1
|
Muggeo P, Grassi M, D’Ascanio V, Forte J, Brescia V, Di Serio F, Piacente L, Giordano P, Santoro N, Faienza MF. Bone Remodeling in Children with Acute Lymphoblastic Leukemia: A Two-Year Prospective Longitudinal Study. Int J Mol Sci 2025; 26:4307. [PMID: 40362542 PMCID: PMC12072470 DOI: 10.3390/ijms26094307] [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] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Childhood leukemia survivors are at risk of long-term complications. Data on bone remodeling in childhood acute lymphoblastic leukemia (ALL) are limited. This 2-year prospective longitudinal study investigated bone remodeling and bone turnover markers at diagnosis, during treatment, and until stopping treatment, in ALL patients < 18 years, to clarify the influence of leukemia itself and/or chemotherapy on bone. METHODS A total of 22 ALL children (12 males, age 5.5 ± 3.6 years) underwent blood sampling at the 5 time point (T0-T4). Osteoprotegerin (OPG), receptor-activator-NF-B-ligand (RANKL), osteocalcin (OC), C-terminal-telopeptide-type-I-collagen (CTX), bone-alkaline-phosphatase (bALP), tartrate-resistant acid-phosphatase-5b (TRACP5b), procollagen-type-I-N-terminal-propeptide (P1NP), Dickkopf-1 (DKK-1), and sclerostin were assessed. Data from patients at T0 were compared to a control group of healthy children. We used the principal component analysis (PCA) for statistics. RESULTS Levels of CTX, OC, P1NP, and bALP resulted lower in ALL children than controls (p = 0.009 for CTX and p < 0.001 for the others), also DKK1 and sclerostin (p < 0.0001 and p = 0.023). RANKL ed OPG were higher in patients. During T0-T4, CTX, OC, P1NP, TRACP5b, and bALP showed a significant increase, in particular at T0-T1 (end-of-induction). Less evident changes were detected onwards. CONCLUSIONS The onset of leukemia has been revealed as a key point in determining a slowing of bone remodeling in ALL children.
Collapse
Affiliation(s)
- Paola Muggeo
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinico, 70124 Bari, Italy; (M.G.); (N.S.)
| | - Massimo Grassi
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinico, 70124 Bari, Italy; (M.G.); (N.S.)
| | - Vito D’Ascanio
- Institute of Sciences of Food Production (ISPA), Italian National Research Council (CNR), 70126 Bari, Italy;
| | - Jessica Forte
- Pediatric Department, Ospedale Della Murgia “F. Perinei”, 70022 Altamura, Italy;
| | - Vincenzo Brescia
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy; (V.B.); (F.D.S.)
| | - Francesca Di Serio
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy; (V.B.); (F.D.S.)
| | - Laura Piacente
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy; (L.P.); (M.F.F.)
| | - Paola Giordano
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Nicola Santoro
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinico, 70124 Bari, Italy; (M.G.); (N.S.)
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy; (L.P.); (M.F.F.)
| |
Collapse
|
2
|
Sukkho T, Khanongnuch C, Lumyong S, Ruangsuriya J, Apichai S, Surh YJ, Pattananandecha T, Saenjum C. Osteoprotective Activity of Sambucus javanica Reinw Ex Blume subsp. javanica Leaf Extracts by Suppressing ROS Production. Antioxidants (Basel) 2025; 14:252. [PMID: 40227226 PMCID: PMC11939775 DOI: 10.3390/antiox14030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Sambucus javanica subsp. javanica (SJ) has been used in traditional medicine in the northern region of Thailand for healing bone fractures; however, studies on how this plant stimulates bone formation are still scarce. The present study aimed to investigate the potential of crude extracts and fractions obtained from SJ leaves for osteoporotic protection. All samples were investigated in murine preosteoblast MC3T3-E1 cells for bone formation and resorption biomarkers, namely alkaline phosphatase (ALP), osteocalcin (OC), osteoprotegerin (OPG), receptor activator of nuclear factor-κB ligand (RANKL), and the OPG/RANKL ratio. Additionally, calcium deposits were determined using the alizarin red S staining technique. The results indicated that the crude water and the crude ethanol extracts contained gallic acid, rutin, and chlorogenic acid as major compounds. The extracts stimulated osteoblastic cell differentiation and enhanced osteoprotective activity, as measured by a significant increase in ALP activity, OC, OPG, the OPG/RANKL ratio, and the degree of calcification. Additionally, they exhibited a negative impact on bone resorption by significantly reducing RANKL and reactive oxygen species (ROS) production. Therefore, our findings add novel evidence indicating that the SJ crude extracts from water and ethanol extraction could be further utilized as a natural active pharmaceutical ingredient (NAPI) for the development of bone health products.
Collapse
Affiliation(s)
- Treethip Sukkho
- Department of Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
| | - Chartchai Khanongnuch
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Research Center for Multidisciplinary Approaches to Miang, Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saisamorn Lumyong
- Department of Biology, Faculty of Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Center of Microbial Diversity and Sustainable Utilization, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jetsada Ruangsuriya
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sutasinee Apichai
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 08828, Republic of Korea;
| | - Thanawat Pattananandecha
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chalermpong Saenjum
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Research Center for Multidisciplinary Approaches to Miang, Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
3
|
Farella I, Chiarito M, Vitale R, D’Amato G, Faienza MF. The "Burden" of Childhood Obesity on Bone Health: A Look at Prevention and Treatment. Nutrients 2025; 17:491. [PMID: 39940349 PMCID: PMC11821239 DOI: 10.3390/nu17030491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Childhood obesity represents a multifaceted challenge to bone health, influenced by a combination of endocrine, metabolic, and mechanical factors. Excess body fat correlates with an increase in bone mineral density (BMD) yet paradoxically elevates fracture risk due to compromised bone quality and increased mechanical loading on atypical sites. Additionally, subjects with syndromic obesity, as well as individuals with atypical nutritional patterns, including those with eating disorders, show bone fragility through unique genetic and hormonal dysregulations. Emerging evidence underscores the adverse effects of new pharmacological treatments for severe obesity on bone health. Novel drugs, such as glucagon-like peptide-1 (GLP-1) receptor agonists, and bariatric surgery demonstrate potential in achieving weight loss, though limited evidence is available regarding their short- and long-term impacts on skeletal health. This review provides a comprehensive analysis of the mechanisms underlying the impact of childhood obesity on bone health. It critically appraises evidence from in vitro studies, animal models, and clinical research in children with exogenous obesity, syndromic obesity, and eating disorders. It also explores the effects of emerging pharmacological and surgical treatments for severe obesity on skeletal integrity, highlights prevention strategies, and identifies research gaps.
Collapse
Affiliation(s)
- Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy;
| | - Mariangela Chiarito
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Rossella Vitale
- Giovanni XXIII Pediatric Hospital, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Gabriele D’Amato
- Neonatal Intensive Care Unit, Di Venere Hospital, 70012 Bari, Italy;
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| |
Collapse
|
4
|
Wang Y, Hu J, Sun L, Zhou B, Lin X, Zhang Q, Wang O, Jiang Y, Xia W, Xing X, Li M. Correlation of serum DKK1 level with skeletal phenotype in children with osteogenesis imperfecta. J Endocrinol Invest 2024; 47:2785-2795. [PMID: 38744806 PMCID: PMC11473575 DOI: 10.1007/s40618-024-02380-9] [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: 01/23/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE We aim to detect serum DKK1 level of pediatric patients with OI and to analyze its relationship with the genotype and phenotype of OI patients. METHODS A cohort of pediatric OI patients and age-matched healthy children were enrolled. Serum levels of DKK1 and bone turnover biomarkers were measured by enzyme-linked immunosorbent assay. Bone mineral density (BMD) was measured by Dual-energy X-ray absorptiometry. Pathogenic mutations of OI were detected by next-generation sequencing and confirmed by Sanger sequencing. RESULTS A total of 62 OI children with mean age of 9.50 (4.86, 12.00) years and 29 healthy children were included in this study. The serum DKK1 concentration in OI children was significantly higher than that in healthy children [5.20 (4.54, 6.32) and 4.08 (3.59, 4.92) ng/mL, P < 0.001]. The serum DKK1 concentration in OI children was negatively correlated with height (r = - 0.282), height Z score (r = - 0.292), ALP concentration (r = - 0.304), lumbar BMD (r = - 0.276), BMD Z score of the lumbar spine and femoral neck (r = - 0.32; r = - 0.27) (all P < 0.05). No significant difference in serum DKK1 concentration was found between OI patients with and without vertebral compression fractures. In patients with spinal deformity (22/62), serum DKK1 concentration was positively correlated with SDI (r = 0.480, P < 0.05). No significant correlation was observed between serum DKK1 concentration and the annual incidence of peripheral fractures, genotype and types of collagen changes in OI children. CONCLUSION The serum DKK1 level was not only significantly elevated in OI children, but also closely correlated to their skeletal phenotype, suggesting that DKK1 may become a new biomarker and a potential therapeutic target of OI.
Collapse
Affiliation(s)
- Y Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - J Hu
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - L Sun
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - B Zhou
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - X Lin
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - Q Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - O Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - Y Jiang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - W Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - X Xing
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China
| | - M Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Beijing, 100730, Dongcheng District, China.
| |
Collapse
|
5
|
Gajewska J, Chełchowska M, Szamotulska K, Klemarczyk W, Strucińska M, Ambroszkiewicz J. Differences in Bone Metabolism between Children with Prader-Willi Syndrome during Growth Hormone Treatment and Healthy Subjects: A Pilot Study. Int J Mol Sci 2024; 25:9159. [PMID: 39273107 PMCID: PMC11394978 DOI: 10.3390/ijms25179159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Despite therapy with growth hormone (GH) in children with Prader-Willi syndrome (PWS), low bone mineral density and various orthopedic deformities have been observed often. Therefore, this study aimed to analyze bone markers, with an emphasis on vitamin K-dependent proteins (VKDPs), in normal-weight children with PWS undergoing GH therapy and a low-energy dietary intervention. Twenty-four children with PWS and 30 healthy children of the same age were included. Serum concentrations of bone alkaline phosphatase (BALP), osteocalcin (OC), carboxylated-OC (Gla-OC), undercarboxylated-OC (Glu-OC), periostin, osteopontin, osteoprotegerin (OPG), sclerostin, C-terminal telopeptide of type I collagen (CTX-I), and insulin-like growth factor-I (IGF-I) were determined using immunoenzymatic methods. OC levels and the OC/CTX-I ratios were lower in children with PWS than in healthy children (p = 0.011, p = 0.006, respectively). Glu-OC concentrations were lower (p = 0.002), but Gla-OC and periostin concentrations were higher in patients with PWS compared with the controls (p = 0.005, p < 0.001, respectively). The relationships between IGF-I and OC (p = 0.013), Gla-OC (p = 0.042), and the OC/CTX-I ratio (p = 0.017) were significant after adjusting for age in children with PWS. Bone turnover disorders in children with PWS may result from impaired bone formation due to the lower concentrations of OC and the OC/CTX-I ratio. The altered profile of OC forms with elevated periostin concentrations may indicate more intensive carboxylation processes of VKDPs in these patients. The detailed relationships between the GH/IGF-I axis and bone metabolism markers, particularly VKDPs, in children with PWS requires further research.
Collapse
Affiliation(s)
- Joanna Gajewska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland
| | - Magdalena Chełchowska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland
| | - Witold Klemarczyk
- Department of Nutrition, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland
| | - Małgorzata Strucińska
- Department of Nutrition, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland
| | - Jadwiga Ambroszkiewicz
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland
| |
Collapse
|
6
|
Atilano-Miguel S, Barbosa-Cortés L, Ortiz-Muñiz R, Maldonado-Hernández J, Martin-Trejo JA, Rodríguez-Cruz M, Balcázar-Hernández L, Solís-Labastida KA, Bautista-Martínez BA, Juárez-Moya A, Hernández-Piñón Z, Galindo-Rodríguez RA, Chávez-Anaya A, Valdez-Avilez RE, Domínguez-Salgado JM, Villa-Morales J, Rodríguez-Palacios ME. Changes in RANKL, OPG, and 25(OH)D Levels in Children with Leukemia from Diagnosis to Remission. Cancers (Basel) 2024; 16:2811. [PMID: 39199584 PMCID: PMC11352827 DOI: 10.3390/cancers16162811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND The receptor activator of the nuclear factor-kB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) pathway is a determining pathway in the balance between bone formation and resorption, and disruptions in this complex can affect bone metabolism. METHODS This study analyzes the changes in RANKL, OPG, and 25(OH)D levels; the RANKL/OPG ratio; and other bone turnover markers (BTMs) from diagnosis to complete remission in children with acute lymphoblastic leukemia (ALL). This is a prospective observational cohort study, carried out at the Instituto Mexicano del Seguro Social, Mexico City, including 33 patients (4-17 years) with newly diagnosed B-cell ALL. The patients were treated with the HP09 chemotherapy protocol. Children who had previously been treated with corticosteroids were excluded. A peripheral blood sample at diagnosis and remission was collected to determine the 25(OH)D and BTM concentrations. RESULTS Increased RANKL (p = 0.001) and osteocalcin (p < 0.001) levels and RANKL/OPG ratio (<0.001) and a decreased OPG level (p = 0.005) were observed at remission, predominantly in the high-risk (HR) relapse and vitamin D deficiency groups. A negative association between RANKL and OPG (r = -0.454, p = 0.008) was observed. CONCLUSIONS we suggest that the RANKL/OPG ratio could serve as a bone remodeling marker in ALL patients.
Collapse
Affiliation(s)
- Salvador Atilano-Miguel
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 14387, Mexico; (S.A.-M.); (R.O.-M.)
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de México 9340, Mexico;
| | - Lourdes Barbosa-Cortés
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - Rocío Ortiz-Muñiz
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 14387, Mexico; (S.A.-M.); (R.O.-M.)
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de México 9340, Mexico;
| | - Jorge Maldonado-Hernández
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - Jorge A. Martin-Trejo
- Departamento Clínico de Hematología, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.A.M.-T.); (K.A.S.-L.); (B.A.B.-M.); (A.J.-M.); (Z.H.-P.)
| | - Maricela Rodríguez-Cruz
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - Lourdes Balcázar-Hernández
- Departamento de Endocrinología, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico;
| | - Karina A. Solís-Labastida
- Departamento Clínico de Hematología, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.A.M.-T.); (K.A.S.-L.); (B.A.B.-M.); (A.J.-M.); (Z.H.-P.)
| | - Benito A. Bautista-Martínez
- Departamento Clínico de Hematología, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.A.M.-T.); (K.A.S.-L.); (B.A.B.-M.); (A.J.-M.); (Z.H.-P.)
| | - Azalia Juárez-Moya
- Departamento Clínico de Hematología, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.A.M.-T.); (K.A.S.-L.); (B.A.B.-M.); (A.J.-M.); (Z.H.-P.)
| | - Zayra Hernández-Piñón
- Departamento Clínico de Hematología, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.A.M.-T.); (K.A.S.-L.); (B.A.B.-M.); (A.J.-M.); (Z.H.-P.)
| | - Raeline A. Galindo-Rodríguez
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - Adriana Chávez-Anaya
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - Rosa E. Valdez-Avilez
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de México 9340, Mexico;
| | - Juan M. Domínguez-Salgado
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - Judith Villa-Morales
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| | - María E. Rodríguez-Palacios
- Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad (UMAE), Instituto Mexicano del Seguro Social (IMSS), Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (J.M.-H.); (M.R.-C.); (R.A.G.-R.); (A.C.-A.); (J.M.D.-S.); (J.V.-M.); (M.E.R.-P.)
| |
Collapse
|
7
|
Faienza MF, Giardinelli S, Annicchiarico A, Chiarito M, Barile B, Corbo F, Brunetti G. Nutraceuticals and Functional Foods: A Comprehensive Review of Their Role in Bone Health. Int J Mol Sci 2024; 25:5873. [PMID: 38892062 PMCID: PMC11172758 DOI: 10.3390/ijms25115873] [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: 04/03/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Bone health is the result of a tightly regulated balance between bone modeling and bone remodeling, and alterations of these processes have been observed in several diseases both in adult and pediatric populations. The imbalance in bone remodeling can ultimately lead to osteoporosis, which is most often associated with aging, but contributing factors can already act during the developmental age, when over a third of bone mass is accumulated. The maintenance of an adequate bone mass is influenced by genetic and environmental factors, such as physical activity and diet, and particularly by an adequate intake of calcium and vitamin D. In addition, it has been claimed that the integration of specific nutraceuticals such as resveratrol, anthocyanins, isoflavones, lycopene, curcumin, lutein, and β-carotene and the intake of bioactive compounds from the diet such as honey, tea, dried plums, blueberry, and olive oil can be efficient strategies for bone loss prevention. Nutraceuticals and functional foods are largely used to provide medical or health benefits, but there is an urge to determine which products have adequate clinical evidence and a strong safety profile. The aim of this review is to explore the scientific and clinical evidence of the positive role of nutraceuticals and functional food in bone health, focusing both on molecular mechanisms and on real-world studies.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, 70124 Bari, Italy; (M.F.F.)
| | - Silvia Giardinelli
- Department of Medical Sciences, Pediatrics, University of Ferrara, 44121 Ferrara, Italy
| | - Alessia Annicchiarico
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (B.B.)
| | - Mariangela Chiarito
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, 70124 Bari, Italy; (M.F.F.)
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (B.B.)
| | - Filomena Corbo
- Department of Pharmacy-Drug Sciences, University of Bari “A. Moro”, 70125 Bari, Italy;
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (B.B.)
| |
Collapse
|
8
|
Madeo SF, Zagaroli L, Vandelli S, Calcaterra V, Crinò A, De Sanctis L, Faienza MF, Fintini D, Guazzarotti L, Licenziati MR, Mozzillo E, Pajno R, Scarano E, Street ME, Wasniewska M, Bocchini S, Bucolo C, Buganza R, Chiarito M, Corica D, Di Candia F, Francavilla R, Fratangeli N, Improda N, Morabito LA, Mozzato C, Rossi V, Schiavariello C, Farello G, Iughetti L, Salpietro V, Salvatoni A, Giordano M, Grugni G, Delvecchio M. Endocrine features of Prader-Willi syndrome: a narrative review focusing on genotype-phenotype correlation. Front Endocrinol (Lausanne) 2024; 15:1382583. [PMID: 38737552 PMCID: PMC11082343 DOI: 10.3389/fendo.2024.1382583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Prader-Willi syndrome (PWS) is a complex genetic disorder caused by three different types of molecular genetic abnormalities. The most common defect is a deletion on the paternal 15q11-q13 chromosome, which is seen in about 60% of individuals. The next most common abnormality is maternal disomy 15, found in around 35% of cases, and a defect in the imprinting center that controls the activity of certain genes on chromosome 15, seen in 1-3% of cases. Individuals with PWS typically experience issues with the hypothalamic-pituitary axis, leading to excessive hunger (hyperphagia), severe obesity, various endocrine disorders, and intellectual disability. Differences in physical and behavioral characteristics between patients with PWS due to deletion versus those with maternal disomy are discussed in literature. Patients with maternal disomy tend to have more frequent neurodevelopmental problems, such as autistic traits and behavioral issues, and generally have higher IQ levels compared to those with deletion of the critical PWS region. This has led us to review the pertinent literature to investigate the possibility of establishing connections between the genetic abnormalities and the endocrine disorders experienced by PWS patients, in order to develop more targeted diagnostic and treatment protocols. In this review, we will review the current state of clinical studies focusing on endocrine disorders in individuals with PWS patients, with a specific focus on the various genetic causes. We will look at topics such as neonatal anthropometry, thyroid issues, adrenal problems, hypogonadism, bone metabolism abnormalities, metabolic syndrome resulting from severe obesity caused by hyperphagia, deficiencies in the GH/IGF-1 axis, and the corresponding responses to treatment.
Collapse
Affiliation(s)
- Simona F. Madeo
- Department of Medical and Surgical Sciences for Mother, Children and Adults, Pediatric Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Zagaroli
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| | - Sara Vandelli
- Department of Medical and Surgical Sciences for Mother, Children and Adults, Post-Graduate School of Pediatrics, University of Modena and Reggio Emilia, Modena, Italy
| | - Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, Milano, Italy
| | - Antonino Crinò
- Center for Rare Diseases and Congenital Defects, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Luisa De Sanctis
- Pediatric Endocrinology, Regina Margherita Children Hospital – Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
| | - Danilo Fintini
- Prader Willi Reference Center, Endocrinology and Diabetology Unit, Pediatric University Department, IRCCS Bambino Gesù Children Hospital, Rome, Italy
| | - Laura Guazzarotti
- Pediatric Endocrinology Unit, University Hospital of Padova, Padova, Italy
| | - Maria Rosaria Licenziati
- Neuro-endocrine Diseases and Obesity Unit, Department of Neurosciences, Santobono-Pausilipon Children’s Hospital, Naples, Italy
| | - Enza Mozzillo
- Department of Translational and Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Roberta Pajno
- Pediatric Unit, IRCCS San Raffaele Institute, Milan, Italy
| | - Emanuela Scarano
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria E. Street
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - Malgorzata Wasniewska
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
- Pediatric Unit, Gaetano Martino University Hospital of Messina, Messina, Italy
| | - Sarah Bocchini
- Prader Willi Reference Center, Endocrinology and Diabetology Unit, Pediatric University Department, IRCCS Bambino Gesù Children Hospital, Rome, Italy
| | - Carmen Bucolo
- Pediatric Unit, IRCCS San Raffaele Institute, Milan, Italy
| | - Raffaele Buganza
- Pediatric Endocrinology, Regina Margherita Children Hospital – Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Mariangela Chiarito
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
| | - Domenico Corica
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
- Pediatric Unit, Gaetano Martino University Hospital of Messina, Messina, Italy
| | - Francesca Di Candia
- Department of Translational and Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | | | - Nadia Fratangeli
- Division of Auxology, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Verbania, Italy
| | - Nicola Improda
- Neuro-endocrine Diseases and Obesity Unit, Department of Neurosciences, Santobono-Pausilipon Children’s Hospital, Naples, Italy
| | | | - Chiara Mozzato
- Child and Women Health Department, University of Padova, Padova, Italy
| | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, Milano, Italy
| | | | - Giovanni Farello
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Lorenzo Iughetti
- Department of Medical and Surgical Sciences for Mother, Children and Adults, Pediatric Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Vincenzo Salpietro
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Mara Giordano
- Laboratory of Genetics, Struttura Complessa a Direzione Universitaria (SCDU) Biochimica Clinica, Ospedale Maggiore della Carità, Novara, Italy
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Graziano Grugni
- Division of Auxology, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Verbania, Italy
| | - Maurizio Delvecchio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
9
|
杜 信, 谢 静, 邹 玲. [Advances in Molecular Regulatory Mechanisms of Jaw Repair and Reconstruction]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:224-229. [PMID: 38322535 PMCID: PMC10839496 DOI: 10.12182/20240160101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Indexed: 02/08/2024]
Abstract
Jawbone injuries resulting from trauma, diseases, and surgical resections are commonly seen in clinical practice, necessitating precise and effective strategies for repair and reconstruction to restore both function and aesthetics. The precise and effective repair and the reconstruction of jawbone injuries pose a significant challenge in the field of oral and maxillofacial surgery, owing to the unique biomechanical characteristics and physiological functions of the jawbone. The natural repair process following jawbone injuries involves stages such as hematoma formation, inflammatory response, ossification, and bone remodeling. Bone morphogenetic proteins (BMPs), transforming growth factor beta (TGF-β), vascular endothelial growth factor (VEGF), and other growth factors play crucial roles in promoting jawbone regeneration. Cytokines such as interleukins and tumor necrosis factor play dual roles in regulating inflammatory response and bone repair. In recent years, significant progress in molecular biology research has been made in the field of jawbone repair and reconstruction. Tissue engineering technologies, including stem cell therapy, bioactive scaffolds, and growth factor delivery systems, have found important applications in jawbone repair. However, the intricate molecular regulatory mechanisms involved in the complex jawbone repair and reconstruction methods are not fully understood and still require further research. Future research directions will be focused on the precise control of these molecular processes and the development of more efficient combination therapeutic strategies to promote the effective and functional reconstruction of the jawbone. This review aims to examine the latest findings on the molecular regulatory mechanisms of the repair and reconstruction of jawbone injuries and the therapeutic strategies. The conclusions drawn in this article provide a molecular-level understanding of the repair of jawbone injuries and highlight potential directions for future research.
Collapse
Affiliation(s)
- 信眉 杜
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓科 (成都 610041)State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 静 谢
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓科 (成都 610041)State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 玲 邹
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓科 (成都 610041)State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Bindels-de Heus KGCB, Hagenaar DA, Mous SE, Dekker I, van der Kaay DCM, Kerkhof GF, Elgersma Y, Moll HA, de Wit MCY. Bone health in children with Angelman syndrome at the ENCORE Expertise Center. Eur J Pediatr 2024; 183:103-111. [PMID: 37831301 PMCID: PMC10857954 DOI: 10.1007/s00431-023-05231-6] [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: 07/17/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Angelman syndrome (AS) is a rare genetic disorder due to lack of UBE3A function on chromosome 15q11.2q13 caused by a deletion, uniparental paternal disomy (UPD), imprinting center disorder (ICD), or pathological variant of the UBE3A gene. AS is characterized by developmental delay, epilepsy, and lack of speech. Although fractures are observed frequently in our clinical practice, there are few studies on bone health in AS. The aim of this study is to investigate bone health in children with AS. In this prospective cohort study, we describe bone health in 91 children with AS visiting the ENCORE Expertise Center for AS between April 2010 and December 2021. Bone health was assessed with the bone health index (BHI) in standard deviation score (SDS) measured by digital radiogrammetry of the left hand using BoneXpert software. Risk factors analyzed were age, sex, genetic subtype, epilepsy, anti-seizure medication use, mobility, body mass index (BMI), and onset of puberty. Children with AS had a mean BHI of -1.77 SDS (SD 1.4). A significantly lower BHI was found in children with a deletion (-2.24 SDS) versus non-deletion (-1.02 SDS). Other factors associated with reduced BHI-SDS were inability to walk and late onset of puberty. Children with a history of one or more fractures (22%) had a significantly lower BHI than children without fractures (-2.60 vs -1.56 SDS). Longitudinal analysis showed a significant decrease in BHI-SDS with age in all genetic subtypes. Conclusions: Children with AS have a reduced bone health. Risk factors are deletion genotype, no independent walking, and late onset of puberty. Bone health decreased significantly with age. What is Known: • Children with neurological disorders often have a low bone health and higher risk of fractures. • Little is known about bone health in children with Angelman syndrome (AS). What is New: • Children with AS showed a reduced bone health and this was significantly associated with having a deletion, not being able to walk independently, and late onset of puberty. • Longitudinal analysis showed a significant decrease in bone health as children got older.
Collapse
Affiliation(s)
- Karen G C B Bindels-de Heus
- Dept. of Pediatrics, Erasmus MC Sophia Children's Hospital, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.
| | - Doesjka A Hagenaar
- Dept. of Pediatrics, Erasmus MC Sophia Children's Hospital, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
- Dept. of Child- and Adolescent Psychiatry and Psychology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Sabine E Mous
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
- Dept. of Child- and Adolescent Psychiatry and Psychology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ilonka Dekker
- Dept. of Pediatrics, Erasmus MC Sophia Children's Hospital, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | | | - Gerthe F Kerkhof
- Dept. of Pediatric Endocrinology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ype Elgersma
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
- Dept. of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Henriette A Moll
- Dept. of Pediatrics, Erasmus MC Sophia Children's Hospital, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
| | - Marie-Claire Y de Wit
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
- Dept. of Neurology and Pediatric Neurology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
11
|
Faienza MF, Urbano F, Chiarito M, Lassandro G, Giordano P. Musculoskeletal health in children and adolescents. Front Pediatr 2023; 11:1226524. [PMID: 38161439 PMCID: PMC10754974 DOI: 10.3389/fped.2023.1226524] [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: 05/21/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024] Open
Abstract
The purpose of this narrative review was to investigate the key determinants of musculoskeletal health in childhood and adolescence, with particular attention to the role of physical activity. First, we examined the importance of bone modeling and remodeling in maintaining the bone health and the integrity and mechanical characteristic of the skeleton. In addition, we reported the evidence on an appropriate calcium and vitamin D intake, as well as local load variation in achieving proper peak bone mass. Proteomic and transcriptomic studies identified the skeletal muscle "secretoma", consisting of several myokines involved in endocrine and paracrine functions. Among these, we explored the role of irisin, a myokine involved in the muscle-bone crosstalk, and in the regulation of metabolic pathways. It is known that physical activity during growing positively impacts on skeleton and can protect by bone loss in adulthood. However, there are still concerns about the optimal interval duration and exercise intensity, particularly at the pubertal growth spurt which represents a window of opportunity to increase skeletal strength. We reported data from clinical trials performed in the last 5 years analyzing the impact of the type and timing of physical activity during childhood on skeletal development. Finally, we reported recent data on the significance of physical activity in some rare diseases.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Mariangela Chiarito
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Paola Giordano
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
12
|
Amr M, Mohie-Eldinn M, Farid A. Evaluation of buffalo, cow, goat and camel milk consumption on multiple health outcomes in male and female Sprague Dawley rats. Int Dairy J 2023; 146:105760. [DOI: 10.1016/j.idairyj.2023.105760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
|
13
|
Xing W, Liang L, Dong N, Chen L, Liu Z. Abnormal changes of bone metabolism markers with age in children with cerebral palsy. Front Pediatr 2023; 11:1214608. [PMID: 37593441 PMCID: PMC10427878 DOI: 10.3389/fped.2023.1214608] [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: 04/30/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Cerebral palsy (CP) is a broad range of diseases with permanent and nonprogressive motor impairments, carrying a high cost for both the individual and the society. The characteristics of low bone mineral density and high risk of fractures suggest that bone metabolism disorders are present in CP. This study aims to investigate the association between indicators of bone metabolism and children with CP. A total of 139 children (75 children with CP and 64 healthy controls) were included in this cross-sectional study. Participants were divided into three age groups (0-2 years, 2.1-4 years, and 4.1-7 years). All children with CP were diagnosed according to clinical criteria and furtherly divided into clinical subtypes. The levels of total procollagen type I N-terminal propeptide (TPINP), N-MID osteocalcin (OC), beta-crosslaps (β-CTX), 25-hydroxyvitamin D (25-OHD) and parathyroid hormone (PTH) in the serum were measured with corresponding detection kits according to the manufacturer's instructions. Serum levels of TPINP and 25-OHD were lower with older age, whereas β-CTX and PTH were higher with older age. In the CP group, TPINP (age 0-2 years and 2.1-4 years) and OC (age 2.1-4 years) levels were higher, while β-CTX (age 2.1-4 years and 4.1-7 years) and PTH (age 2.1-4 years) values were lower than the control group. In addition, there were no statistically significant differences in the levels of these indicators among the CP subgroups with different clinical characteristics. Our study shows that bone turnover markers, indicators of bone metabolism, in children with CP differ significantly from healthy controls. The indicators we studied changed with age, and they did not correlate with disease severity.
Collapse
Affiliation(s)
| | | | | | | | - Zhizhong Liu
- Department of Clinical Laboratory, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| |
Collapse
|
14
|
Muggeo P, Grassi M, D'Ascanio V, Brescia V, Fontana A, Piacente L, Di Serio F, Giordano P, Faienza MF, Santoro N. Bone Remodeling Markers in Children with Acute Lymphoblastic Leukemia after Intensive Chemotherapy: The Screenshot of a Biochemical Signature. Cancers (Basel) 2023; 15:cancers15092554. [PMID: 37174020 PMCID: PMC10177249 DOI: 10.3390/cancers15092554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
PURPOSE to investigate the effects of intensive chemotherapy and glucocorticoid (GC) treatment on bone remodeling markers in children with acute lymphoblastic leukemia (ALL). METHODS A cross-sectional study was carried out in 39 ALL children (aged 7.64 ± 4.47) and 49 controls (aged 8.7 ± 4.7 years). Osteoprotegerin (OPG), receptor activator of NF-κB ligand (RANKL), osteocalcin (OC), C-terminal telopeptide of type I collagen (CTX), bone alkaline phosphatase (bALP), tartrate-resistant acid phosphatase 5b (TRACP5b), procollagen type I N-terminal propeptide (P1NP), Dickkopf-1 (DKK-1), and sclerostin were assessed. Statistical analysis was conducted using the principal component analysis (PCA) to study patterns of associations in bone markers. RESULTS ALL patients showed significantly higher OPG, RANKL, OC, CTX, and TRACP5b than the controls (p ≤ 0.02). Considering ALL group, we found a strong positive correlation among OC, TRACP5b, P1NP, CTX, and PTH (r = 0.43-0.69; p < 0.001); between CTX and P1NP (r = 0.5; p = 0.001); and between P1NP and TRAcP (r = 0.63; p < 0.001). The PCA revealed OC, CTX, and P1NP as the main markers explaining the variability of the ALL cohort. CONCLUSIONS Children with ALL showed a signature of bone resorption. The assessment of bone biomarkers could help identify ALL individuals who are most at risk of developing bone damage and who need preventive interventions.
Collapse
Affiliation(s)
- Paola Muggeo
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinic, 70124 Bari, Italy
| | - Massimo Grassi
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinic, 70124 Bari, Italy
| | - Vito D'Ascanio
- Institute of Sciences of Food Production (ISPA), Italian National Research Council (CNR), 70126 Bari, Italy
| | - Vincenzo Brescia
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | - Antonietta Fontana
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | - Laura Piacente
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University "A. Moro", 70124 Bari, Italy
| | - Francesca Di Serio
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | - Paola Giordano
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University "A. Moro", 70124 Bari, Italy
| | - Nicola Santoro
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinic, 70124 Bari, Italy
| |
Collapse
|
15
|
Galeone A, Grano M, Brunetti G. Tumor Necrosis Factor Family Members and Myocardial Ischemia-Reperfusion Injury: State of the Art and Therapeutic Implications. Int J Mol Sci 2023; 24:4606. [PMID: 36902036 PMCID: PMC10003149 DOI: 10.3390/ijms24054606] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Ischemic heart disease is the principal cause of death worldwide and clinically manifests as myocardial infarction (MI), stable angina, and ischemic cardiomyopathy. Myocardial infarction is defined as an irreversible injury due to severe and prolonged myocardial ischemia inducing myocardial cell death. Revascularization is helpful in reducing loss of contractile myocardium and improving clinical outcome. Reperfusion rescues myocardium from cell death but also induces an additional injury called ischemia-reperfusion injury. Multiple mechanisms are involved in ischemia-reperfusion injury, such as oxidative stress, intracellular calcium overload, apoptosis, necroptosis, pyroptosis, and inflammation. Various members of the tumor necrosis factor family play a key role in myocardial ischemia-reperfusion injury. In this article, the role of TNFα, CD95L/CD95, TRAIL, and the RANK/RANKL/OPG axis in the regulation of myocardial tissue damage is reviewed together with their potential use as a therapeutic target.
Collapse
Affiliation(s)
- Antonella Galeone
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| |
Collapse
|
16
|
Brain-Type Creatine Kinase Release from Cultured Osteoclasts Exposed to Neridronate in Children Affected by Osteogenesis Imperfecta Type 1. Biomedicines 2023; 11:biomedicines11020458. [PMID: 36830994 PMCID: PMC9953364 DOI: 10.3390/biomedicines11020458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Brain-type creatine kinase (CK-BB) increases during osteoclastogenesis, with high circulating amounts in type I osteogenesis imperfecta (OI) following treatment with neridronate, a bisphosphonate able to inhibit osteoclast activity and survival. The aim of this study was to demonstrate the correlation between osteoclastogenesis and CK-BB release from OI patients' osteoclasts treated with different concentrations of neridronate. Our patients showed reduced bone quality, increased levels of CTX I, a marker of bone resorption, and decreased levels of OPG, an inhibitor of osteoclastogenesis. In OI patients, the presence of MCSF and RANKL determined an increased secretion of CK-BB from osteoclasts (p = 0.04) compared with control conditions without these cytokines; interestingly, in the absence of these factors, the secretion of CK-BB is significantly elevated at 3 µmol/L compared with 0.03 and 1 µmol/L (p = 0.007). In healthy donors' cultures, the higher concentration of CK-BB can be detected following stimulation with 3 µmol/L neridronate compared with the untreated condition both with and without MCSF and RANKL (p = 0.03 and p = 0.006, respectively). Consistently, in osteoclast cultures, neridronate treatment is associated with a decrease in multinucleated TRAP+ cells, together with morphology changes typical of apoptosis. Consistently, in the media of the same osteoclast cultures, we demonstrated a significant increase in caspase-3 levels. In conclusion, our findings support the idea that CK-BB levels increase in the serum of OI-treated patients.
Collapse
|
17
|
Faienza MF, Pontrelli P, Brunetti G. Type 2 diabetes and bone fragility in children and adults. World J Diabetes 2022; 13:900-911. [PMID: 36437868 PMCID: PMC9693736 DOI: 10.4239/wjd.v13.i11.900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/17/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022] Open
Abstract
Type 2 diabetes (T2D) is a global epidemic disease. The prevalence of T2D in adolescents and young adults is increasing alarmingly. The mechanisms leading to T2D in young people are similar to those in older patients. However, the severity of onset, reduced insulin sensitivity and defective insulin secretion can be different in subjects who develop the disease at a younger age. T2D is associated with different complications, including bone fragility with consequent susceptibility to fractures. The purpose of this systematic review was to describe T2D bone fragility together with all the possible involved pathways. Numerous studies have reported that patients with T2D show preserved, or even increased, bone mineral density compared with controls. This apparent paradox can be explained by the altered bone quality with increased cortical bone porosity and compr-omised mechanical properties. Furthermore, reduced bone turnover has been described in T2D with reduced markers of bone formation and resorption. These findings prompted different researchers to highlight the mechanisms leading to bone fragility, and numerous critical altered pathways have been identified and studied. In detail, we focused our attention on the role of microvascular disease, advanced glycation end products, the senescence pathway, the Wnt/β-catenin pathway, the osteoprotegerin/receptor-activator of nuclear factor kappa B ligand, osteonectin and fibroblast growth factor 23. The understanding of type 2 myeloid bone fragility is an important issue as it could suggest possible interventions for the prevention of poor bone quality in T2D and/or how to target these pathways when bone disease is clearly evident.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari Aldo Moro, Bari 70124, Italy
| | - Paola Pontrelli
- Division of Nephrology, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari 70124, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari 70125, Italy
| |
Collapse
|
18
|
Rodriguez-Merchan EC. Osteoporosis in hemophilia: what is its importance in clinical practice? Expert Rev Hematol 2022; 15:697-710. [PMID: 35912904 DOI: 10.1080/17474086.2022.2108783] [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: 03/31/2022] [Accepted: 07/29/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The relationship between severe hemophilia and osteoporosis has been well established in the literature. However, although the importance of its prevention in order to reduce the risk of bone fractures has been reported, the importance of its treatment in clinical practice has not been well analyzed. AREAS COVERED In this paper, a review of the available clinical and experimental information on osteoporosis in hemophilia has been performed, to better understand the relationship between hemophilia and osteoporosis. Prevention of osteoporosis in hemophilia should include primary hematological prophylaxis; a diet appropriate in calcium and vitamin D; a regular exercise program that includes aerobics, strength training and balance and flexibility activities; restriction of tobacco and alcohol use; and limitation of the duration of immobilization. EXPERT OPINION Prevention of osteoporosis in hemophilic patients is paramount. However, it is noteworthy that there is only one publication on the treatment of osteoporosis in patients with hemophilia. Until further research is done on this topic, the existing recommendations for non-hemophilic patients should be followed. They include the use of antiresorptives (estrogens, selective estrogen receptor modulators, bisphosphonates, denosumab) and anabolic agents (teriparatide, abaloparatide, romosozumab). Further studies on the management of osteoporosis in patients with hemophilia are required.
Collapse
Affiliation(s)
- E Carlos Rodriguez-Merchan
- Department of Orthopaedic Surgery, La Paz University Hospital, Madrid, Spain
- Osteoarticular Surgery Research, Hospital La Paz Institute for Health Research, IdiPAZ (La Paz University Hospital - Autonomous University of Madrid), Madrid, Spain
| |
Collapse
|
19
|
Chiarito M, Piacente L, Chaoul N, Pontrelli P, D'Amato G, Grandone A, Russo G, Street ME, Wasniewska MG, Brunetti G, Faienza MF. Role of Wnt-signaling inhibitors DKK-1 and sclerostin in bone fragility associated with Turner syndrome. J Endocrinol Invest 2022; 45:1255-1263. [PMID: 35237949 PMCID: PMC9098532 DOI: 10.1007/s40618-022-01760-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/07/2022] [Indexed: 12/22/2022]
Abstract
PURPOSE Girls affected with Turner syndrome (TS) present with low bone mineral density (BMD) and osteopenia/osteoporosis. Thus, they have an increased risk to develop fractures compared to normal population. The aim of this study was to deepen the pathophysiology of skeletal fragility in TS subjects by evaluating the serum levels of Dickkopf-1 (DKK-1) and sclerostin, main regulators of bone mass, as well as the percentage of circulating osteoblast precursors (OCPs). METHODS Thirty-four TS girls and 24 controls were recruited. All subjects underwent anthropometric measures (height, weight, body mass index-BMI). A peripheral venous blood sample was collected to determine serum levels of active intact parathyroid hormone (PTH), 25-OH vitamin D, calcium, phosphorus, bone alkaline phosphatase (bALP), osteocalcin, sclerostin, DKK-1, RANKL and OPG. OCPs were detected by flow cytometry. In TS subjects bone mineralization was measured at lumbar spine by dual energy X-ray absorptiometry (DXA). RESULTS bALP, 25-OH Vitamin D, and osteocalcin levels were significant lower in TS subjects than in the controls. Statistically significant higher levels of sclerostin, DKK-1 and RANKL were measured in patients compared with the controls. The percentage of OCPs did not show significant differences between patients and controls. Sclerostin and DKK-1 levels were related with anthropometric parameters, bone metabolism markers, HRT, rhGH therapy, RANKL and lumbar BMAD-Z-score. CONCLUSION TS patients showed higher levels of sclerostin and DKK-1 than controls which can be related to HRT, and to reduced bone formation markers as well as the increased bone resorption activity.
Collapse
Affiliation(s)
- M Chiarito
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari "A. Moro", Bari, Italy
| | - L Piacente
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari "A. Moro", Bari, Italy
| | - N Chaoul
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Bari, Italy
| | - P Pontrelli
- Department of Emergency and Organ Transplantation, Division of Nephrology, University of Bari "A. Moro", Bari, Italy
| | - G D'Amato
- Neonatal Intensive Care Unit, "Di Venere" Hospital, Bari, Italy
| | - A Grandone
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - G Russo
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - M E Street
- Department of Mother and Child, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - M G Wasniewska
- Pediatric Unit, Department of Human Pathology in Adulthood and Childhood, University of Messina, Messina, Italy
| | - G Brunetti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University "A. Moro" of Bari, Bari, Italy
| | - M F Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari "A. Moro", Bari, Italy.
| |
Collapse
|
20
|
Jazbinšek S, Koce M, Kotnik P. Novel Treatment Options in Childhood Bone Diseases. Horm Res Paediatr 2022; 96:590-598. [PMID: 35235937 DOI: 10.1159/000523868] [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: 11/29/2021] [Accepted: 02/17/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Several novel treatment options have recently become available in childhood bone diseases. The purpose of this article is to provide an update on some of the therapeutic agents used in the treatment of pediatric osteoporosis, X-linked hypophosphatemic rickets, and achondroplasia (ACH). SUMMARY Vitamin D3 and Ca supplementation remains the basis of childhood osteoporosis treatment. Bisphosphonate (BP) therapy is the main antiresorptive therapeutic option, while denosumab, a human monoclonal IgG2 antibody with high affinity and specificity for a primary regulator of bone resorption - RANKL, represents a possible alternative. Its potent inhibition of bone resorption and turnover process leads to continuous increase of bone mineral density throughout the treatment also in the pediatric population. With a half-life much shorter than BPs, its effects are rapidly reversible upon discontinuation. Safety and dosing concerns in children remain. Novel treatment options have recently become available in two rare bone diseases. Burosumab, a monoclonal antibody against FGF-23, has been approved for the treatment of children with X-linked hypophosphatemic rickets older than 1 year. It presents an effective, more etiology-based treatment for rickets compared to conventional therapy, without the need for multiple daily oral phosphate supplementation. Its long-term efficacy and safety are currently being investigated. After years of anticipation, a novel treatment option for ACH has become available. C-type natriuretic peptide analog vosoritide effectively increases proportional growth and has a reasonable safety profile in children >2 years. Its effect on other features of the disease and the final height is yet to be determined. Several other treatment options for ACH exploring different therapeutic approaches are currently being investigated. KEY MESSAGES Denosumab is effective in the treatment of childhood-onset osteoporosis; however, further studies are necessary to determine the optimal treatment protocol. Burosumab is more etiology-based and convenient in comparison to conventional treatment of X-linked hypophospha--temic rickets in children and adults. Vosoritide importantly changes the natural course of achondroplasia, at least in the short term.
Collapse
Affiliation(s)
- Sončka Jazbinšek
- Division of Pediatrics, Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Maša Koce
- Division of Pediatrics, Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Primož Kotnik
- Division of Pediatrics, Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Division of Pediatrics, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
21
|
Gebetsberger J, Schirmer M, Wurzer WJ, Streif W. Low Bone Mineral Density in Hemophiliacs. Front Med (Lausanne) 2022; 9:794456. [PMID: 35186990 PMCID: PMC8849249 DOI: 10.3389/fmed.2022.794456] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Objective To review the current knowledge on bone health in patients with hemophilia A and the underlying pathogenetic mechanisms. Data Sources Original research articles, meta-analyses, and scientific reviews. Data Synthesis Already in childhood, patients with hemophilia A are prone to low bone mineral density, leading to osteopenia and/or osteoporosis. Initially associated with the life style of hemophilia, today we are faced with accumulating evidence that coagulation factor VIII is involved directly or indirectly in bone physiology. Conclusion Understanding the role of factor VIII and the mechanisms of decreased bone mineral density in hemophilia A is critically important, especially as non-factor replacement therapies are available, and treatment decisions potentially impact bone health.
Collapse
Affiliation(s)
| | - Michael Schirmer
- Department of Internal Medicine, Clinic II, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Werner Streif
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
ZHU Y, JIN S, ZHANG D, DUAN Z, ZHAO D, HAN K, GU S, LI T, HUO N. Sheep bone collagen peptide ameliorates osteoporosis by regulating RANK/RANKL/OPG signal pathway. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.18822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yixin ZHU
- College of Veterinary Medicine, China
| | | | | | - Zhuo DUAN
- College of Veterinary Medicine, China
| | | | | | | | - Tao LI
- College of Veterinary Medicine, China
| | | |
Collapse
|
23
|
Giordano P, Del Vecchio GC, Russo G, Palmieri VV, Piacente L, Fidone C, Urbano F, Faienza MF. High Dickkopf-1 (DKK-1) levels are associated with chronic inflammation in children with Sickle Cell Disease. Eur J Haematol 2021; 108:336-341. [PMID: 34962669 DOI: 10.1111/ejh.13741] [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: 11/13/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Sickle bone disease (SBD) is a chronic complication of Sickle Cell Disease (SCD) whose pathogenesis is not completely understood. Chronic inflammation associated with SCD could alter bone remodeling. Our aim was to analyze the serum levels of bone remodeling markers in a group of SCD children to evaluate their involvement in the SBD. METHODS We enrolled 26 SCD subjects and 26 age-matched controls, who lived in the same geographic area. DKK-1, sclerostin, RANKL, and OPG serum levels were evaluated. Neutrophil-lymphocyte ratio (NLR) was also evaluated as a marker of inflammation. RESULTS The analysis of bone remodeling markers did not show any significant difference between the two groups except for DKK-1 levels that were significantly higher in the patients than controls (p<0.05). A significant direct correlation between NLR and DKK-1 (p=0.004) was found. An inverse correlation between NLR and osteocalcin (p=0.01) has also been observed. CONCLUSIONS The chronic inflammation, which represents a peculiar characteristic in SCD patients, would represent the primary causal agent of the activation of osteoblastogenesis inhibitors responsible of bone impairment in these subjects. Further studies will be needed to better explain the role of these inhibitors in SCD, to prevent or treat bone damage in this population.
Collapse
Affiliation(s)
- Paola Giordano
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University "A. Moro", Bari, Italy
| | | | - Giovanna Russo
- Clinica di Onco-Ematologia Pediatrica, Azienda Ospedaliero Universitaria "Policlinico Vittorio Emanuele", Catania, Italy
| | - Viviana Valeria Palmieri
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University "A. Moro", Bari, Italy
| | - Laura Piacente
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University "A. Moro", Bari, Italy
| | | | | | - Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University "A. Moro", Bari, Italy
| |
Collapse
|
24
|
Shi P, Hou A, Li C, Wu X, Jia S, Cen H, Hu X, Gong H. Continuous subcutaneous insulin infusion ameliorates bone structures and mechanical properties in type 2 diabetic rats by regulating bone remodeling. Bone 2021; 153:116101. [PMID: 34245934 DOI: 10.1016/j.bone.2021.116101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/21/2021] [Accepted: 07/01/2021] [Indexed: 12/29/2022]
Abstract
Continuous subcutaneous insulin infusion (CSII) is an intensive insulin therapy for patients with type 2 diabetes mellitus (T2DM) who have poor glycemic control, but its effect on T2DM-related bone disorder is unclear. This study described the possible mechanisms by which CSII affects bone remodeling, structures, and mechanical properties in T2DM rats. Herein, male rats (6-week-old) were assigned randomly to 4-week and 8-week administration groups, each of which included healthy control, T2DM, CSII, and Placebo groups. Then, metabolic markers, bone formation and resorption markers in serum and protein expressions of osteoclastogenesis regulators in tibias were detected. Meanwhile, microstructures, nanostructures, macro-mechanical properties, nano-mechanical properties, and mineral compositions in femurs were evaluated. 4-week later, CSII treatment restored circulatory metabolites, bone formation and resorption markers, and osteoclastogenesis regulators, improved certain bone microstructures, decreased matrix mineralization, and increased fracture toughness in T2DM rats. For 8-week group, CSII treatment restored bone formation and resorption markers, osteoclastogenesis regulators, and bone microstructures, besides improved bone mineral compositions and nanostructures, enhanced bone mechanical properties such as fracture toughness, maximum load, elastic modulus, indentation modulus and hardness. Collectively, 8-week CSII treatment is more conducive to ameliorating bone structures and mechanical properties in T2DM rats by regulating bone remodeling compared with 4-week CSII treatment, thus improving whole bone quality and providing valuable information for clinical prevention and treatment of T2DM-related bone disorders.
Collapse
Affiliation(s)
- Peipei Shi
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Aiqi Hou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Chenchen Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiaodan Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shaowei Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Haipeng Cen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiaorong Hu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
25
|
Karim K, Giribabu N, Salleh N. Marantodes pumilum Var Alata (Kacip Fatimah) ameliorates derangement in RANK/RANKL/OPG pathway and reduces inflammation and oxidative stress in the bone of estrogen-deficient female rats with type-2 diabetes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153677. [PMID: 34333329 DOI: 10.1016/j.phymed.2021.153677] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND M. pumilum has been claimed to protect the bone against the adverse effect of estrogen deficiency. Additionally, it also exhibits anti-diabetic activity. In view of these, this study aims to identify the mechanisms underlying the bone protective effect of M. pumilum in the presence of both estrogen deficiency and diabetes mellitus (DM). METHODS Ovariectomized, diabetic female rats were given M. pumilum leave aqueous extract (MPLA) (50 and 100 mg/kg/day), estrogen, glibenclamide and estrogen plus glibenclamide for 28 consecutive days. At the end of the treatment, fasting blood glucose (FBG), serum insulin, Ca2+, PO43- and bone alkaline phosphatase (BALP) levels were measured. Rats were sacrificed and femur bones were harvested for determination of expression level and distribution of RANK, RANKL, OPG and oxidative stress and inflammatory proteins by molecular biological techniques. RESULTS 100 mg/kg/day MPLA treatment decreased the FBG and BALP levels but increased the serum insulin, Ca2+ and PO43- levels in estrogen deficient, diabetic rats. Expression and distribution of RANKL, NF-κB p65, IKKβ, IL-6, IL-1β and Keap-1 decreased however expression and distribution of RANK, OPG, BMP-2, Type-1 collagen, Runx2, TRAF6, Nrf2, NQO-1, HO-1, SOD and CAT increased in the bone of estrogen deficient, diabetic rats which received 100 mg/kg/day MPLA with greater effects than estrogen-only, glibenclamide-only and estrogen plus glibenclamide treatments. CONCLUSION MPLA helps to overcome the adverse effect of estrogen deficiency and DM on the bone and thus this herb could potentially be used for the treatment and prevention of osteoporosis in postmenopausal women with diabetes.
Collapse
Affiliation(s)
- Kamarulzaman Karim
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
26
|
Brunetti G, D'Amato G, De Santis S, Grano M, Faienza MF. Mechanisms of altered bone remodeling in children with type 1 diabetes. World J Diabetes 2021; 12:997-1009. [PMID: 34326950 PMCID: PMC8311475 DOI: 10.4239/wjd.v12.i7.997] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/17/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Bone loss associated with type 1 diabetes mellitus (T1DM) begins at the onset of the disease, already in childhood, determining a lower bone mass peak and hence a greater risk of osteoporosis and fractures later in life. The mechanisms underlying diabetic bone fragility are not yet completely understood. Hyperglycemia and insulin deficiency can affect the bone cells functions, as well as the bone marrow fat, thus impairing the bone strength, geometry, and microarchitecture. Several factors, like insulin and growth hormone/insulin-like growth factor 1, can control bone marrow mesenchymal stem cell commitment, and the receptor activator of nuclear factor-κB ligand/osteoprotegerin and Wnt-b catenin pathways can impair bone turnover. Some myokines may have a key role in regulating metabolic control and improving bone mass in T1DM subjects. The aim of this review is to provide an overview of the current knowledge of the mechanisms underlying altered bone remodeling in children affected by T1DM.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University "A. Moro" of Bari, Bari 70125, Italy
| | - Gabriele D'Amato
- Department of Women’s and Children’s Health, ASL Bari, Neonatal Intensive Care Unit, Di Venere Hospital, Bari 70124, Italy
| | - Stefania De Santis
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, Bari 70126, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, Univ Bari, Bari 70124, Italy
| | - Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University "A.Moro", Bari 70124, Italy
| |
Collapse
|
27
|
Mechanisms of Bone Impairment in Sickle Bone Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041832. [PMID: 33668588 PMCID: PMC7918363 DOI: 10.3390/ijerph18041832] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 11/23/2022]
Abstract
Sickle bone disease (SBD) is a chronic and invalidating complication of Sickle cell disease (SCD), a multisystem autosomal recessive genetic disorder affecting millions of people worldwide. Mechanisms involved in SBD are not completely known, especially in pediatric age. Among the hypothesized pathogenetic mechanisms underlying SBD are bone marrow compensatory hyperplasia and bone ischemic damage, both secondary to vaso-occlusive crisis (VOC), which leads to cell sickling, thus worsening local hypoxia with a negative impact on osteoblast recruitment. Furthermore, the hypoxia is a strong activator of erythropoietin, which in turn stimulates osteoclast precursors and induces bone loss. Hemolysis and iron overload due to a chronic transfusion regimen could also contribute to the onset of bone complications. Vitamin D deficiency, which is frequently seen in SCD subjects, may worsen SBD by increasing the resorptive state that is responsible for low bone mineral density, acute/chronic bone pain, and high fracture risk. An imbalance between osteoblasts and osteoclasts, with a relative decrease of osteoblast recruitment and activity, is a further possible mechanism responsible for the impairment of bone health in SCD. Moreover, delayed pubertal growth spurt and low peak bone mass may explain the high incidence of fracture in SCD adolescents. The aim of this review was to focus on the pathogenesis of SBD, updating the studies on biochemical, instrumental, and biological markers of bone metabolism. We also evaluated the growth development and endocrine complications in subjects affected with SCD.
Collapse
|
28
|
Correlation between Serum Bone Turnover Markers and Estimated Glomerular Filtration Rate in Chinese Patients with Diabetes. DISEASE MARKERS 2021; 2021:6731218. [PMID: 33505536 PMCID: PMC7806398 DOI: 10.1155/2021/6731218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022]
Abstract
Objective Diabetes is a growing global public health concern with many significant disease complications. Multiple studies show that bone turnover markers (BTMs) are decreased in diabetes patients, indicating impaired bone metabolism in diabetes patients. A recent study also showed that in diabetes patients, BTMs are correlated with urine albumin to creatinine ratio, an indicator of nephropathy. However, whether BTMs are correlated with estimated glomerular filtration rate (eGFR) in diabetes remains unknown. This retrospective study accessed correlations between serum BTMs and eGFR in Chinese patients with diabetes and compare levels of BTMs and eGFR between diabetic patients and healthy individuals. Methods This study analyzed data from 221 diabetic patients (include type1 and type 2 diabetes) and 155 healthy individuals. Serum BTM levels and eGFR were compared between diabetic patients and healthy individuals. Pearson correlation analysis was used to assess correlations between BTMs and eGFR. Multiple logistic regression analysis adjusted for gender and age was performed to measure odd ratio (OR) and 95% confidence interval (95% CI) of BTMs on diabetes. Results Patients with diabetes had significant lower 25-hydroxyvitamin D (25(OH)D) levels (15.07 ± 6.20 ng/mL) than healthy group (17.89 ± 6.41 ng/mL) (P < 0.05). For patients with diabetes, eGFR was negatively correlated with osteocalcin (OC) (r = −0.434, P < 0.05), procollagen type 1 intact N-terminal propeptide (P1NP) (r = −0.350, P < 0.05), and β-carboxy-terminal cross-linking telopeptide of type I collagen (β-CTX) (r = −0.179, P < 0.05) levels. For healthy people, eGFR was negatively correlated with 25(OH)D (r = −0.290, P < 0.05) levels. Multiple logistic regression analysis adjusted for age and gender (mean age of diabetes was 64.9 years and the percentage of female was 66.9%, mean age of healthy people was 48.4 years and the percentage of female was 37.4%) showed that 25(OH)D (OR = 0.909, 95%CI = 0.862 − 0.959, P < 0.05) was protective factors for diabetes. Conclusions In the stage of diabetic nephropathy, bone turnover may accelerate. It is important to detect BTMs in the stage of diabetic nephropathy.
Collapse
|
29
|
Faienza MF, Chiarito M, Brunetti G, D'Amato G. Growth plate gene involment and isolated short stature. Endocrine 2021; 71:28-34. [PMID: 32504378 DOI: 10.1007/s12020-020-02362-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Short stature is a common clinical presentation, thus it is widely accepted that it is a polygenic trait. However, genome wide association and next generation sequencing studies have recently challenged this view, suggesting that many of the children classified as idiopathic short stature could instead have monogenic defects. Linear growth is determined primarily by chondrogenesis at the growth plate. This process results from chondrocyte proliferation, hypertrophy, and extracellular matrix secretion, and it is perfectly coordinated by complex networks of local paracrine and endocrine factors. Alterations in genes which control growth plate development can explain a large number of cases of isolated short stature, allowing an etiological diagnosis. METHODS/RESULTS We reviewed recent data on the genetic alterations in fundamental cellular processes, paracrine signaling, and cartilage matrix formation associated with impaired growth plate chondrogenesis. In particular we focused on growth plate gene involvement in nonsyndromic short stature. CONCLUSIONS The identification of genetic basis of growth failure will have a significant impact on the care of children affected with short stature.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Paediatric Unit, Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy.
| | - Mariangela Chiarito
- Paediatric Unit, Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Human Anatomy and Histology, University of Bari "A. Moro", Bari, Italy
| | | |
Collapse
|
30
|
Chiarito M, Brunetti G, D'Amato G, Faienza MF. Monitoring and maintaining bone health in patients with Turner syndrome. Expert Rev Endocrinol Metab 2020; 15:431-438. [PMID: 33074770 DOI: 10.1080/17446651.2020.1834846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Subjects affected with Turner Syndrome (TS) suffer low bone mineral density and high risk of fracture from a young age. Estrogen deficiency is considered the main risk factor but other factors, such as intrinsic bone abnormalities, enhanced osteoclastogenesis, vitamin D deficiency and other comorbidities may contribute to the exalted bone fragility. AREAS COVERED The authors performed a literature search in PubMed and EMBASE, using selected key words. They focused their search on pathogenetic mechanisms of osteoporosis in TS and updated the diagnosis, prevention and therapeutic interventions. EXPERT OPINION Bone health is a concern in subjects with TS, and strategies to prevent osteoporosis and fractures should be considered from childhood. Advice on how to live a healthy lifestyle, including physical activity and correct nutrition, should be given during childhood in order to prevent bone impairment later in life. The screening for vitamin D deficiency should be performed between the ages of 9 and 11, and every 2-3 years thereafter. Early initiation of estrogen replacement therapy (ERT) between 11-12 years of age, prompt titration to the adult dose after 2 years, and long-term follow-up to guarantee compliance with ERT, are the key points of osteoporosis prevention in women with TS.
Collapse
Affiliation(s)
- Mariangela Chiarito
- Department of Biomedical Sciences and Human Oncology, University "A.Moro" , Bari, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Human Anatomy and Histology, University "A. Moro" , Bari, Italy
| | - Gabriele D'Amato
- Department of Women's and Children's Health, ASL Bari, Neonatal Intensive Care Unit, Di Venere Hospital , Bari, Italy
| | - Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, University "A.Moro" , Bari, Italy
| |
Collapse
|
31
|
How Physical Activity across the Lifespan Can Reduce the Impact of Bone Ageing: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17061862. [PMID: 32183049 PMCID: PMC7143872 DOI: 10.3390/ijerph17061862] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Bone remodeling is a lifelong process, due to the balanced activity of the osteoblasts (OBs), the bone-forming cells, and osteoclasts (OCs), the bone-resorbing cells. This equilibrium is mainly regulated by the WNT-ß-cathenin pathway and the RANK-RANKL/OPG system, respectively. Bone ageing is a process which normally occurs during life due to the imbalance between bone formation and bone resorption, potentially leading to osteoporosis. Bone loss associated with bone ageing is determined by oxidative stress, the result of the increasing production of reactive oxygen species (ROS). The promotion of physical exercise during growth increases the chances of accruing bone and delaying the onset of osteoporosis. Several studies demonstrate that physical exercise is associated with higher bone mineral density and lower fracture incidence, and the resulting bone mineral gain is maintained with ageing, despite a reduction of physical activity in adulthood. The benefits of exercise are widely recognized, thus physical activity is considered the best non-pharmacologic treatment for pathologies such as osteoporosis, obesity, diabetes and cardiovascular disease. We reviewed the physiological mechanisms which control bone remodeling, the effects of physical activity on bone health, and studies on the impact of exercise in reducing bone ageing.
Collapse
|
32
|
Bao T, Yang K, Long Z, Zeng L, Li Y. Systematic Pharmacological Methodology to Explore the Pharmacological Mechanism of Siwu Decoction for Osteoporosis. Med Sci Monit 2019; 25:8152-8171. [PMID: 31666500 PMCID: PMC6844540 DOI: 10.12659/msm.917393] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/08/2019] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis is an important health problem worldwide. Siwu decoction (SWD) and its modification have a good clinical effect on osteoporosis. However, the molecular mechanism of SWD on osteoporosis has not been thoroughly explained. A systematic pharmacological methodology was utilized to predict the active compounds and potential targets of SWD, collect the genes of osteoporosis and the known targets of SWD, and analyze the osteoporosis and SWD's network. Five networks were constructed and analyzed: (1) Osteoporosis genes' protein-protein interaction (PPI) network; (2) Compound-compound target network of SWD; (3) SWD-osteoporosis PPI network; (4) Compound-known target network of SWD; and (5) SWD known target- osteoporosis PPI network. Several osteoporosis and treatment-related targets (eg.,. HSP90AB1, FGFR1, HRAS, GRB2, and PGF), clusters, biological processes, and signaling pathways (e.g., PI3K-Akt signaling pathway, insulin signaling pathway, MAPK signaling pathway and FoxO signaling pathway) were found. The therapeutic effect of SWD on osteoporosis may be achieved by interfering with the biological processes and signaling pathways related to the development of osteoporosis.
Collapse
Affiliation(s)
- Tingting Bao
- Beijing University of Chinese Medicine, Beijing, P.R. China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P.R. China
| | - Kailin Yang
- Capital Medical University, Beijing, P.R. China
- Beijing Anzhen Hospital, Capital Medical University, Beijing, P.R. China
| | - Zhiyong Long
- Shantou University Medical College, Shantou, Guangdong, P.R. China
- Department of Rehabilitation Medicine, Guangdong Geriatric Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Liuting Zeng
- Hunan University of Chinese Medicine, Changsha, Hunan, P.R. China
| | - Yuehua Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P.R. China
| |
Collapse
|
33
|
Cannarella R, Barbagallo F, Condorelli RA, Aversa A, La Vignera S, Calogero AE. Osteoporosis from an Endocrine Perspective: The Role of Hormonal Changes in the Elderly. J Clin Med 2019; 8:jcm8101564. [PMID: 31581477 PMCID: PMC6832998 DOI: 10.3390/jcm8101564] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/09/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Introduction: Osteoporosis is increasingly prevalent in the elderly, with fractures mostly occurring in women and men who are older than 55 and 65 years of age, respectively. The aim of this review was to examine the evidence regarding the influence of hormones on bone metabolism, followed by clinical data of hormonal changes in the elderly, in the attempt to provide possible poorly explored diagnostic and therapeutic candidate targets for the management of primary osteoporosis in the aging population. Material and methods: An extensive Medline search using PubMed, Embase, and Cochrane Library was performed. Results: While the rise in Thyroid-stimulating hormone (TSH) levels has a protective role on bone mass, the decline of estrogen, testosterone, Insulin-like growth factor 1 (IGF1), and vitamin D and the rise of cortisol, parathyroid hormone, and follicle-stimulating hormone (FSH) favor bone loss in the elderly. Particularly, the AA rs6166 FSH receptor (FSHR) genotype, encoding for a more sensitive FSHR than that encoded by the GG one, is associated with low total body mass density (BMD), independently of circulating estrogen. A polyclonal antibody with a FSHR-binding sequence against the β-subunit of murine FSH seems to be effective in ameliorating bone loss in ovariectomized mice. Conclusions: A complete hormonal assessment should be completed for both women and men during bone loss evaluation. Novel possible diagnostic and therapeutic tools might be developed for the management of male and female osteoporosis.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Federica Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
34
|
Garcia AP, Hatvany JB, Murphy MA, Atchley DH, Gurley BJ, Kamdem LK. Effect of Aromatase Inhibition (Exemestane) on Urine Concentration of Osteoprotegerin in Healthy Postmenopausal Women. J Clin Pharmacol 2019; 60:209-217. [PMID: 31535401 DOI: 10.1002/jcph.1519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/12/2019] [Indexed: 11/06/2022]
Abstract
This pilot study examined how exemestane (an aromatase inhibitor [AI]) affected osteoprotegerin (OPG) urine concentrations in postmenopausal women. Exemestane (25 mg, single dose) was given to 14 disease-free women past menopause in this nonrandomized, open-label study. Before dosing, urine specimens were gathered. Three days later, these women returned to provide urine specimens for pharmacokinetic (measurement of major parent drug and enzymatic product) and pharmacodynamic (profiling of OPG) analysis. Urine concentrations of the major parent drug (exemestane) and enzymatic product (17-hydroexemestane) were quantified using liquid chromatography-tandem mass spectrometry. An analyst software package was used for data processing. Following the manufacturer's guidelines, OPG urine concentrations were quantified using a human osteoprotegerin TNFRSF11b ELISA kit from Sigma-Aldrich. A microplate reader helped to carry out OPG data analysis and processing. Our results highlight that OPG urine concentrations were decreased 3 days after drug dosage (mean predosage OPG concentration, 61.4 ± 24.1 pg/mL; vs mean postdosage OPG concentration, 45.7 ± 22.1 pg/mL; P = .02, Wilcoxon rank test). Among the 14 volunteers enrolled in the study, 4 subjects had an increase of less than 1-fold, and the rest showed an average of a 2-fold decrease in OPG concentration (range, 1.1-5.4; standard deviation, 1.3) after exemestane administration. There was no association between fold decrease in OPG urine concentration and the pharmacokinetics of the major parent drug (exemestane) and its enzymatic product (17-hydroexemestane). We concluded that one of the off-target pharmacological effects of AIs (eg ,exemestane) may result in the reduction of osteoprotegerin.
Collapse
Affiliation(s)
| | - Jacob B Hatvany
- Harding University College of Pharmacy, Searcy, Arkansas, USA
| | - Michael A Murphy
- Harding University Physician Assistant Program, Searcy, Arkansas, USA
| | - Daniel H Atchley
- University of Pikeville-Kentucky College of Osteopathic Medicine, KYCOM, Pikeville, Kentucky, USA
| | - Bill J Gurley
- University of Arkansas for Medical Sciences, College of Pharmacy, Little Rock, Arkansas, USA
| | - Landry K Kamdem
- Harding University College of Pharmacy, Searcy, Arkansas, USA
| |
Collapse
|