1
|
Weldon KC, Longaker MT, Ambrosi TH. Harnessing the diversity and potential of endogenous skeletal stem cells for musculoskeletal tissue regeneration. Stem Cells 2025; 43:sxaf006. [PMID: 39945760 PMCID: PMC11892563 DOI: 10.1093/stmcls/sxaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/21/2025] [Indexed: 03/11/2025]
Abstract
In our aging society, the degeneration of the musculoskeletal system and adjacent tissues is a growing orthopedic concern. As bones age, they become more fragile, increasing the risk of fractures and injuries. Furthermore, tissues like cartilage accumulate damage, leading to widespread joint issues. Compounding this, the regenerative capacity of these tissues declines with age, exacerbating the consequences of fractures and cartilage deterioration. With rising demand for fracture and cartilage repair, bone-derived stem cells have attracted significant research interest. However, the therapeutic use of stem cells has produced inconsistent results, largely due to ongoing debates and uncertainties regarding the precise identity of the stem cells responsible for musculoskeletal growth, maintenance and repair. This review focuses on the potential to leverage endogenous skeletal stem cells (SSCs)-a well-defined population of stem cells with specific markers, reliable isolation techniques, and functional properties-in bone repair and cartilage regeneration. Understanding SSC behavior in response to injury, including their activation to a functional state, could provide insights into improving treatment outcomes. Techniques like microfracture surgery, which aim to stimulate SSC activity for cartilage repair, are of particular interest. Here, we explore the latest advances in how such interventions may modulate SSC function to enhance bone healing and cartilage regeneration.
Collapse
Affiliation(s)
- Kelly C Weldon
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
- School of Medicine, University of California, Sacramento, CA 95817, United States
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Thomas H Ambrosi
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| |
Collapse
|
2
|
Cáceres TM, Patiño LH, Ramírez JD. Understanding Host-Pathogen Interactions in Congenital Chagas Disease Through Transcriptomic Approaches. Pathogens 2025; 14:106. [PMID: 40005483 PMCID: PMC11858232 DOI: 10.3390/pathogens14020106] [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: 12/16/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, is a parasitic zoonosis with significant health impacts, particularly in Latin America. While traditionally associated with vector-borne transmission, increased migration has expanded its reach into urban and non-endemic regions. Congenital transmission has become a critical route of infection, involving intricate maternal-fetal immune interactions that challenge diagnosis and treatment. This review synthesizes findings from three RNA-seq studies that explore the molecular underpinnings of congenital Chagas disease, emphasizing differentially expressed genes (DEGs) implicated in host-pathogen interactions. The DAVID tool analysis highlighted the overexpression of genes associated with the innate immune response, including pro-inflammatory cytokines that drive chemotaxis and neutrophil activation. Additionally, calcium-dependent pathways critical for parasite invasion were modulated. T. cruzi exploits the maternal-fetal immune axis to establish a tolerogenic environment conducive to congenital transmission. Alterations in placental angiogenesis, cellular regeneration, and metabolic processes further demonstrate the parasite's ability to manipulate host responses for its survival and persistence. These findings underscore the complex interplay between the host and pathogen that facilitates disease progression. Future research integrating transcriptomic, proteomic, and metabolomic approaches is essential to unravel the molecular mechanisms underlying congenital Chagas disease, with a particular focus on the contributions of genetic diversity and non-coding RNAs in immune evasion and disease pathogenesis.
Collapse
Affiliation(s)
- Tatiana M. Cáceres
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (T.M.C.); (L.H.P.)
| | - Luz Helena Patiño
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (T.M.C.); (L.H.P.)
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (T.M.C.); (L.H.P.)
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
3
|
Chang YH, Wu KC, Wang KH, Ding DC. Role of Leucine-Rich Repeat-Containing G-Protein-Coupled Receptors 4-6 (LGR4-6) in the Ovary and Other Female Reproductive Organs: A Literature Review. Cell Transplant 2025; 34:9636897241303441. [PMID: 39874091 PMCID: PMC11776010 DOI: 10.1177/09636897241303441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 01/30/2025] Open
Abstract
Leucine-rich repeat-containing G-protein-coupled receptors regulate stem cell activity and tissue homeostasis within female reproductive organs, primarily through their interaction with the Wnt/β-catenin signaling pathway. LGR4-6 are increasingly recognized for their roles in organ development, regeneration, and cancer. This review aims to provide a comprehensive overview of the roles of LGR4-6 in female reproductive organs, highlighting their significance in normal physiology and disease states, specifically in the context of ovarian cancer. LGR4 is essential for the proper development of the female reproductive system; its deficiency leads to significant reproductive abnormalities, including delayed menarche and follicle development issues. LGR5 is a well-established marker of stem cells in the ovary and fallopian tubes. It has been implicated in the pathogenesis of high-grade serous ovarian cancer. LGR6, while less studied, shares functional similarities with LGR5 and can maintain stemness. It contributes to chemoresistance in ovarian cancer. LGR6 is a marker for fallopian tube stem cells and is involved in stem cell maintenance and differentiation. LGR4-6 regulate the pathophysiology of female reproductive tissues. LGR4-6 are promising therapeutic targets for treating reproductive cancers and other related disorders. Molecular mechanisms underlying the functions of LGR4-6 should be studied.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
| | - Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
| | - Kai-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
- Institute of Medical Sciences, Tzu Chi University, Hualien
| |
Collapse
|
4
|
King JS, Wan M, Kim A, Novak S, Prabhu S, Kalajzic I, Delany AM, Sanjay A. Effects of Aging on the Immune and Periosteal Response to Fracture in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622348. [PMID: 39574733 PMCID: PMC11580938 DOI: 10.1101/2024.11.06.622348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Aging predisposes individuals to reduced bone mass and fragility fractures, which are costly and linked to high mortality. Understanding how aging affects fracture healing is essential for developing therapies to enhance bone regeneration in older adults. During the inflammatory phase of fracture healing, immune cells are recruited to the injury site as periosteal skeletal stem/progenitor cells (pSSPCs) rapidly proliferate and differentiate into osteochondral lineages, allowing for fibrocartilaginous callus formation and complete bone healing. Irrespective of age, how periosteal mesenchymal and immune cells interact during early fracture healing is incompletely understood, limiting our ability to potentially modulate these processes. To address this, we directly analyzed, in parallel, at a single-cell level, isolated murine CD45(+) and CD45(-) periosteal cells dissected from intact and fractured bones, collected three days after injury. Through comprehensive analysis, corroborated by bulk RNA-sequencing, flow cytometry, and histology, we found aging decreases pSSPCs proliferative, marked by a reduced expression of genes required for callus formation and an increased senescence signature. We found that the chemokine Cxcl9 was highly upregulated in aged intact Prrx1+ pSSPCs, predicted to interact with other pSSPCs directly, and associated with increased recruitment of CD8+ T cells at the fracture site three days after injury. Cell-to-cell communication analysis provided insight into the complexity of interactions among the many cell types regulating fracture healing and the impact of aging on these processes. Together, these results provide insight into age-induced alterations in fracture healing, informing the development of improved therapeutic approaches for fragility fractures.
Collapse
|
5
|
Kobelski MM, Ramchand SK, Tsai JN, Leder BZ, Demay MB. Evaluation of Osteogenic Phenotype in Postmenopausal Women Receiving Anabolic and Antiresorptive Osteoporosis Therapies. Endocrinology 2024; 165:bqae142. [PMID: 39445778 PMCID: PMC11523613 DOI: 10.1210/endocr/bqae142] [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: 07/11/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
Aging of the general population has led to a substantial increase in the prevalence of osteoporosis over the past decades. While there are effective pharmacological agents that increase bone formation, decrease bone resorption, and decrease fracture risk, they do not uniformly cure osteoporosis. This has prompted investigations to examine whether combination therapy (COMBO) with these agents can result in an additive benefit. Since concomitant therapy with denosumab and teriparatide has shown promise in this respect, investigations were undertaken to explore whether the changes in osteogenic phenotype could provide insight into the cellular and molecular mechanism of this effect. Investigations were performed in postmenopausal women receiving denosumab, teriparatide, or both for 3 months. Histomorphometric parameters were the primary outcome, while exploratory studies examined RNA expression in bone biopsies as well as in sorted and cultured bone marrow stromal cells (BMSCs). Osteogenic colony forming units of BMSCs were also evaluated. The studies demonstrated that COMBO results in an increase in osteoprogenitors, evidenced by an increase in osteoblastic colony-forming units. This was associated with an increased in BMSC expression of LGR6 (leucine-rich repeat containing G protein-coupled receptor 6), a stem cell marker and activator of the canonical Wnt signaling pathway. These data suggest that enhancement of canonical Wnt signaling contributes to the increase in osteoprogenitors and consequently an increase in bone density in postmenopausal women receiving COMBO for osteoporosis.
Collapse
Affiliation(s)
| | - Sabashini K Ramchand
- Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Joy N Tsai
- Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin Z Leder
- Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Cotta GC, Teixeira dos Santos RC, Costa GMJ, Lacerda SMDSN. Reporter Alleles in hiPSCs: Visual Cues on Development and Disease. Int J Mol Sci 2024; 25:11009. [PMID: 39456792 PMCID: PMC11507014 DOI: 10.3390/ijms252011009] [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: 08/30/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Reporter alleles are essential for advancing research with human induced pluripotent stem cells (hiPSCs), notably in developmental biology and disease modeling. This study investigates the state-of-the-art gene-editing techniques tailored for generating reporter alleles in hiPSCs, emphasizing their effectiveness in investigating cellular dynamics and disease mechanisms. Various methodologies, including the application of CRISPR/Cas9 technology, are discussed for accurately integrating reporter genes into the specific genomic loci. The synthesis of findings from the studies utilizing these reporter alleles reveals insights into developmental processes, genetic disorder modeling, and therapeutic screening, consolidating the existing knowledge. These hiPSC-derived models demonstrate remarkable versatility in replicating human diseases and evaluating drug efficacy, thereby accelerating translational research. Furthermore, this review addresses challenges and future directions in refining the reporter allele design and application to bolster their reliability and relevance in biomedical research. Overall, this investigation offers a comprehensive perspective on the methodologies, applications, and implications of reporter alleles in hiPSC-based studies, underscoring their essential role in advancing both fundamental scientific understanding and clinical practice.
Collapse
Affiliation(s)
| | | | | | - Samyra Maria dos Santos Nassif Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil; (G.C.C.); (R.C.T.d.S.); (G.M.J.C.)
| |
Collapse
|
7
|
King JS, Wan M, Wagley Y, Stestiv M, Kalajzic I, Hankenson KD, Sanjay A. Signaling pathways associated with Lgr6 to regulate osteogenesis. Bone 2024; 187:117207. [PMID: 39033993 DOI: 10.1016/j.bone.2024.117207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Fracture management largely relies on the bone's inherent healing capabilities and, when necessary, surgical intervention. Currently, there are limited osteoinductive therapies to promote healing, making targeting skeletal stem/progenitor cells (SSPCs) a promising avenue for therapeutic development. A limiting factor for this approach is our incomplete understanding of the molecular mechanisms governing SSPCs' behavior. We have recently identified that the Leucine-rich repeat-containing G-protein coupled receptor 6 (Lgr6) is expressed in sub-populations of SSPCs, and is required for maintaining bone volume during adulthood and for proper fracture healing. Lgr family members (Lgr4-6) are markers of stem cell niches and play a role in tissue regeneration primarily by binding R-Spondin (Rspo1-4). This interaction promotes canonical Wnt (cWnt) signaling by stabilizing Frizzled receptors. Interestingly, our findings here indicate that Lgr6 may also influence cWnt-independent pathways. Remarkably, Lgr6 expression was enhanced during Bmp-mediated osteogenesis of both human and murine cells. Using biochemical approaches, RNA sequencing, and bioinformatic analysis of published single-cell data, we found that elements of BMP signaling, including its target gene, pSMAD, and gene ontology pathways, are downregulated in the absence of Lgr6. Our findings uncover a molecular interdependency between the Bmp pathway and Lgr6, offering new insights into osteogenesis and potential targets for enhancing fracture healing.
Collapse
Affiliation(s)
- Justin S King
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Marta Stestiv
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA.
| |
Collapse
|
8
|
Huang M, Chen B, Chen X, Liu T, Liang S, Hu H, Bai X, Gong Y. RanGAP1 maintains chromosome stability in limb bud mesenchymal cells during bone development. Cell Signal 2024; 120:111222. [PMID: 38729327 DOI: 10.1016/j.cellsig.2024.111222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Bone development involves the rapid proliferation and differentiation of osteogenic lineage cells, which makes accurate chromosomal segregation crucial for ensuring cell proliferation and maintaining chromosomal stability. However, the mechanism underlying the maintenance of chromosome stability during the rapid proliferation and differentiation of Prx1-expressing limb bud mesenchymal cells into osteoblastic precursor cells remains unexplored. METHODS A transgenic mouse model of RanGAP1 knockout of limb and head mesenchymal progenitor cells was constructed to explore the impact of RanGAP1 deletion on bone development by histomorphology and immunostaining. Subsequently, G-banding karyotyping analysis and immunofluorescence staining were used to examine the effects of RanGAP1 deficiency on chromosome instability. Finally, the effects of RanGAP1 deficiency on chromothripsis and bone development signaling pathways were elucidated by whole-genome sequencing, RNA-sequencing, and qPCR. RESULTS The ablation of RanGAP1 in limb and head mesenchymal progenitor cells expressing Prx1 in mice resulted in embryonic lethality, severe cartilage and bone dysplasia, and complete loss of cranial vault formation. Moreover, RanGAP1 loss inhibited chondrogenic or osteogenic differentiation of mesenchymal stem cells (MSCs). Most importantly, we found that RanGAP1 loss in limb bud mesenchymal cells triggered missegregation of chromosomes, resulting in chromothripsis of chromosomes 1q and 14q, further inhibiting the expression of key genes involved in multiple bone development signaling pathways such as WNT, Hedgehog, TGF-β/BMP, and PI3K/AKT in the chromothripsis regions, ultimately disrupting skeletal development. CONCLUSIONS Our results establish RanGAP1 as a critical regulator of bone development, as it supports this process by preserving chromosome stability in Prx1-expressing limb bud mesenchymal cells.
Collapse
Affiliation(s)
- Minjun Huang
- Department of Spine Surgery, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China; Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Bochong Chen
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoli Chen
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tianxiao Liu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Siying Liang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongling Hu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan 528399, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Yan Gong
- Department of Spine Surgery, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
9
|
Zhong F, Hao W, Chao H, Li Z, Chen S, Li S, Zhang S, Zhang K, Li Z, Liu H. Tenascin-C promotes endochondral ossification and fracture healing through Hedgehog and Hippo signaling. Biochem Biophys Res Commun 2024; 703:149634. [PMID: 38354465 DOI: 10.1016/j.bbrc.2024.149634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/24/2023] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Fractures are frequent and severe musculoskeletal injuries. This study aimed to investigate the function of tenascin-C (TNC) in regulating chondrogenic during fracture healing and elucidate the underlying molecular mechanisms. A well-established femur fracture model in male C57BL/6J mice was used to transect the middle diaphysis of the femur. To identify the essential role of TNC, shTNC lentiviruses or TNC protein were administered in the animal model. Micro-CT analysis, histologic analysis, immunostaining assays, and gene expression analysis were employed to investigate the effect of TNC during fracture healing. An in vitro mesenchymal stem cell culture system was developed to investigate the role and molecular mechanism of TNC in regulating chondrogenesis. TNC expression was induced at the inflammatory phase and peaked at the cartilaginous callus phase during fracture healing. Knockdown of TNC expression in callus results in decreased callus formation and impaired fracture healing. Conversely, administration of exogenous TNC promoted chondrogenic differentiation, cartilage template formation and ultimately improved fracture healing. Both the Hedgehog and Hippo signaling pathways were found to be involved in the pro-chondrogenic function of TNC. Our observations demonstrate that TNC is a crucial factor responsible for endochondral ossification in fracture healing and provide a potential therapeutic strategy for promoting fracture healing.
Collapse
Affiliation(s)
- Fangling Zhong
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Wenjun Hao
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Hua Chao
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Zihao Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Siwen Chen
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Sifang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Shuai Zhang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Kuibo Zhang
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Zemin Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China.
| | - Hui Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
10
|
Aydin A, Klenk C, Nemec K, Işbilir A, Martin LM, Zauber H, Rrustemi T, Toka HR, Schuster H, Gong M, Stricker S, Bock A, Bähring S, Selbach M, Lohse MJ, Luft FC. ADAM19 cleaves the PTH receptor and associates with brachydactyly type E. Life Sci Alliance 2024; 7:e202302400. [PMID: 38331475 PMCID: PMC10853454 DOI: 10.26508/lsa.202302400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Brachydactyly type E (BDE), shortened metacarpals, metatarsals, cone-shaped epiphyses, and short stature commonly occurs as a sole phenotype. Parathyroid hormone-like protein (PTHrP) has been shown to be responsible in all forms to date, either directly or indirectly. We used linkage and then whole genome sequencing in a small pedigree, to elucidate BDE and identified a truncated disintegrin-and-metalloproteinase-19 (ADAM19) allele in all affected family members, but not in nonaffected persons. Since we had shown earlier that the extracellular domain of the parathyroid hormone receptor (PTHR1) is subject to an unidentified metalloproteinase cleavage, we tested the hypothesis that ADAM19 is a sheddase for PTHR1. WT ADAM19 cleaved PTHR1, while mutated ADAM-19 did not. We mapped the cleavage site that we verified with mass spectrometry between amino acids 64-65. ADAM-19 cleavage increased Gq and decreased Gs activation. Moreover, perturbed PTHR1 cleavage by ADAM19 increased ß-arrestin2 recruitment, while cAMP accumulation was not altered. We suggest that ADAM19 serves as a regulatory element for PTHR1 and could be responsible for BDE. This sheddase may affect other PTHrP or PTH-related functions.
Collapse
Affiliation(s)
- Atakan Aydin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Christoph Klenk
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Katarina Nemec
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Department of Structural Biology and Center of Excellence for Data-Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ali Işbilir
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Lisa M Martin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Henrik Zauber
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Trendelina Rrustemi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hakan R Toka
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Herbert Schuster
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Maolian Gong
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Andreas Bock
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sylvia Bähring
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Matthias Selbach
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Martin J Lohse
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- ISAR Bioscience Institute, Munich, Germany
| | - Friedrich C Luft
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
11
|
Xia D, Wu R, Xue Q, Jiang G, Xu S. Metabolomics provides insights into acceleration of bone healing in fractured patients with traumatic brain injuries. Biomed Chromatogr 2023; 37:e5733. [PMID: 37705144 DOI: 10.1002/bmc.5733] [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: 04/07/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023]
Abstract
While clinical surveys have frequently reported that patients with traumatic brain injuries (TBIs) and comorbidities experience faster healing, the underlying mechanisms have been investigated but remain unclear. As a comprehensive comparison and analysis of the metabolic characteristics of these two pathologies have not been undertaken, we developed a rat model of fracture and TBI and collected serum samples for metabolomic analysis using ultra-high performance liquid chromatography-quadrupole time-of-flight MS (UHPLC-Q-TOF/MS). In total, we identified 40 differential metabolites and uncovered related pathways and potential mechanisms, including aminoacyl-transfer RNA biosynthesis; differential amino acids such as leucine, cholylhistidine, aspartyl-lysine; and related lipid metabolism, and discussed their impacts on bone formation in detail. This study highlights that the UHPLC-Q-TOF/MS-based metabolomics approach offers a better understanding of the metabolic links between TBI and accelerated bone recovery.
Collapse
Affiliation(s)
- Demeng Xia
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Wu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qian Xue
- Cinical Research Unit, Changhai Hospital, Naval Military University, Shanghai, China
| | - Gehan Jiang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Shuogui Xu
- Department of Orthopedics, Changhai Hospital, Naval Military University, Shanghai, China
| |
Collapse
|
12
|
Kitakaze T, Tatsumi R, Yamaguchi M, Nakatsuji A, Harada N, Yamaji R. All- Trans Retinoic Acid-Responsive LGR6 Is Transiently Expressed during Myogenic Differentiation and Is Required for Myoblast Differentiation and Fusion. Int J Mol Sci 2023; 24:ijms24109035. [PMID: 37240382 DOI: 10.3390/ijms24109035] [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: 04/11/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
All-trans retinoic acid (ATRA) promotes myoblast differentiation into myotubes. Leucine-rich repeat-containing G-protein-coupled receptor 6 (LGR6) is a candidate ATRA-responsive gene; however, its role in skeletal muscles remains unclear. Here, we demonstrated that during the differentiation of murine C2C12 myoblasts into myotubes, Lgr6 mRNA expression transiently increased before the increase in the expression of the mRNAs encoding myogenic regulatory factors, such as myogenin, myomaker, and myomerger. The loss of LGR6 decreased the differentiation and fusion indices. The exogenous expression of LGR6 up to 3 and 24 h after the induction of differentiation increased and decreased the mRNA levels of myogenin, myomaker, and myomerger, respectively. Lgr6 mRNA was transiently expressed after myogenic differentiation in the presence of a retinoic acid receptor α (RARα) agonist and an RARγ agonist in addition to ATRA, but not in the absence of ATRA. Furthermore, a proteasome inhibitor or Znfr3 knockdown increased exogenous LGR6 expression. The loss of LGR6 attenuated the Wnt/β-catenin signaling activity induced by Wnt3a alone or in combination with Wnt3a and R-spondin 2. These results indicate that LGR6 promotes myogenic differentiation and that ATRA is required for the transient expression of LGR6 during differentiation. Furthermore, LGR6 expression appeared to be downregulated by the ubiquitin-proteasome system involving ZNRF3.
Collapse
Affiliation(s)
- Tomoya Kitakaze
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 5998531, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 5998531, Osaka, Japan
| | - Rina Tatsumi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 5998531, Osaka, Japan
| | - Mayu Yamaguchi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 5998531, Osaka, Japan
| | - Aino Nakatsuji
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 5998531, Osaka, Japan
| | - Naoki Harada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 5998531, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 5998531, Osaka, Japan
| | - Ryoichi Yamaji
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 5998531, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 5998531, Osaka, Japan
- Center for Research and Development of Bioresources, Osaka Metropolitan University, Sakai 5998531, Osaka, Japan
| |
Collapse
|