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
|
Tracey LJ, El-Maklizi M, Sokolowski DJ, Gams MS, Brooke-Bisschop T, Ruston J, Taylor C, Khozin A, Rajakumar SA, Wilson MD, Guidos CJ, Justice MJ. Functional and molecular single-cell analyses implicate PRDM14 in the initiation of B cell leukemia in mice. Sci Rep 2025; 15:8827. [PMID: 40087379 PMCID: PMC11909259 DOI: 10.1038/s41598-025-93043-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
The transcription factor Prdm14 is a potent oncogene implicated in the initiation of many cancers. PRDM14 resets and maintains the pluripotent state in normal cells, but the molecular mechanisms through which PRDM14 drives oncogenesis are poorly understood. Here, we interrogated the heterogeneity of Prdm14-expressing cells in a T cell lymphoblastic leukemia/lymphoma mouse model. Using mass cytometry (CyTOF) of bone marrow at a pre-leukemic timepoint, an unexpected abnormal progenitor B cell population was identified. Prdm14-expressing progenitor B cells demonstrated short-term self-renewal and a block in differentiation when transferred to syngeneic hosts. Consistently, aged host mice succumb to a highly penetrant B-LL. Single-cell RNA-seq analyses suggests that the expression signature of these pre-leukemia cells is more consistent with that of B-1 cells than B-2 cells. B-1 cells are a self-renewing population of unconventional B cells established during embryonic development. Overlaying the chromatin binding of transcriptional marks H3K4me1 and H3K4me3 with PRDM14 suggests that PRDM14 initiates cancers through promiscuous DNA binding, activating oncogenic pathways and skewing development towards a self-renewing B-1-like phenotype. Together, our data show that Prdm14 can initiate premature T and B cell cancer programs when expressed in hematopoietic progenitor cells.
Collapse
Affiliation(s)
- Lauren J Tracey
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Mahmoud El-Maklizi
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Dustin J Sokolowski
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Miki S Gams
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Travis Brooke-Bisschop
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Julie Ruston
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Christine Taylor
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Alexandra Khozin
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Sujeetha A Rajakumar
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Michael D Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Cynthia J Guidos
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Monica J Justice
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
3
|
Ma G, Cheng S, Han Y, Tang W, Pang W, Chen L, Ding Z, Cao H. The p53-miR17 family-Rankl axis bridges liver-bone communication. Mol Ther 2025; 33:631-648. [PMID: 40308192 PMCID: PMC11853355 DOI: 10.1016/j.ymthe.2024.12.046] [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: 07/15/2024] [Revised: 11/08/2024] [Accepted: 12/27/2024] [Indexed: 05/02/2025] Open
Abstract
Our study elucidates the crucial role of the liver in bone homeostasis through the p53-miR17 family (miR17-miR20/miR20-miR106/miR93-miR106)-Rankl axis. We demonstrate the enhanced hepatocyte Rankl expression in inflammaging conditions, such as aging, ovariectomized (OVX) mice, and elderly humans. Mice with hepatocyte-specific Rankl deletion exhibit significant resistance to bone mass loss associated with aging, lipopolysaccharide (LPS)-induced inflammation, or estrogen deficiency, compared with controls. Our study highlights hepatocytes as the primary source of Rankl in the liver and serum under these conditions. We identify the p53-miR17 family axis as a crucial regulator for hepatocyte Rankl expression, with p53 inhibiting the miR17 family transcription. Through bioinformatics analysis and in vitro validation, we identify Rankl mRNA as a direct target of the miR17 family. Targeting this axis via CasRx-mediated mRNA editing or miRNA interference significantly attenuates bone mass loss in mice. Our investigation underscores the pivotal significance and therapeutic potential of modulating the p53-miR17 family-Rankl axis in the treatment of inflammaging-associated osteoporosis.
Collapse
Affiliation(s)
- Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Siyuan Cheng
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yingying Han
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Pang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhen Ding
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
4
|
Xiang XN, He HC, He CQ. Advances in mechanism and management of bone homeostasis in osteonecrosis: a review article from basic to clinical applications. Int J Surg 2025; 111:1101-1122. [PMID: 39311934 PMCID: PMC11745759 DOI: 10.1097/js9.0000000000002094] [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: 03/04/2024] [Accepted: 09/15/2024] [Indexed: 01/23/2025]
Abstract
Osteonecrosis, characterized by bone cell death leading to impaired bone recovery, causes challenges in bone homeostasis maintenance. Bone homeostasis relies on the delicate balance between osteoclasts and osteoblasts, encompassing a series of complex and strictly regulated biological functions. Current treatments, including conservative therapies and surgeries, often fall short of expected outcomes, necessitating a reorientation towards more effective therapeutic strategies according to the pathogenesis. In this review, the authors hierarchically outlined risk factors, emerging mechanisms, and last-decade treatment approaches in osteonecrosis. By connecting mechanisms of bone homeostasis, the authors proposed future research directions should be focused on elucidating risk factors and key molecules, performing high-quality clinical trial, updating practice, and accelerating translational potential.
Collapse
Affiliation(s)
- Xiao-Na Xiang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, P. R. China
| | - Hong-Chen He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, P. R. China
| | - Cheng-Qi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, P. R. China
| |
Collapse
|
5
|
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
|
6
|
Hong J, Luo F, Du X, Xian F, Li X. The immune cells in modulating osteoclast formation and bone metabolism. Int Immunopharmacol 2024; 133:112151. [PMID: 38685175 DOI: 10.1016/j.intimp.2024.112151] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Osteoclasts are pivotal in regulating bone metabolism, with immune cells significantly influencing both physiological and pathological processes by modulating osteoclast functions. This is particularly evident in conditions of inflammatory bone resorption, such as rheumatoid arthritis and periodontitis. This review summarizes and comprehensively analyzes the research progress on the regulation of osteoclast formation by immune cells, aiming to unveil the underlying mechanisms and pathways through which diseases, such as rheumatoid arthritis and periodontitis, impact bone metabolism.
Collapse
Affiliation(s)
- Jiale Hong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Fang Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xingyue Du
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Fa Xian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
7
|
Zhao Z, Du Y, Yan K, Zhang L, Guo Q. Exercise and osteoimmunology in bone remodeling. FASEB J 2024; 38:e23554. [PMID: 38588175 DOI: 10.1096/fj.202301508rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 04/10/2024]
Abstract
Bones can form the scaffolding of the body, support the organism, coordinate somatic movements, and control mineral homeostasis and hematopoiesis. The immune system plays immune supervisory, defensive, and regulatory roles in the organism, which mainly consists of immune organs (spleen, bone marrow, tonsils, lymph nodes, etc.), immune cells (granulocytes, platelets, lymphocytes, etc.), and immune molecules (immune factors, interferons, interleukins, tumor necrosis factors, etc.). Bone and the immune system have long been considered two distinct fields of study, and the bone marrow, as a shared microenvironment between the bone and the immune system, closely links the two. Osteoimmunology organically combines bone and the immune system, elucidates the role of the immune system in bone, and creatively emphasizes its interdisciplinary characteristics and the function of immune cells and factors in maintaining bone homeostasis, providing new perspectives for skeletal-related field research. In recent years, bone immunology has gradually become a hot spot in the study of bone-related diseases. As a new branch of immunology, bone immunology emphasizes that the immune system can directly or indirectly affect bones through the RANKL/RANK/OPG signaling pathway, IL family, TNF-α, TGF-β, and IFN-γ. These effects are of great significance for understanding inflammatory bone loss caused by various autoimmune or infectious diseases. In addition, as an external environment that plays an important role in immunity and bone, this study pays attention to the role of exercise-mediated bone immunity in bone reconstruction.
Collapse
Affiliation(s)
- Zhonghan Zhao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yuxiang Du
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kai Yan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiang Guo
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Ma L, Wang J, Yang Y, Lu J, Ling J, Chu X, Zhang Z, Tao Y, Li X, Tian Y, Li Z, Zhang Y, Sang X, Lu L, Wan X, Zhang K, Chen Y, Yu J, Zhuo R, Wu S, Pan J, Zhou X, Hu Y, Hu S. BRD4 PROTAC degrader MZ1 exhibits anti-B-cell acute lymphoblastic leukemia effects via targeting CCND3. Hematology 2023; 28:2247253. [PMID: 37594294 DOI: 10.1080/16078454.2023.2247253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
INTRODUCTION B-cell acute lymphoblastic leukemia (B-ALL) is the most prevalent malignant tumor affecting children. While the majority of B-ALL patients (90%) experience successful recovery, early relapse cases of B-ALL continue to exhibit high mortality rates. MZ1, a novel inhibitor of Bromodomains and extra-terminal (BET) proteins, has demonstrated potent antitumor activity against hematological malignancies. The objective of this study was to examine the role and therapeutic potential of MZ1 in the treatment of B-ALL. METHODS In order to ascertain the fundamental mechanism of MZ1, a sequence of in vitro assays was conducted on B-ALL cell lines, encompassing Cell Counting Kit 8 (CCK8) assay, Propidium iodide (PI) staining, and Annexin V/PI staining. Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) were employed to examine protein and mRNA expression levels. Transcriptomic RNA sequencing (RNA-seq) was utilized to screen the target genes of MZ1, and lentiviral transfection was employed to establish stably-expressing/knockdown cell lines. RESULTS MZ1 has been observed to induce the degradation of Bromodomain Containing 4 (BRD4), Bromodomain Containing 3 (BRD3), and Bromodomain Containing 2 (BRD2) in B-ALL cell strains, leading to inhibited cell growth and induction of cell apoptosis and cycle arrest in vitro. These findings suggest that MZ1 exhibits cytotoxic effects on two distinct molecular subtypes of B-ALL, namely 697 (TCF3/PBX1) and RS4;11 (MLL-AF4) B-ALL cell lines. Additionally, RNA-sequencing analysis revealed that MZ1 significantly downregulated the expression of Cyclin D3 (CCND3) gene in B-ALL cell lines, which in turn promoted cell apoptosis, blocked cell cycle, and caused cell proliferation inhibition. CONCLUSION Our results suggest that MZ1 has potential anti-B-ALL effects and might be a novel therapeutic target.
Collapse
Affiliation(s)
- Li Ma
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
- Department of Pediatrics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, People's Republic of China
| | - Jianwei Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yang Yang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Lu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jing Ling
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xinran Chu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yanfang Tao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yuanyuan Tian
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongping Zhang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xu Sang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Lihui Lu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiaomei Wan
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Kunlong Zhang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yanling Chen
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Juanjuan Yu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ran Zhuo
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Shuiyan Wu
- Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiuxia Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yixin Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
9
|
Garcia-Gimenez A, Richardson SE. The role of microenvironment in the initiation and evolution of B-cell precursor acute lymphoblastic leukemia. Front Oncol 2023; 13:1150612. [PMID: 36959797 PMCID: PMC10029760 DOI: 10.3389/fonc.2023.1150612] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/21/2023] [Indexed: 03/09/2023] Open
Abstract
B cell precursor acute lymphoblastic leukemia (BCP-ALL) is a malignant disorder of immature B lineage immune progenitors and is the commonest cancer in children. Despite treatment advances it remains a leading cause of death in childhood and response rates in adults remain poor. A preleukemic state predisposing children to BCP-ALL frequently arises in utero, with an incidence far higher than that of transformed leukemia, offering the potential for early intervention to prevent disease. Understanding the natural history of this disease requires an appreciation of how cell-extrinsic pressures, including microenvironment, immune surveillance and chemotherapy direct cell-intrinsic genetic and epigenetic evolution. In this review, we outline how microenvironmental factors interact with BCP-ALL at different stages of tumorigenesis and highlight emerging therapeutic avenues.
Collapse
Affiliation(s)
- Alicia Garcia-Gimenez
- Department of Haematology, Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Simon E. Richardson
- Department of Haematology, Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals, Cambridge, United Kingdom
- *Correspondence: Simon E. Richardson,
| |
Collapse
|
10
|
Chao P, Zhang X, Zhang L, Cui X, Wang S, Yang Y. Causal effects for genetic variants of osteoprotegerin on the risk of acute myocardial infarction and coronary heart disease: A two-sample Mendelian randomization study. Front Cardiovasc Med 2023; 10:1041231. [PMID: 36960470 PMCID: PMC10028206 DOI: 10.3389/fcvm.2023.1041231] [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: 09/10/2022] [Accepted: 01/26/2023] [Indexed: 03/09/2023] Open
Abstract
Although since the 1980s, the mortality of coronary heart disease(CHD) has obviously decreased due to the rise of coronary intervention, the mortality and disability of CHD were still high in some countries. Etiological studies of acute myocardial infarction(AMI) and CHD were extremely important. In this study, we used two-sample Mendelian randomization(TSMR) method to collect GWAS statistics of osteoprotegerin (OPG), AMI and CHD to reveal the causal relationship between OPG and these two diseases. In total, we identified 7 genetic variants associated with AMI and 7 genetic variants associated with CHD that were not found to be in linkage disequilibrium(LD; r 2 < 0.001). Evidence of a positive effect of an OPG genetic susceptibility on AMI was discovered(IVW OR = 0.877; 95% CI = 0.787-0.977; p = 0.017; 7 SNPs) and CHD (IVW OR = 0.892; 95% CI = 0.803-0.991; p = 0.033; 7 SNPs). After removing the influence of rs1385492, we found that there was a correlation between OPG and AMI/CHD (AMI: weighted median OR = 0.818;95% CI = 0.724-0.950; p = 0.001; 6SNPs;CHD: weighted median OR = 0.842; 95% CI = 0.755-0.938; p = 1.893 × 10-3; 6SNPs). The findings of our study indicated that OPG had a tight genetic causation association with MI or CHD. This genetic causal relationship presented us with fresh ideas for the etiology of AMI and CHD, which is an area of research that will continue in the future.
Collapse
Affiliation(s)
- Peng Chao
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Xueqin Zhang
- Department of Nephropathy, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Lei Zhang
- Department of Endocrine, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Xinyue Cui
- Department of Nephropathy, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Shanshan Wang
- Department of Nephropathy, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Yining Yang
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
- *Correspondence: Yining Yang,
| |
Collapse
|
11
|
Safety and Efficacy of Alendronate to Treat Osteopenia in Children During Therapy for Acute Lymphoblastic Leukemia: A Retrospective Cohort Study of Sequential Outcomes. J Pediatr Hematol Oncol 2022; 45:200-206. [PMID: 36729669 DOI: 10.1097/mph.0000000000002606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 11/07/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Low bone mineral density is encountered in children with acute lymphoblastic leukemia (ALL) before, during, and after treatment. Prior experience with alendronate, an oral bisphosphonate, demonstrated high tolerability and evident clinical efficacy. However, concerns have been expressed about the long-term safety and utility of such agents in children. PROCEDURE Sixty-nine children with ALL received alendronate for a mean of 87 weeks after dual-energy x-ray absorptiometry. Dual-energy x-ray absorptiometry was repeated following the completion of alendronate, and 5 to 9 years later in a subgroup of 32 children. Lumbar spine areal bone mineral density (LS aBMD) Z scores were obtained. RESULTS The mean LS aBMD Z score rose from -1.78 to-0.47 (P <0.0001). There was a modest median loss of LS aBMD subsequently in the 32 subjects on long-term follow-up. Almost 80% (N=172) of the children remain in continuous complete remission at a mean of 14.5 years from diagnosis. Of those who received alendronate, which was almost uniformly well tolerated, 7/69 (10.3%) relapsed compared with 19/89 (21.3%) who did not receive the drug. DISCUSSION Alendronate appears to be well tolerated and moderately effective in osteopenic children with ALL. Whether it offers protection against relapse of leukemia needs further study.
Collapse
|
12
|
Haselager MV, Eldering E. The Therapeutic Potential of Targeting NIK in B Cell Malignancies. Front Immunol 2022; 13:930986. [PMID: 35911754 PMCID: PMC9326486 DOI: 10.3389/fimmu.2022.930986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/08/2022] [Indexed: 11/24/2022] Open
Abstract
NF-κB-inducing kinase (NIK) is a key player in non-canonical NF-κB signaling, involved in several fundamental cellular processes, and is crucial for B cell function and development. In response to certain signals and ligands, such as CD40, BAFF and lymphotoxin-β activation, NIK protein stabilization and subsequent NF-κB activation is achieved. Overexpression or overactivation of NIK is associated with several malignancies, including activating mutations in multiple myeloma (MM) and gain-of-function in MALT lymphoma as a result of post-translational modifications. Consequently, drug discovery studies are devoted to pharmacologic modulation of NIK and development of specific novel small molecule inhibitors. However, disease-specific in vitro and in vivo studies investigating NIK inhibition are as of yet lacking, and clinical trials with NIK inhibitors remain to be initiated. In order to bridge the gap between bench and bedside, this review first briefly summarizes our current knowledge on NIK activation, functional activity and stability. Secondly, we compare current inhibitors targeting NIK based on efficacy and specificity, and provide a future perspective on the therapeutic potential of NIK inhibition in B cell malignancies.
Collapse
Affiliation(s)
- Marco V. Haselager
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, Netherlands
- *Correspondence: Eric Eldering,
| |
Collapse
|
13
|
The roles of osteoprotegerin in cancer, far beyond a bone player. Cell Death Dis 2022; 8:252. [PMID: 35523775 PMCID: PMC9076607 DOI: 10.1038/s41420-022-01042-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 11/08/2022]
Abstract
Osteoprotegerin (OPG), also known as tumor necrosis factor receptor superfamily member 11B (TNFRSF11B), is a member of the tumor necrosis factor (TNF) receptor superfamily. Characterized by its ability to bind to receptor activator of nuclear factor kappa B ligand (RANKL), OPG is critically involved in bone remodeling. Emerging evidence implies that OPG is far beyond a bone-specific modulator, and is involved in multiple physiological and pathological processes, such as immunoregulation, vascular function, and fibrosis. Notably, numerous preclinical and clinical studies have been conducted to assess the participation of OPG in tumorigenesis and cancer development. Mechanistic studies have demonstrated that OPG is involved in multiple hallmarks of cancer, including tumor survival, epithelial to mesenchymal transition (EMT), neo-angiogenesis, invasion, and metastasis. In this review, we systematically summarize the basis and advances of OPG from its molecular structure to translational applications. In addition to its role in bone homeostasis, the physiological and pathological impacts of OPG on human health and its function in cancer progression are reviewed, providing a comprehensive understanding of OPG. We aim to draw more attention to OPG in the field of cancer, and to propose it as a promising diagnostic or prognostic biomarker as well as potential therapeutic target for cancer.
Collapse
|
14
|
Ruiz-Aparicio PF, Vernot JP. Bone Marrow Aging and the Leukaemia-Induced Senescence of Mesenchymal Stem/Stromal Cells: Exploring Similarities. J Pers Med 2022; 12:jpm12050716. [PMID: 35629139 PMCID: PMC9147878 DOI: 10.3390/jpm12050716] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Bone marrow aging is associated with multiple cellular dysfunctions, including perturbed haematopoiesis, the propensity to haematological transformation, and the maintenance of leukaemia. It has been shown that instructive signals from different leukemic cells are delivered to stromal cells to remodel the bone marrow into a supportive leukemic niche. In particular, cellular senescence, a physiological program with both beneficial and deleterious effects on the health of the organisms, may be responsible for the increased incidence of haematological malignancies in the elderly and for the survival of diverse leukemic cells. Here, we will review the connection between BM aging and cellular senescence and the role that these processes play in leukaemia progression. Specifically, we discuss the role of mesenchymal stem cells as a central component of the supportive niche. Due to the specificity of the genetic defects present in leukaemia, one would think that bone marrow alterations would also have particular changes, making it difficult to envisage a shared therapeutic use. We have tried to summarize the coincident features present in BM stromal cells during aging and senescence and in two different leukaemias, acute myeloid leukaemia, with high frequency in the elderly, and B-acute lymphoblastic leukaemia, mainly a childhood disease. We propose that mesenchymal stem cells are similarly affected in these different leukaemias, and that the changes that we observed in terms of cellular function, redox balance, genetics and epigenetics, soluble factor repertoire and stemness are equivalent to those occurring during BM aging and cellular senescence. These coincident features may be used to explore strategies useful to treat various haematological malignancies.
Collapse
Affiliation(s)
- Paola Fernanda Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
15
|
Hughes AM, Kuek V, Kotecha RS, Cheung LC. The Bone Marrow Microenvironment in B-Cell Development and Malignancy. Cancers (Basel) 2022; 14:2089. [PMID: 35565219 PMCID: PMC9102980 DOI: 10.3390/cancers14092089] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022] Open
Abstract
B lymphopoiesis is characterized by progressive loss of multipotent potential in hematopoietic stem cells, followed by commitment to differentiate into B cells, which mediate the humoral response of the adaptive immune system. This process is tightly regulated by spatially distinct bone marrow niches where cells, including mesenchymal stem and progenitor cells, endothelial cells, osteoblasts, osteoclasts, and adipocytes, interact with B-cell progenitors to direct their proliferation and differentiation. Recently, the B-cell niche has been implicated in initiating and facilitating B-cell precursor acute lymphoblastic leukemia. Leukemic cells are also capable of remodeling the B-cell niche to promote their growth and survival and evade treatment. Here, we discuss the major cellular components of bone marrow niches for B lymphopoiesis and the role of the malignant B-cell niche in disease development, treatment resistance and relapse. Further understanding of the crosstalk between leukemic cells and bone marrow niche cells will enable development of additional therapeutic strategies that target the niches in order to hinder leukemia progression.
Collapse
Affiliation(s)
- Anastasia M. Hughes
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia; (A.M.H.); (V.K.); (R.S.K.)
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Vincent Kuek
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia; (A.M.H.); (V.K.); (R.S.K.)
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Rishi S. Kotecha
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia; (A.M.H.); (V.K.); (R.S.K.)
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
- School of Medicine, University of Western Australia, Perth, WA 6009, Australia
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children’s Hospital, Perth, WA 6009, Australia
| | - Laurence C. Cheung
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia; (A.M.H.); (V.K.); (R.S.K.)
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
16
|
Bone Health in Osteosarcoma at Presentation and Its Impact on Cancer Treatment: A Case Series of 3 Pediatric Patients. J Pediatr Hematol Oncol 2022; 44:e782-e787. [PMID: 35091517 DOI: 10.1097/mph.0000000000002400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/12/2021] [Indexed: 11/26/2022]
Abstract
Osteosarcoma is the most common pediatric malignant bone tumor. Concomitant osteoporosis has typically been attributed to oncologic therapy. The present case series is aimed to describe 3 patients who presented with osteoporosis or osteopenia before, or early in, their oncology treatment. In our patients, bone health and its complications had significant impacts including pain, reduced mobility, prolonged admission, and delays in recovery. Our patients experienced improvement with resection of their primary tumor and with bisphosphonate infusion. Future studies are required to determine the prevalence osteoporosis at presentation of osteosarcoma and the role of bisphosphonates.
Collapse
|
17
|
Rajakumar SA, Grandal I, Minden MD, Hitzler JK, Guidos CJ, Danska JS. Targeted blockade of immune mechanisms inhibit B precursor acute lymphoblastic leukemia cell invasion of the central nervous system. Cell Rep Med 2021; 2:100470. [PMID: 35028611 PMCID: PMC8714910 DOI: 10.1016/j.xcrm.2021.100470] [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: 04/29/2021] [Revised: 10/05/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022]
Abstract
Acute lymphoblastic leukemia (ALL) dissemination to the central nervous system (CNS) is a challenging clinical problem whose underlying mechanisms are poorly understood. Here, we show that primary human ALL samples injected into the femora of immunodeficient mice migrate to the skull and vertebral bone marrow and provoke bone lesions that enable passage into the subarachnoid space. Treatment of leukemia xenografted mice with a biologic antagonist of receptor activator of nuclear factor κB ligand (RANKL) blocks this entry route. In addition to erosion of cranial and vertebral bone, samples from individuals with B-ALL also penetrate the blood-cerebrospinal fluid barrier of recipient mice. Co-administration of C-X-C chemokine receptor 4 (CXCR4) and RANKL antagonists attenuate both identified routes of entry. Our findings suggest that targeted RANKL and CXCR4 pathway inhibitors could attenuate routes of leukemia blast CNS invasion and provide benefit for B-ALL-affected individuals.
Collapse
Affiliation(s)
- Sujeetha A. Rajakumar
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ildiko Grandal
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Mark D. Minden
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Johann K. Hitzler
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Pediatrics, Division of Hematology and Oncology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Cynthia J. Guidos
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jayne S. Danska
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
18
|
Clar KL, Weber LM, Schmied BJ, Heitmann JS, Marconato M, Tandler C, Schneider P, Salih HR. Receptor Activator of NF-κB (RANK) Confers Resistance to Chemotherapy in AML and Associates with Dismal Disease Course. Cancers (Basel) 2021; 13:cancers13236122. [PMID: 34885231 PMCID: PMC8657109 DOI: 10.3390/cancers13236122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/19/2022] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults. Despite the emergence of new therapeutic agents in recent years, curation remains challenging, and new and better treatment options are needed. In the present study, we investigated the expression, prognostic significance, and functional role of the Receptor Activator of Nuclear Factor-κB (RANK) in AML. We found that RANK is expressed on leukemic cells in a substantial proportion of AML patients and is associated with a dismal disease course. We further demonstrated that signaling via RANK induces release of factors that favor AML cell survival and confers resistance to chemotherapeutics in AML treatment. Together, our findings identify RANK as novel prognostic marker and putative candidate for therapeutic intervention in AML to enhance response to treatment. Abstract Although treatment options of acute myeloid leukemia (AML) have improved over the recent years, prognosis remains poor. Better understanding of the molecular mechanisms influencing and predicting treatment efficacy may improve disease control and outcome. Here we studied the expression, prognostic relevance and functional role of the tumor necrosis factor receptor (TNFR) family member Receptor Activator of Nuclear Factor (NF)-κB (RANK) in AML. We conducted an experimental ex vivo study using leukemic cells of 54 AML patients. Substantial surface expression of RANK was detected on primary AML cells in 35% of the analyzed patients. We further found that RANK signaling induced the release of cytokines acting as growth and survival factors for the leukemic cells and mediated resistance of AML cells to treatment with doxorubicin and cytarabine, the most commonly used cytostatic compounds in AML treatment. In line, RANK expression correlated with a dismal disease course as revealed by reduced overall survival. Together, our results show that RANK plays a yet unrecognized role in AML pathophysiology and resistance to treatment, and identify RANK as “functional” prognostic marker in AML. Therapeutic modulation of RANK holds promise to improve treatment response in AML patients.
Collapse
Affiliation(s)
- Kim L. Clar
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy (iFIT)”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Lisa M. Weber
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy (iFIT)”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Bastian J. Schmied
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy (iFIT)”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy (iFIT)”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Maddalena Marconato
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
| | - Claudia Tandler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy (iFIT)”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland;
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (K.L.C.); (L.M.W.); (B.J.S.); (J.S.H.); (M.M.); (C.T.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy (iFIT)”, University of Tuebingen, 72076 Tuebingen, Germany
- Correspondence: ; Tel.: +49-7071-29-83275
| |
Collapse
|
19
|
Li N, Fu L, Li Z, Ke Y, Wang Y, Wu J, Yu J. The Role of Immune Microenvironment in Maxillofacial Bone Homeostasis. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.780973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Maxillofacial bone defects are common medical problems caused by congenital defects, necrosis, trauma, tumor, inflammation, and fractures non-union. Maxillofacial bone defects often need bone graft, which has many difficulties, such as limited autogenous bone supply and donor site morbidity. Bone tissue engineering is a promising strategy to overcome the above-mentioned problems. Osteoimmunology is the inter-discipline that focuses on the relationship between the skeletal and immune systems. The immune microenvironment plays a crucial role in bone healing, tissue repair and regeneration in maxillofacial region. Recent studies have revealed the vital role of immune microenvironment and bone homeostasis. In this study, we analyzed the complex interaction between immune microenvironment and bone regeneration process in oral and maxillofacial region, which will be important to improve the clinical outcome of the bone injury treatment.
Collapse
|
20
|
Li K, Jin R, Wu X. The role of macrophages and osteoclasts in the progression of leukemia. ACTA ACUST UNITED AC 2021; 26:724-733. [PMID: 34555294 DOI: 10.1080/16078454.2021.1976911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
ABSTRACTBone marrow microenvironment provides critical regulatory signals for lineage differentiation and maintenance of HSC quiescence, and these signals also contribute to hematological myeloid malignancies. Macrophages exhibit high phenotypic heterogeneity under both physiological and pathological conditions and are mainly divided into proinflammatory M1 and anti-inflammatory M2 macrophages. Furthermore, osteoclasts are multinucleated giant cells that arise by fusion of monocyte/macrophage-like cells, which are commonly known as bone macrophages. Emerging evidence suggests that macrophages and osteoclasts originating from myeloid progenitors lead to two competing differentiation outcomes, and they appear to play an important role in the onset, progression, and bone metastasis of solid cancers. However, little is known about their role in the development of hematological malignancies. In this review, we focus on macrophages and osteoclasts, their role in leukemia, and the potential for targeting these cells in this disease.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
21
|
Dander E, Palmi C, D’Amico G, Cazzaniga G. The Bone Marrow Niche in B-Cell Acute Lymphoblastic Leukemia: The Role of Microenvironment from Pre-Leukemia to Overt Leukemia. Int J Mol Sci 2021; 22:ijms22094426. [PMID: 33922612 PMCID: PMC8122951 DOI: 10.3390/ijms22094426] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic lesions predisposing to pediatric B-cell acute lymphoblastic leukemia (B-ALL) arise in utero, generating a clinically silent pre-leukemic phase. We here reviewed the role of the surrounding bone marrow (BM) microenvironment in the persistence and transformation of pre-leukemic clones into fully leukemic cells. In this context, inflammation has been highlighted as a crucial microenvironmental stimulus able to promote genetic instability, leading to the disease manifestation. Moreover, we focused on the cross-talk between the bulk of leukemic cells with the surrounding microenvironment, which creates a “corrupted” BM malignant niche, unfavorable for healthy hematopoietic precursors. In detail, several cell subsets, including stromal, endothelial cells, osteoblasts and immune cells, composing the peculiar leukemic niche, can actively interact with B-ALL blasts. Through deregulated molecular pathways they are able to influence leukemia development, survival, chemoresistance, migratory and invasive properties. The concept that the pre-leukemic and leukemic cell survival and evolution are strictly dependent both on genetic lesions and on the external signals coming from the microenvironment paves the way to a new idea of dual targeting therapeutic strategy.
Collapse
Affiliation(s)
- Erica Dander
- Correspondence: (E.D.); (C.P.); Tel.: +39-(0)-39-2332229 (E.D. & C.P.); Fax: +39-(0)39-2332167 (E.D. & C.P.)
| | - Chiara Palmi
- Correspondence: (E.D.); (C.P.); Tel.: +39-(0)-39-2332229 (E.D. & C.P.); Fax: +39-(0)39-2332167 (E.D. & C.P.)
| | | | | |
Collapse
|
22
|
Abstract
Skeletal morbidities continue to cause acute and long-term burdens for B-ALL patients underscoring the need to identify the mechanisms underlying these processes and to develop effective therapies. Our recent findings demonstrated that B-ALL cells isolated at patient diagnosis can cause bone destruction and have identified the receptor activator of nuclear factor κ-B (RANK-RANKL) ligand axis as a critical effector of these effects.
Collapse
Affiliation(s)
- Sujeetha A Rajakumar
- Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Canada.,Department of Medical Biophysics, Faculty of Medicine University of Toronto, Toronto, Canada
| | - Jayne S Danska
- Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Canada.,Department of Medical Biophysics, Faculty of Medicine University of Toronto, Toronto, Canada.,Department of Immunology, Faculty of Medicine University of Toronto, Toronto, Canada
| |
Collapse
|