inhibitory effect of ERFE on hepcidin is not evident, our results still indicate that ERFE may have a beneficial erythropoietic effect in the context of ACD in RA disease activity.
Collapse
Affiliation(s)
- Soha R Youssef
- Clinical Pathology Department, Ain Shams University, Cairo, Egypt
| | | | - Caroline S Morad
- Internal Medicine and Rheumatology Department, Ain Shams University, Cairo, Egypt
| | - Adel A Elazab Elged
- Clinical Pathology Department, Ain Shams University, Cairo, Egypt.,Galala University, Suez, Egypt
| | - Rasha A El-Gamal
- Clinical Pathology Department, Ain Shams University, Cairo, Egypt
| |
Collapse
|
42
|
Lindquist DM, Dillman JR, Tkach JA. Editorial for "Hepatic Iron Quantification Using a Free-Breathing 3D Radial Gradient Echo Technique and Validation with a 2D Biopsy-Calibrated R2* Relaxometry Method". J Magn Reson Imaging 2021; 55:1417-1418. [PMID: 34523184 DOI: 10.1002/jmri.27904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Diana M Lindquist
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jonathan R Dillman
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jean A Tkach
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
43
|
Naemi FMA, Al-Adwani S, Al-Nazawi A, Al-Khatabi H. Association between HLA genotype and ferritin levels in COVID-19 infection: a study of a Saudi cohort. Infect Dis (Lond) 2021; 53:891-899. [PMID: 34304676 DOI: 10.1080/23744235.2021.1955149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND An elevation in serum ferritin levels through an unknown mechanism was observed in COVID-19 infected patients. This study examined the association between patients' HLA genotype and serum ferritin level modulation and also assessed the effect of serum ferritin levels on infection severity/mortality. MATERIALS AND METHODS One hundred and thirty-six COVID-19 Saudi patients were divided into two groups according to their ferritin levels: group 1 (<500 ng/mL, N = 67) and group 2 (>500 ng/mL, N = 69). HLA genotyping (class I and II) was carried out through the rPCR-SSO method. RESULTS High serum ferritin levels were associated with a significant increase in infection severity, as 75% of ICU patients showed high levels of ferritin compared to 43% of patients with moderate symptoms, p = .002. This elevation indicated a gender skew in that 56% of the infected male patients displayed high ferritin levels compared to 36.6% of the female patients, p = .03. In terms of mortality, 74% of patients with fatal outcomes had a high level of serum ferritin compared to 47% of recovered patients, p = .039. There was a significant difference in the HLA frequency between the two groups, with a predominance of HLA-A*01 in the low-ferritin group (19.4 vs. 6.5%, p = .002, pc = .016) and predominance of C*03 in the high ferritin group (10.9 vs. 3%, p = .047, pc = .27). CONCLUSION High serum ferritin levels are associated with an increase in COVID-19 severity, which may be affected by HLA polymorphism.
Collapse
Affiliation(s)
- Fatmah M A Naemi
- Histocompatibility and Immunogenetics Laboratory, King Fahd General Hospital, Ministry of Health, Jeddah, Saudi Arabia
| | - Shurooq Al-Adwani
- Histocompatibility and Immunogenetics Laboratory, King Fahd General Hospital, Ministry of Health, Jeddah, Saudi Arabia
| | | | - Heba Al-Khatabi
- Center of Excellence in Genomic Medicine Research, King Fahd Medical Research Center, King Abdul-Aziz University, Jeddah, Saudi Arabia
| |
Collapse
|
44
|
DePalma RG, Hayes VW, O'Leary TJ. Optimal serum ferritin level range: iron status measure and inflammatory biomarker. Metallomics 2021; 13:6287580. [PMID: 34048587 PMCID: PMC8195161 DOI: 10.1093/mtomcs/mfab030] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022]
Abstract
This report provides perspectives concerning dual roles of serum ferritin as a measure of both iron status and inflammation. We suggest benefits of a lower range of serum ferritin as has occurred for total serum cholesterol and fasting blood glucose levels. Observations during a prospective randomized study using phlebotomy in patients with peripheral arterial disease offered unique insights into dual roles of serum ferritin both as an iron status marker and acute phase reactant. Robust positive associations between serum ferritin, interleukin 6 [IL-6], tissue necrosis factor-alpha, and high sensitivity C-reactive protein were discovered. Elevated serum ferritin and IL-6 levels associated with increased mortality and with reduced mortality at ferritin levels <100 ng mL-1. Epidemiologic studies demonstrate similar outcomes. Extremely elevated ferritin and IL-6 levels also occur in individuals with high mortality due to SARS-CoV-2 infection. Disordered iron metabolism reflected by a high range of serum ferritin level signals disease severity and outcomes. Based upon experimental and epidemiologic data, we suggest testing the hypotheses that optimal ferritin levels for cardiovascular mortality reduction range from 20 to 100 ng mL-1 with % transferrin levels from 20 to 50%, to ensure adequate iron status and that ferritin levels above 194 ng mL-1 associate with all-cause mortality in population cohorts.
Collapse
Affiliation(s)
- Ralph G DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington, DC 20420, USA.,Department of Surgery, Uniformed University of the Health Sciences, Bethesda, MD 20814, USA
| | - Virginia W Hayes
- Virginia W Hayes, Ambulatory Care Service, Sierra Nevada Health Care System, Reno, NV 89502, USA
| | - Timothy J O'Leary
- Office of Research and Development, Department of Veterans Affairs, Washington, DC 20420, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
45
|
Wáng YXJ. Gender-specific liver aging and magnetic resonance imaging. Quant Imaging Med Surg 2021; 11:2893-2904. [PMID: 34249621 DOI: 10.21037/qims-21-227] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/15/2021] [Indexed: 12/14/2022]
Abstract
The number of imaging studies performed on elderly individuals will increase in the next several decades. It is important to understand normal age-related changes in the structural and functional imaging appearance of the liver. This article highlights a number of liver aging aspects which are particularly relevant to magnetic resonance imaging (MRI). Physiology of aging liver is associated with a reduction in size, in perfusion, and in function. Pulsed echo-Doppler showed substantial reduction of portal flow in elderly subjects, particularly those after the age of 75 years old. An MRI biomarker diffusion derived vessel density (DDVD) demonstrated that liver microperfusion volume in healthy females starts to decrease even before menopause age. Liver fat content and iron content increase with aging, and the change is more substantial in women after menopause. Adult men have higher liver fat and iron contents than women from the start and change less during aging. Nonalcoholic fatty liver disease (NAFLD) is very common among assumed healthy subjects. There is a male predominance of NAFLD from the paediatric population up to fifth decade of life in adults. After the age of 60 years, women overtake their male counterparts in prevalence of NAFLD. Higher liver fat leads to decreased apparent diffusion coefficient (ADC) and intravoxel incoherent motion (IVIM)-Dslow measures. Higher liver iron content shortens T2* measure, lower ADC and IVIM-Dslow measures, increases imaging noises and decreases liver visibility. Young women have high liver T1rho value and then decrease substantially, while liver T1rho in men remains relatively unchanged with aging. In positron emission tomography (PET) studies, aging is associated with an increase of both liver fluorine-18-fluorodeoxyglucose maximum standard uptake and mean standard uptake values.
Collapse
Affiliation(s)
- Yì Xiáng J Wáng
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| |
Collapse
|
46
|
Ledesma-Colunga MG, Weidner H, Vujic Spasic M, Hofbauer LC, Baschant U, Rauner M. Shaping the bone through iron and iron-related proteins. Semin Hematol 2021; 58:188-200. [PMID: 34389111 DOI: 10.1053/j.seminhematol.2021.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/18/2021] [Accepted: 06/08/2021] [Indexed: 01/04/2023]
Abstract
Well-controlled iron levels are indispensable for health. Iron deficiency is the most common cause of anemia, whereas iron overload, either hereditary or secondary due to disorders of ineffective erythropoiesis, causes widespread organ failure. Bone is particularly sensitive to fluctuations in systemic iron levels as both iron deficiency and overload are associated with low bone mineral density and fragility. Recent studies have shown that not only iron itself, but also iron-regulatory proteins that are mutated in hereditary hemochromatosis can control bone mass. This review will summarize the current knowledge on the effects of iron on bone homeostasis and bone cell activities, and on the role of proteins that regulate iron homeostasis, i.e. hemochromatosis proteins and proteins of the bone morphogenetic protein pathway, on bone remodeling. As disorders of iron homeostasis are closely linked to bone fragility, deeper insights into common regulatory mechanisms may provide new opportunities to concurrently treat disorders affecting iron homeostasis and bone.
Collapse
Affiliation(s)
- Maria G Ledesma-Colunga
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Heike Weidner
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Maja Vujic Spasic
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Lorenz C Hofbauer
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
47
|
Rago V, Leo I, Marrocco A, Izzo R, Filetti C. Blood and performance adaptations to individual training load in professional soccer: a team study. J Sports Med Phys Fitness 2021; 62:1237-1245. [PMID: 34137574 DOI: 10.23736/s0022-4707.21.12690-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
PURPOSE The aim of this study was to describe seasonal changes in iron storage, hormonal status and functional capacity in relation to accumulated training load in a professional male soccer team. METHODS Resting blood samples, countermovement jump (CMJ) and aerobic capacity (45-15 test) were collected over a 6-month period from the start of the preparatory period to the middle-season (E1 to E4) in a professional male soccer team (n=15 outfield players). External training load was regularly quantified using a wearable 10-Hz global positioning system. RESULTS One player systematically showed reduced iron storage throughout the season (ferritin<110 μg·l-1, hemoglobin<14 g·dl-1). No significant differences in blood and performance parameters were observed throughout the season (P>0.05). However, accumulated total distance and high-intensity distance (above maximal aerobic speed) from E1 to E3 were negatively correlated to changes in haematocrit, hemoglobin and red blood cells (r=-0.85 to -0.67; P<0.05) and positively to changes in ferritin (r=0.63-0.69; P<0.05). Additionally, high-intensity distance covered between E1 and E3 was negatively correlated to changes in testosterone concentrations (r [95%CI]=-0.71 [-0.93; -0.15]; P=0.021). CONCLUSIONS Resting blood parameters and functional capacity of male soccer players appeared to be stable throughout the early competitive period. However, iron storage and hormonal status are likely to be affected by accumulated high-intensity activity performed during practice and competition. Practitioners involved with GPS-based TL monitoring could consider the accumulated amount of high-intensity activity to inform medical staffs about possible changes in oxygen-carrying capacity and anaerobic overtraining.
Collapse
Affiliation(s)
- Vincenzo Rago
- Faculty of Health Sciences and Sports, Universidade Europeia, Lisbon, Portugal -
| | - Italo Leo
- Department of Human, Philosophical and Educational Sciences, University of Salerno, Fisciano, Salerno, Italy
| | - Arcano Marrocco
- Department of Biomolecular Sciences, University Carlo Bo, Urbino, Italy
| | - Riccardo Izzo
- Department of Biomolecular Sciences, University Carlo Bo, Urbino, Italy
| | - Cristoforo Filetti
- Performance Department, Paris Saint-Germain Football Club, Paris, France.,Italian School of Sport Sciences and Exercise, Faculty of Medicine and Surgery, University of Tor Vergata, Rome, Italy
| |
Collapse
|
48
|
Weyand AC, McGann PT. Eliminating race-based reference ranges in haematology: a call to action. LANCET HAEMATOLOGY 2021; 8:e462-e466. [PMID: 34048684 DOI: 10.1016/s2352-3026(21)00106-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022]
Abstract
In haematology, as in all of medicine, the use of reference intervals for laboratory variables is essential to define disease states and inform treatment decisions. There are many haematological variables, including haemoglobin, mean corpuscular volume, absolute neutrophil count, and iron indices, that are often reported to be different on the basis of a person's race or ethnicity. Although there are many haematological conditions with a genetic basis, such that it is appropriate to consider ancestry in the diagnostic algorithm, defining pathology on the basis of a social construct such as race is unacceptable. The inclusion of separate thresholds or simple statements that so-called normal values vary by race further validates the common misperception that there are physiological differences between Black and white patients. These statements might have downstream effects on diagnostic and treatment decisions that exacerbate existing racial health disparities. In this Viewpoint, we argued for the removal of race-based reference intervals across haematology.
Collapse
Affiliation(s)
- Angela C Weyand
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Patrick T McGann
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
49
|
Kang W, Barad A, Clark AG, Wang Y, Lin X, Gu Z, O'Brien KO. Ethnic Differences in Iron Status. Adv Nutr 2021; 12:1838-1853. [PMID: 34009254 PMCID: PMC8483971 DOI: 10.1093/advances/nmab035] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Iron is unique among all minerals in that humans have no regulatable excretory pathway to eliminate excess iron after it is absorbed. Iron deficiency anemia occurs when absorbed iron is not sufficient to meet body iron demands, whereas iron overload and subsequent deposition of iron in key organs occur when absorbed iron exceeds body iron demands. Over time, iron accumulation in the body can increase risk of chronic diseases, including cirrhosis, diabetes, and heart failure. To date, only ∼30% of the interindividual variability in iron absorption can be captured by iron status biomarkers or iron regulatory hormones. Much of the regulation of iron absorption may be under genetic control, but these pathways have yet to be fully elucidated. Genome-wide and candidate gene association studies have identified several genetic variants that are associated with variations in iron status, but the majority of these data were generated in European populations. The purpose of this review is to summarize genetic variants that have been associated with alterations in iron status and to highlight the influence of ethnicity on the risk of iron deficiency or overload. Using extant data in the literature, linear mixed-effects models were constructed to explore ethnic differences in iron status biomarkers. This approach found that East Asians had significantly higher concentrations of iron status indicators (serum ferritin, transferrin saturation, and hemoglobin) than Europeans, African Americans, or South Asians. African Americans exhibited significantly lower hemoglobin concentrations compared with other ethnic groups. Further studies of the genetic basis for ethnic differences in iron metabolism and on how it affects disease susceptibility among different ethnic groups are needed to inform population-specific recommendations and personalized nutrition interventions for iron-related disorders.
Collapse
Affiliation(s)
- Wanhui Kang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Alexa Barad
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Andrew G Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA,Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Yiqin Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
50
|
Iron Deficiency - Not Only a Premenopausal Topic After Bariatric Surgery? Obes Surg 2021; 31:3242-3250. [PMID: 33821393 PMCID: PMC8175328 DOI: 10.1007/s11695-021-05380-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 11/11/2022]
Abstract
Purpose In our centre, specialized high dose multivitamin supplementation designed to meet the needs of patients after gastric bypass surgery is routinely recommended in the early postoperative period. The aim of the present study was to analyse whether iron supplementation prescribed in clinical practice is sufficient in both sexes and whether multivitamin supplementation standardized for women might potentially lead to iron overload in men. Materials/Methods This was a retrospective study covering the period up to 36 months after bariatric surgery. Three groups were compared (men, premenopausal and postmenopausal women). The iron status was evaluated employing serum ferritin concentrations. Results A total of 283 patients who had at least one follow-up visit between January 2015 and April 2018 at a specialized academic outpatient centre were included (71 men, 130 premenopausal women, 82 postmenopausal women). Thirty-six months after surgery, 33.3%, 68.4% and 54.5% of the men, pre- and postmenopausal women, respectively, were iron deficient. The preoperative prevalence of excess ferritin levels was 13.7% in premenopausal, 3.0% in postmenopausal women, 5.7% in men and declined in the following months. Conclusion Iron deficiency is very common after gastric bypass surgery, and even high dosages of multivitamin and mineral supplements might not be sufficient to prevent the development of iron deficiency. Men, pre- and postmenopausal women differ in their prevalence of iron deficiency which demands adapted iron dosage regimens based on the sex and the age. Iron overload is rare in all observed groups and highest in premenopausal women. Graphical abstract ![]()
Collapse
|