1
|
Yamaguchi H, Tran LT, Bi J, Urayama A, Yutzey KE, Mishina Y, Komatsu Y. Enhanced BMP signaling in cranial neural crest cells induces aberrant chondrogenesis by upregulating Tbx20 expression during craniofacial development. Biochem Biophys Res Commun 2025; 765:151834. [PMID: 40273622 DOI: 10.1016/j.bbrc.2025.151834] [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: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Bone morphogenetic proteins (BMPs) are critical for craniofacial development. We previously reported that cranial neural crest cell (CNCC)-specific enhanced BMP signaling through the ALK2 receptor causes ectopic cartilage formation in the face during mouse embryonic development. However, the downstream effectors triggering this ectopic chondrogenesis remain unclear. Here, we investigated the targets of BMP signaling responsible for ectopic cartilage formation. A microarray analysis using CNC-derived ectomesenchymal cells from the first branchial arches identified T-box transcription factor 20 (Tbx20) as the top candidate gene in CNC-specific gain-of-function ALK2 mouse embryos. This prompted us to hypothesize that enhanced BMP signaling increases Tbx20 expression, which triggers ectopic cartilage formation in the craniofacial region. To examine whether Tbx20 overexpression in CNCCs alters craniofacial development, we utilized a Cre-LoxP system to augment Tbx20 expression in a neural crest-specific manner in mice. CNCC-specific overexpression of Tbx20 led to neonatal death with severe craniofacial defects, such as orofacial clefts and exencephaly. Interestingly, aberrant chondrogenesis was observed in the posterior frontal (PF) suture, a structure derived from CNCCs, suggesting that augmented Tbx20 expression triggers ectopic cartilage formation in the PF suture. This study reveals that enhanced BMP-Tbx20 signaling in CNCCs causes aberrant chondrogenesis in the PF suture during craniofacial development.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Lauren T Tran
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jiarui Bi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Periodontics & Dental Hygiene, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Katherine E Yutzey
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA; The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics and Epigenetics, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.
| |
Collapse
|
2
|
Yamaguchi H, Meyer MD, Barrell WB, Faisal M, Berdeaux R, Liu KJ, Komatsu Y. The primary cilia: Orchestrating cranial neural crest cell development. Differentiation 2025; 142:100818. [PMID: 39500655 PMCID: PMC11911094 DOI: 10.1016/j.diff.2024.100818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 03/17/2025]
Abstract
Primary cilia (hereafter "cilia") are microtubule-based antenna-like organelles projecting from the surface of vertebrate cells. Cilia can serve as cellular antennae controlling cell growth and differentiation. Absent or dysfunctional cilia frequently lead to craniofacial anomalies known as craniofacial ciliopathies. However, the detailed pathological mechanisms of craniofacial ciliopathies remain unclear. This perspective discusses our current understanding of the role of cilia in cranial neural crest cells. We also describe potential mechanisms of ciliogenesis in cranial neural crest cells, which may contribute to unraveling the complex pathogenesis of craniofacial ciliopathies.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, USA
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Maryam Faisal
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Bioengineering, Rice University George R. Brown School of Engineering, 77005, Houston, TX, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; CellChorus INC, Houston, TX, USA
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 77030, Houston, TX, USA.
| |
Collapse
|
3
|
Sakinah-Syed G, Liew JS, Abdul Majid N, Inche Zainal Abidin SA. Alteration of primary cilia and intraflagellar transport 20 (IFT20) expression in oral squamous cell carcinoma (OSCC) cell lines. PeerJ 2025; 13:e18931. [PMID: 40017656 PMCID: PMC11867036 DOI: 10.7717/peerj.18931] [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: 09/19/2024] [Accepted: 01/13/2025] [Indexed: 03/01/2025] Open
Abstract
Background Aberrations in primary cilia expression and intraflagellar transport (IFT) protein function have been implicated in tumourigenesis. This study explores the relationship between the alteration of primary cilia and tumourigenesis by investigating primary cilia expression and the role of IFT20 in regulating matrix metalloproteinase 9 (MMP-9) expression in oral squamous cell carcinoma (OSCC) cell lines. Methods The frequency and length of primary cilia were determined in OKF6-TERT2 cells, HSC-2 cells, and HSC-3 cells using immunofluorescence. Additionally, primary cilia presence in non-proliferating OSCC cells was examined. OSCC cells were treated with either small interfering RNA (siRNA) negative control or siRNA targeting IFT20 for functional analysis. mRNA expression levels of IFT20 and MMP-9 were quantified using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results Results showed that HSC-2 cells exhibit abundant primary cilia when cultured in low serum media (2% serum) for 48 h, followed by serum starvation for over 72 h. No significant changes in cilia expression were observed in HSC-3 cells compared to OKF6-TERT2 cells. Ciliated cells were found in non-proliferating HSC-2 and HSC-3 cells. OSCC cells showed longer cilia than OKF6-TERT2 cells, indicating ciliary abnormalities. Changes in ciliation and cilium length of OSCC cells were accompanied by increased expression of IFT20, an intraflagellar transport protein crucial for the primary cilia assembly. However, IFT20 knockdown did not affect MMP-9 at the mRNA level in these cells. Conclusions This study reveals the differences in primary cilia expression among OSCC cells. Furthermore, the increased abundance and elongation of primary cilia in OSCC cells are accompanied by elevated expression of IFT20. Nonetheless, IFT20 did not affect MMP-9 mRNA expression in OSCC cells.
Collapse
Affiliation(s)
- Gulam Sakinah-Syed
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| | - Jia Shi Liew
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| | - Siti Amalina Inche Zainal Abidin
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
- Oral Cancer Research & Coordinating Center, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Yamaguchi H, Kitami M, Li M, Swaminathan S, Darabi R, Takemaru KI, Komatsu Y. Disruption of distal appendage protein CEP164 causes skeletal malformation in mice. Biochem Biophys Res Commun 2024; 741:151063. [PMID: 39612644 PMCID: PMC12011135 DOI: 10.1016/j.bbrc.2024.151063] [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: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
The primary cilium is a cellular antenna to orchestrate cell growth and differentiation. Deficient or dysfunctional cilia are frequently linked to skeletal abnormalities. Previous research demonstrated that ciliary proteins regulating axoneme elongation are essential for skeletogenesis. However, the role of the ciliary proteins responsible for initiating cilium assembly in skeletal development remains unknown. Here, we investigate the function of centrosomal protein of 164 kDa (CEP164), a key ciliogenesis regulator that localizes at the distal appendages of the mother centriole, during skeletal development in mice. Interestingly, the mesodermal cell-specific Cep164 deletion resulted in severe bone defects and osteoblast-specific deletion of Cep164 affected bone development. In contrast, chondrocyte-specific Cep164 deletion did not cause overt skeletal abnormalities, indicating that CEP164 functions in a cell type-specific manner within skeletal tissues. Importantly, Cep164-mutant osteoblasts not only displayed a lack of cilia but also showed an increased number of γH2AX-positive cells, indicating the involvement of defective DNA damage response in the etiology of skeletal lesions of Cep164-mutant mice. These results demonstrate that CEP164 has both ciliary and non-ciliary functions to control osteoblast growth and survival. Our study therefore reveals a novel understanding of the pathogenesis of skeletal ciliopathies associated with CEP164 dysfunction.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Megumi Kitami
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Margaret Li
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Kinesiology, Rice University Wiess School of Natural Science, Houston, TX, USA
| | - Sowmya Swaminathan
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Radbod Darabi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA; Institute of Muscle Biology and Cachexia, University of Houston, Houston, TX, USA
| | - Ken-Ichi Takemaru
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
5
|
Kitami M, Kaku M, Thant L, Maeda T. A loss of primary cilia by a reduction in mTOR signaling correlates with age-related deteriorations in condylar cartilage. GeroScience 2024; 46:5995-6007. [PMID: 38526843 PMCID: PMC11493995 DOI: 10.1007/s11357-024-01143-x] [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: 11/01/2023] [Accepted: 03/16/2024] [Indexed: 03/27/2024] Open
Abstract
Age-related deterioration of condylar cartilage is an etiological factor in temporomandibular joint-osteoarthritis (TMJ-OA). However, its underlying mechanism remains unknown. Therefore, we examined age-related changes and the relationship between mTOR signaling and primary cilia in condylar cartilage to determine the intrinsic mechanisms of age-related TMJ-OA. Age-related morphological changes were analyzed using micro-computed tomography and safranin O-stained histological samples of the mandibular condyle of C57BL/6J mice (up to 78 weeks old). Immunohistochemistry was used to assess the activity of mTOR signaling, primary cilia frequency, and Golgi size of condylar chondrocytes. Four-week-old mice receiving an 11-week series of intraperitoneal injections of rapamycin, a potent mTOR signaling inhibitor, were used for the histological evaluation of the condylar cartilage. The condylar cartilage demonstrated an age-related reduction in cartilage area, including chondrocyte size, cell density, and cell size distribution. The Golgi size, primary cilia frequency, and mTOR signaling also decreased with age. Rapamycin injections resulted in both diminished cartilage area and cell size, resembling the phenotypes observed in aged mice. Rapamycin-injected mice also exhibited a smaller Golgi size and lower primary cilia frequency in condylar cartilage. We demonstrated that a loss of primary cilia due to a decline in mTOR signaling was correlated with age-related deteriorations in condylar cartilage. Our findings provide new insights into the tissue homeostasis of condylar cartilage, contributing to understanding the etiology of age-related TMJ-OA.
Collapse
Affiliation(s)
- Megumi Kitami
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Masaru Kaku
- Division of Bio-Prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Lay Thant
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
6
|
Boldizar H, Friedman A, Stanley T, Padilla M, Galdieri J, Sclar A, Stawicki TM. The role of cilia in the development, survival, and regeneration of hair cells. Biol Open 2024; 13:bio061690. [PMID: 39263863 PMCID: PMC11413933 DOI: 10.1242/bio.061690] [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: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Mutations impacting cilia genes lead to a class of human diseases known as ciliopathies. This is due to the role of cilia in the development, survival, and regeneration of many cell types. We investigated the extent to which disrupting cilia impacted these processes in lateral line hair cells of zebrafish. We found that mutations in two intraflagellar transport (IFT) genes, ift88 and dync2h1, which lead to the loss of kinocilia, caused increased hair cell apoptosis. IFT gene mutants also have a decreased mitochondrial membrane potential, and blocking the mitochondrial uniporter causes a loss of hair cells in wild-type zebrafish but not mutants, suggesting mitochondria dysfunction may contribute to the apoptosis seen in these mutants. These mutants also showed decreased proliferation during hair cell regeneration but did not show consistent changes in support cell number or proliferation during hair cell development. These results show that the loss of hair cells seen following disruption of cilia through either mutations in anterograde or retrograde IFT genes appears to be due to impacts on hair cell survival but not necessarily development in the zebrafish lateral line.
Collapse
Affiliation(s)
- Hope Boldizar
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | - Amanda Friedman
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | - Tess Stanley
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | - María Padilla
- Biology Department, Lafayette College, Easton, PA 18042, USA
| | | | - Arielle Sclar
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | | |
Collapse
|
7
|
Yamaguchi H, Barrell WB, Faisal M, Liu KJ, Komatsu Y. Ciliary and non-ciliary functions of Rab34 during craniofacial bone development. Biochem Biophys Res Commun 2024; 724:150174. [PMID: 38852507 DOI: 10.1016/j.bbrc.2024.150174] [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: 05/22/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024]
Abstract
The primary cilium is a hair-like projection that controls cell development and tissue homeostasis. Although accumulated studies identify the molecular link between cilia and cilia-related diseases, the underlying etiology of ciliopathies has not been fully understood. In this paper, we determine the function of Rab34, a small GTPase, as a key regulator for controlling ciliogenesis and type I collagen trafficking in craniofacial development. Mechanistically, Rab34 is required to form cilia that control osteogenic proliferation, survival, and differentiation via cilia-mediated Hedgehog signaling. In addition, Rab34 is indispensable for regulating type I collagen trafficking from the ER to the Golgi. These results demonstrate that Rab34 has both ciliary and non-ciliary functions to regulate osteogenesis. Our study highlights the critical function of Rab34, which may contribute to understanding the novel etiology of ciliopathies that are associated with the dysfunction of RAB34 in humans.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Maryam Faisal
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Department of Bioengineering, Rice University George R. Brown School of Engineering, Houston, TX, 77005, USA
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Graduate Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Lai B, Jiang H, Gao Y, Zhou X. Skeletal ciliopathy: pathogenesis and related signaling pathways. Mol Cell Biochem 2024; 479:811-823. [PMID: 37188988 DOI: 10.1007/s11010-023-04765-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
Cilia are tiny organelles with conserved structures and components in eukaryotic cells. Ciliopathy is a set of diseases resulting from cilium dysfunction classified into first-order and second-order ciliopathy. With the advancement of clinical diagnosis and radiography, numerous skeletal phenotypes, including polydactyly, short limbs, short ribs, scoliosis, a narrow thorax, and numerous anomalies in bone and cartilage, have been discovered in ciliopathies. Mutation in genes encoding cilia core components or other cilia-related molecules have been found in skeletal ciliopathies. Meanwhile, various signaling pathways associated with cilia and skeleton development have been deemed to be significant for the occurrence and progression of diseases. Herein, we review the structure and key components of the cilium and summarize several skeletal ciliopathies with their presumable pathology. We also emphasize the signaling pathways involved in skeletal ciliopathies, which may assist in developing potential therapies for these diseases.
Collapse
Affiliation(s)
- Bowen Lai
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Fengyang Road 415, Shanghai, 200003, China
| | - Heng Jiang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Fengyang Road 415, Shanghai, 200003, China
| | - Yuan Gao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Fengyang Road 415, Shanghai, 200003, China
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Fengyang Road 415, Shanghai, 200003, China.
| |
Collapse
|
9
|
Li Y, Yang S, Yang S. IFT20 and WWTR1 govern bone homeostasis via synchronously regulating the expression and stability of TβRII in osteoblast lineage cells. RESEARCH SQUARE 2024:rs.3.rs-4009802. [PMID: 38562782 PMCID: PMC10984095 DOI: 10.21203/rs.3.rs-4009802/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Balance of bone and marrow fat formation is critical for bone homeostasis. The imbalance of bone homeostasis will cause various bone diseases, such as osteoporosis. However, the precise mechanisms governing osteoporotic bone loss and marrow adipose tissue (MAT) accumulation remain poorly understood. By analysis of publicly available databases from bone samples of osteoporosis patients, we found that the expression of intraflagellar transport 20 (IFT20) and WW domain containing transcription regulator 1 (WWTR1) were significantly downregulated in osteoblast lineage cells. Additionally, we found that double deletions of IFT20 and WWTR1 in osteoblasts resulted in a significant accumulation of MAT and bone loss. Moreover, IFT20 and WWTR1 deficiency in osteoblasts exacerbated bone-fat imbalance in ovariectomy (OVX)- and high-fat-diet (HFD)-induced osteoporosis mouse models. Mechanistically, we found that deletions of IFT20 and WWTR1 in osteoblasts synergistically inhibited osteogenesis and promoted adipogenesis and osteoclastogenesis. We also found that IFT20 interacted with TGF-β receptor type II (TβRII) to enhance TβRII stability by blocking c-Cbl-mediated ubiquitination and degradation of TβRII. WWTR1 transcriptionally upregulated TβRII expression by directly binding its promoter. These findings indicate that targeting IFT20/WWTR1 may be a potential therapeutic strategy for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yang Li
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthopaedic Surgery, School of Medicine, Johns Hopkins University Baltimore, MD 21205, USA
| | - Shuting Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Yamaguchi H, Li M, Kitami M, Swaminathan S, Mishina Y, Komatsu Y. Enhanced BMP signaling in Cathepsin K-positive tendon progenitors induces heterotopic ossification. Biochem Biophys Res Commun 2023; 688:149147. [PMID: 37948912 PMCID: PMC10952113 DOI: 10.1016/j.bbrc.2023.149147] [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: 08/29/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Heterotopic ossification (HO) is abnormal bone growth in soft tissues that results from injury, trauma, and rare genetic disorders. Bone morphogenetic proteins (BMPs) are critical osteogenic regulators which are involved in HO. However, it remains unclear how BMP signaling interacts with other extracellular stimuli to form HO. To address this question, using the Cre-loxP recombination system in mice, we conditionally expressed the constitutively activated BMP type I receptor ALK2 with a Q207D mutation (Ca-ALK2) in Cathepsin K-Cre labeled tendon progenitors (hereafter "Ca-Alk2:Ctsk-Cre"). Ca-Alk2:Ctsk-Cre mice were viable but they formed spontaneous HO in the Achilles tendon. Histological and molecular marker analysis revealed that HO is formed via endochondral ossification. Ectopic chondrogenesis coincided with enhanced GLI1 production, suggesting that elevated Hedgehog (Hh) signaling is involved in the pathogenesis of HO. Interestingly, focal adhesion kinase, a critical mediator for the mechanotransduction pathway, was also activated in Ca-Alk2:Ctsk-Cre mice. Our findings suggest that enhanced BMP signaling may elevate Hh and mechanotransduction pathways, thereby causing HO in the regions of the Achilles tendon.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Margaret Li
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Department of Kinesiology, Rice University Wiess School of Natural Science, Houston, TX, 77005, USA
| | - Megumi Kitami
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan; Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan
| | - Sowmya Swaminathan
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; The College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Graduate Program in Genetics and Epigenetics, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Kretschmer V, Schneider S, Matthiessen PA, Reichert D, Hotaling N, Glasßer G, Lieberwirth I, Bharti K, De Cegli R, Conte I, Nandrot EF, May-Simera HL. Deletion of IFT20 exclusively in the RPE ablates primary cilia and leads to retinal degeneration. PLoS Biol 2023; 21:e3002402. [PMID: 38048369 PMCID: PMC10721183 DOI: 10.1371/journal.pbio.3002402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/14/2023] [Accepted: 10/26/2023] [Indexed: 12/06/2023] Open
Abstract
Vision impairment places a serious burden on the aging society, affecting the lives of millions of people. Many retinal diseases are of genetic origin, of which over 50% are due to mutations in cilia-associated genes. Most research on retinal degeneration has focused on the ciliated photoreceptor cells of the retina. However, the contribution of primary cilia in other ocular cell types has largely been ignored. The retinal pigment epithelium (RPE) is a monolayer epithelium at the back of the eye intricately associated with photoreceptors and essential for visual function. It is already known that primary cilia in the RPE are critical for its development and maturation; however, it remains unclear whether this affects RPE function and retinal tissue homeostasis. We generated a conditional knockout mouse model, in which IFT20 is exclusively deleted in the RPE, ablating primary cilia. This leads to defective RPE function, followed by photoreceptor degeneration and, ultimately, vision impairment. Transcriptomic analysis offers insights into mechanisms underlying pathogenic changes, which include transcripts related to epithelial homeostasis, the visual cycle, and phagocytosis. Due to the loss of cilia exclusively in the RPE, this mouse model enables us to tease out the functional role of RPE cilia and their contribution to retinal degeneration, providing a powerful tool for basic and translational research in syndromic and non-syndromic retinal degeneration. Non-ciliary mechanisms of IFT20 in the RPE may also contribute to pathogenesis and cannot be excluded, especially considering the increasing evidence of non-ciliary functions of ciliary proteins.
Collapse
Affiliation(s)
- Viola Kretschmer
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Sandra Schneider
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Peter Andreas Matthiessen
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Dominik Reichert
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nathan Hotaling
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gunnar Glasßer
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- University of Naples “Federico II”, Naples, Italy
| | | | - Helen Louise May-Simera
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
12
|
Miao LW, Liu TZ, Sun YH, Cai N, Xuan YY, Wei Z, Cui BB, Jing LL, Ma HP, Xian CJ, Wang JF, Gao YH, Chen KM. Simulated microgravity-induced oxidative stress and loss of osteogenic potential of osteoblasts can be prevented by protection of primary cilia. J Cell Physiol 2023; 238:2692-2709. [PMID: 37796139 DOI: 10.1002/jcp.31127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023]
Abstract
Oxidative stress has been considered to be closely related to spaceflight-induced bone loss; however, mechanism is elusive and there are no effective countermeasures. Using cultured rat calvarial osteoblasts exposed to microgravity simulated by a random positioning machine, this study addressed the hypotheses that microgravity-induced shortening of primary cilia leads to oxidative stress and that primary cilium protection prevents oxidative stress and osteogenesis loss. Microgravity was found to induce oxidative stress (as represented by increased levels of reactive oxygen species (ROS) and malondialdehyde production, and decreased activities of antioxidant enzymes), which was perfectly replicated in osteoblasts growing in NG with abrogated primary cilia (created by transfection of an interfering RNA), suggesting the possibility that shortening of primary cilia leads to oxidative stress. Oxidative stress was accompanied by mitochondrial dysfunction (represented by increased mitochondrial ROS and decreased mitochondrial membrane potential) and intracellular Ca2+ overload, and the latter was found to be caused by increased activity of Ca2+ channel transient receptor potential vanilloid 4 (TRPV4), as also evidenced by TRPV4 agonist GSK1016790A-elicited Ca2+ influx. Supplementation of HC-067047, a specific antagonist of TRPV4, attenuated microgravity-induced mitochondrial dysfunction, oxidative stress, and osteogenesis loss. Although TRPV4 was found localized in primary cilia and expressed at low levels in NG, microgravity-induced shortening of primary cilia led to increased TRPV4 levels and Ca2+ influx. When primary cilia were protected by miR-129-3p overexpression or supplementation with a natural flavonoid moslosooflavone, microgravity-induced increased TRPV4 expression, mitochondrial dysfunction, oxidative stress, and osteogenesis loss were all prevented. Our data revealed a new mechanism that primary cilia function as a controller for TRPV4 expression. Microgravity-induced injury on primary cilia leads to increased expression and overactive channel of TRPV4, causing intracellular Ca2+ overload and oxidative stress, and primary cilium protection could be an effective countermeasure against microgravity-induced oxidative stress and loss of osteogenic potential of osteoblasts.
Collapse
Affiliation(s)
- Lu-Wei Miao
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Tian-Zhen Liu
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Yue-Hong Sun
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Nan Cai
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Ying-Ying Xuan
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Zhenlong Wei
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Bing-Bing Cui
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Lin-Lin Jing
- Department of Pharmacy, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Hui-Ping Ma
- Department of Pharmacy, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Cory J Xian
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ju-Fang Wang
- Gansu Key Laboratory of Space Radiobiology, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Yu-Hai Gao
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Ke-Ming Chen
- Fundamental Medical Science Research Laboratories, Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, China
| |
Collapse
|
13
|
Terada N, Saitoh Y, Saito M, Yamada T, Kamijo A, Yoshizawa T, Sakamoto T. Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families. Genes (Basel) 2023; 14:1942. [PMID: 37895291 PMCID: PMC10606877 DOI: 10.3390/genes14101942] [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: 10/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The protein 4.1 and membrane palmitoylated protein (MPP) families were originally found as components in the erythrocyte membrane skeletal protein complex, which helps maintain the stability of erythrocyte membranes by linking intramembranous proteins and meshwork structures composed of actin and spectrin under the membranes. Recently, it has been recognized that cells and tissues ubiquitously use this membrane skeletal system. Various intramembranous proteins, including adhesion molecules, ion channels, and receptors, have been shown to interact with the 4.1 and MPP families, regulating cellular and tissue dynamics by binding to intracellular signal transduction proteins. In this review, we focus on our previous studies regarding genetically modified animal models, especially on 4.1G, MPP6, and MPP2, to describe their functional roles in the peripheral nervous system, the central nervous system, the testis, and bone formation. As the membrane skeletal proteins are located at sites that receive signals from outside the cell and transduce signals inside the cell, it is necessary to elucidate their molecular interrelationships, which may broaden the understanding of cell and tissue functions.
Collapse
Affiliation(s)
- Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
| | - Yurika Saitoh
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo 120-0045, Japan
| | - Masaki Saito
- School of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan;
| | - Tomoki Yamada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
| | - Akio Kamijo
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
- Division of Basic & Clinical Medicine, Nagano College of Nursing, Komagane City, Nagano 399-4117, Japan
| | - Takahiro Yoshizawa
- Division of Animal Research, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto City, Nagano 390-8621, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| |
Collapse
|
14
|
Onodera S, Azuma T. Hedgehog-Related Mutation Causes Bone Malformations with or without Hereditary Gene Mutations. Int J Mol Sci 2023; 24:12903. [PMID: 37629084 PMCID: PMC10454035 DOI: 10.3390/ijms241612903] [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: 07/18/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The hedgehog (Hh) family consists of numerous signaling mediators that play important roles at various stages of development. Thus, the Hh pathway is essential for bone tissue development and tumorigenesis. Gorlin syndrome is a skeletal and tumorigenic disorder caused by gain-of-function mutations in Hh signaling. In this review, we first present the phenotype of Gorlin syndrome and the relationship between genotype and phenotype in bone and craniofacial tissues, including the causative gene as well as other Hh-related genes. Next, the importance of new diagnostic methods using next-generation sequencing and multiple gene panels will be discussed. We summarize Hh-related genetic disorders, including cilia disease, and the genetics of Hh-related bone diseases.
Collapse
Affiliation(s)
- Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, 2-9-18 Kanda Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan;
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, 2-9-18 Kanda Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan;
- Oral Health Science Center, Tokyo Dental College, 2-9-18 Kanda Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
15
|
Jeong J, Kang I, Kim Y, Ku KB, Park JH, Kim HJ, Kim CW, La J, Jung HE, Kim HC, Choi YJ, Kim J, Kim J, Lee HK. Regulation of c-SMAC formation and AKT-mTOR signaling by the TSG101-IFT20 axis in CD4 + T cells. Cell Mol Immunol 2023; 20:525-539. [PMID: 37029318 PMCID: PMC10202954 DOI: 10.1038/s41423-023-01008-x] [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: 12/14/2021] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
CD4+ T cells play major roles in the adaptive immune system, which requires antigen recognition, costimulation, and cytokines for its elaborate orchestration. Recent studies have provided new insight into the importance of the supramolecular activation cluster (SMAC), which comprises concentric circles and is involved in the amplification of CD4+ T cell activation. However, the underlying mechanism of SMAC formation remains poorly understood. Here, we performed single-cell RNA sequencing of CD4+ T cells left unstimulated and stimulated with anti-CD3 and anti-CD28 antibodies to identify novel proteins involved in their regulation. We found that intraflagellar transport 20 (IFT20), previously known as cilia-forming protein, was upregulated in antibody-stimulated CD4+ T cells compared to unstimulated CD4+ T cells. We also found that IFT20 interacted with tumor susceptibility gene 101 (TSG101), a protein that endocytoses ubiquitinated T-cell receptors. The interaction between IFT20 and TSG101 promoted SMAC formation, which led to amplification of AKT-mTOR signaling. However, IFT20-deficient CD4+ T cells showed SMAC malformation, resulting in reduced CD4+ T cell proliferation, aerobic glycolysis, and cellular respiration. Finally, mice with T-cell-specific IFT20 deficiency exhibited reduced allergen-induced airway inflammation. Thus, our data suggest that the IFT20-TSG101 axis regulates AKT-mTOR signaling via SMAC formation.
Collapse
Affiliation(s)
- Jiung Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - In Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yumin Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Keun Bon Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jang Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyun-Jin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Chae Won Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jeongwoo La
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hi Eun Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyeon Cheol Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Young Joon Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jaeho Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
16
|
Stevenson NL. The factory, the antenna and the scaffold: the three-way interplay between the Golgi, cilium and extracellular matrix underlying tissue function. Biol Open 2023; 12:287059. [PMID: 36802341 PMCID: PMC9986613 DOI: 10.1242/bio.059719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
The growth and development of healthy tissues is dependent on the construction of a highly specialised extracellular matrix (ECM) to provide support for cell growth and migration and to determine the biomechanical properties of the tissue. These scaffolds are composed of extensively glycosylated proteins which are secreted and assembled into well-ordered structures that can hydrate, mineralise, and store growth factors as required. The proteolytic processing and glycosylation of ECM components is vital to their function. These modifications are under the control of the Golgi apparatus, an intracellular factory hosting spatially organised, protein-modifying enzymes. Regulation also requires a cellular antenna, the cilium, which integrates extracellular growth signals and mechanical cues to inform ECM production. Consequently, mutations in either Golgi or ciliary genes frequently lead to connective tissue disorders. The individual importance of each of these organelles to ECM function is well-studied. However, emerging evidence points towards a more tightly linked system of interdependence between the Golgi, cilium and ECM. This review examines how the interplay between all three compartments underpins healthy tissue. As an example, it will look at several members of the golgin family of Golgi-resident proteins whose loss is detrimental to connective tissue function. This perspective will be important for many future studies looking to dissect the cause and effect of mutations impacting tissue integrity.
Collapse
Affiliation(s)
- Nicola L Stevenson
- Cell Biology Laboratories, School of Biochemistry, Faculty of Biomedical Sciences University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
17
|
Liu J, Leng F, Gao Y, He W, Wang J, Xian CJ, Ma H, Chen K. Protection of primary cilia is an effective countermeasure against the impairment of osteoblast function induced by simulated microgravity. J Cell Mol Med 2022; 27:36-51. [PMID: 36512344 PMCID: PMC9806295 DOI: 10.1111/jcmm.17628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
The molecular mechanism for the microgravity-induced decrease in bone formation remains unclear and there is a lack of effective specific preventative therapies. We recently reported that primary cilia of osteoblasts became shorter and even disappeared when the cells were exposed to random positioning machine (RPM)-simulated microgravity and that the microgravity-induced loss of osteogenic potential of osteoblasts could be attenuated when the resorption of primary cilia was prevented by treatment with 0.1 μM cytochalasin D. In the current study, it was further found that the loss of the osteogenic capacity of rat calvarial osteoblasts (ROBs) was associated with the inhibition of the BMP-2/Smad1/5/8 signalling pathway, of which most of the signalling proteins including BMP-2, BMPRII, Smad1/5/8 and p-Smad1/5/8 were found localized to primary cilia. Accompanying the resorption of primary cilia following the cells being exposed to simulated microgravity, the expression levels of these signalling proteins were reduced significantly. Furthermore, the expression of miRNA-129-3p, a microRNA previously reported to control cilium biogenesis, was found to be reduced quickly and changed in a similar tendency with the length of primary cilia. Moreover, overexpression of miRNA-129-3p in ROBs significantly attenuated microgravity-induced inhibition of BMP-2 signalling and loss of osteogenic differentiation and mineralization. These results indicated the important role of miRNA-129-3p in microgravity-induced resorption of primary cilia of osteoblasts and the potential of replenishing the miRNA-129-3p as an effective countermeasure against microgravity-induced loss of primary cilia and impairment of osteoblast function.
Collapse
Affiliation(s)
- Jing Liu
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support ForcePeople's Liberation Army of ChinaLanzhouChina
| | - Fei‐Fan Leng
- Department of Bioengineering, School of Life Science and EngineeringLanzhou University of TechnologyLanzhouChina
| | - Yu‐Hai Gao
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support ForcePeople's Liberation Army of ChinaLanzhouChina
| | - Wen‐Fang He
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support ForcePeople's Liberation Army of ChinaLanzhouChina
| | - Ju‐Fang Wang
- Gansu Key Laboratory of Space RadiobiologyInstitute of Modern Physics, Chinese Academy of SciencesLanzhouChina
| | - Cory J. Xian
- UniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Hui‐Ping Ma
- Department of Pharmacy, The 940th Hospital of Joint Logistic Support ForcePeople's Liberation Army of ChinaLanzhouChina
| | - Ke‐Ming Chen
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support ForcePeople's Liberation Army of ChinaLanzhouChina
| |
Collapse
|
18
|
Yamaguchi H, Shen J, Little DR, Li M, Sozen S, Suzuki K, Mishina Y, Komatsu Y. Enhanced BMP signaling through ALK2 attenuates keratinocyte differentiation. Biochem Biophys Res Commun 2022; 629:101-105. [PMID: 36116371 DOI: 10.1016/j.bbrc.2022.09.014] [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: 07/25/2022] [Revised: 07/29/2022] [Accepted: 09/02/2022] [Indexed: 11/02/2022]
Abstract
Accumulated studies have suggested that bone morphogenetic proteins (BMPs) are critical for skin development. However, it remains elusive how BMP signaling via ALK2 (aka ACVR1), one of the important BMP type I receptors, regulates keratinocyte differentiation. To address this question, we utilized a genetic system that enhances BMP signaling via ALK2 in an epidermis-specific manner in mice (hereafter ca-Alk2:K14-Cre). Ca-Alk2:K14-Cre mice displayed a sticky and hairless skin phenotype with a thinner epidermis incapable of differentiating. Although cellular proliferation and survival were comparable between wild-type and ca-Alk2:K14-Cre mice, skin differentiation was severely hampered in ca-Alk2:K14-Cre mice. To uncover the mechanism of altered keratinocyte differentiation, we performed a transcriptome analysis. As a result, we found that the expression levels of cell cycle inhibitor p21 were increased in ca-Alk2:K14-Cre mice. Our findings suggest that aberrant BMP signaling via ALK2 positively regulates p21 expression that attenuates keratinocyte differentiation, and further highlights the critical role of BMP signaling in skin development.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jingling Shen
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China.
| | - Danielle R Little
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Margaret Li
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Department of Kinesiology, Rice University Wiess School of Natural Science, Houston, TX, 77005, USA
| | - Serra Sozen
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Department of Medicine, The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Kentaro Suzuki
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, 400-8510, Japan
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Graduate Program in Genes and Development, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Finetti F, Onnis A, Baldari CT. IFT20: An Eclectic Regulator of Cellular Processes beyond Intraflagellar Transport. Int J Mol Sci 2022; 23:ijms232012147. [PMID: 36292997 PMCID: PMC9603483 DOI: 10.3390/ijms232012147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Initially discovered as the smallest component of the intraflagellar transport (IFT) system, the IFT20 protein has been found to be implicated in several unconventional mechanisms beyond its essential role in the assembly and maintenance of the primary cilium. IFT20 is now considered a key player not only in ciliogenesis but also in vesicular trafficking of membrane receptors and signaling proteins. Moreover, its ability to associate with a wide array of interacting partners in a cell-type specific manner has expanded the function of IFT20 to the regulation of intracellular degradative and secretory pathways. In this review, we will present an overview of the multifaceted role of IFT20 in both ciliated and non-ciliated cells.
Collapse
|
20
|
Wang Z, Sa G, Zheng L, Wei Z, Zhang Z, Hu Y, Yang X. Intraflagellar transport protein 88 interacts with polycystin 2 to regulate mechanosensitive hedgehog signaling in mandibular condylar chondrocytes. Arch Oral Biol 2022; 143:105548. [PMID: 36155344 DOI: 10.1016/j.archoralbio.2022.105548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/03/2022] [Accepted: 09/17/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aimed to explore whether intraflagellar transport protein 88 (IFT88) was associated with polycystin 2 during mechanotransduction of mandibular condylar chondrocytes. METHODS Rat mandibular condylar chondrocytes isolated from the condylar bone-cartilage junction were subjected to cyclic tensile strain (0.1 Hz, 10% elongation). Overexpression of IFT88 was achieved by lentiviral vector-mediated transfection. Knockdown of IFT88 and polycystin 2 was achieved by small interfering RNA (siRNA). The prevalence and length of cilia were reflected by immunofluorescence staining. The activities of hedgehog signaling were evaluated by western blot analysis. The interaction between polycystin 2 and IFT88 was evaluated by conducting a co-immunoprecipitation (co-IP) assay. RESULTS Overexpression of IFT88 increased the length of cilia. Protein levels of polycystin 2, Indian hedgehog (Ihh), Patched 1 (Ptch1), Smoothened (Smo), and Glioma-associated oncogene homolog 1 (Gli1) were elevated in IFT88-overexpressing mandibular condylar chondrocytes under cyclic tensile strain. Knockdown of the protein level of IFT88 reduced the prevalence and length of cilia, and protein levels of polycystin 2, Ihh, Ptch1, Smo, and Gli1. A co-IP assay showed that IFT88 formed a complex with polycystin 2 under cyclic tensile strain. Knockdown of polycystin 2 decreased the protein levels of IFT88, Ihh, Ptch1, Smo, and Gli1 in mandibular condylar chondrocytes following cyclic tensile strain. CONCLUSION These findings highlight the vital role of an interaction between IFT88 and polycystin 2 in mechanosensitive hedgehog signaling in mandibular condylar chondrocytes following cyclic tensile strain, which suggest that therapies regulating polycystin 2 may be considered for the disorders of temporomandibular joints.
Collapse
Affiliation(s)
- Zhuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guoliang Sa
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Liwu Zheng
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Zequan Wei
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhuoyu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yanping Hu
- Stomatological Hospital of Xiamen Medical Collage, Xiamen, China
| | - Xuewen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
21
|
Xu J, Li Z, Tower RJ, Negri S, Wang Y, Meyers CA, Sono T, Qin Q, Lu A, Xing X, McCarthy EF, Clemens TL, James AW. NGF-p75 signaling coordinates skeletal cell migration during bone repair. SCIENCE ADVANCES 2022; 8:eabl5716. [PMID: 35302859 PMCID: PMC8932666 DOI: 10.1126/sciadv.abl5716] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/26/2022] [Indexed: 05/26/2023]
Abstract
Bone regeneration following injury is initiated by inflammatory signals and occurs in association with infiltration by sensory nerve fibers. Together, these events are believed to coordinate angiogenesis and tissue reprogramming, but the mechanism of coupling immune signals to reinnervation and osteogenesis is unknown. Here, we found that nerve growth factor (NGF) is expressed following cranial bone injury and signals via p75 in resident mesenchymal osteogenic precursors to affect their migration into the damaged tissue. Mice lacking Ngf in myeloid cells demonstrated reduced migration of osteogenic precursors to the injury site with consequently delayed bone healing. These features were phenocopied by mice lacking p75 in Pdgfra+ osteoblast precursors. Single-cell transcriptomics identified mesenchymal subpopulations with potential roles in cell migration and immune response, altered in the context of p75 deletion. Together, these results identify the role of p75 signaling pathway in coordinating skeletal cell migration during early bone repair.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert J. Tower
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Orthopaedics and Traumatology, University of Verona, Verona 37129, Italy
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Carolyn A. Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Amy Lu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Edward F. McCarthy
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thomas L. Clemens
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Abstract
The primary cilium is a nonmotile microtubule-based organelle in most vertebrate cell types. The primary cilium plays a critical role in tissue development and homeostasis by sensing and transducing various signaling pathways. Ciliary proteins such as intraflagellar transport (IFT) proteins as well as ciliary motor proteins, kinesin and dynein, comprise a bidirectional intraflagellar transport system needed for cilia formation and function. Mutations in ciliary proteins that lead to loss or dysfunction of primary cilia cause ciliopathies such as Jeune syndrome and Ellis-van Creveld syndrome and cause abnormalities in tooth development. These diseases exhibit severe skeletal and craniofacial dysplasia, highlighting the significance of primary cilia in skeletal development. Cilia are necessary for the propagation of hedgehog, transforming growth factor β, platelet-derived growth factor, and fibroblast growth factor signaling during osteogenesis and chondrogenesis. Ablation of ciliary proteins such as IFT80 or IFT20 blocks cilia formation, which inhibits osteoblast differentiation, osteoblast polarity, and alignment and reduces bone formation. Similarly, cilia facilitate chondrocyte differentiation and production of a cartilage matrix. Cilia also play a key role in mechanosensing and are needed for increased bone formation in response to mechanical forces.
Collapse
Affiliation(s)
- Z. Chinipardaz
- Department of Basic and
Translational Sciences, University of Pennsylvania, School of Dental
Medicine, Philadelphia, PA, USA,Department of Periodontics,
School of Dental Medicine, University of Pennsylvania, Philadelphia, PA,
USA
| | - M. Liu
- Department of Periodontics,
School of Dental Medicine, University of Pennsylvania, Philadelphia, PA,
USA
| | - D.T. Graves
- Department of Periodontics,
School of Dental Medicine, University of Pennsylvania, Philadelphia, PA,
USA
| | - S. Yang
- Department of Basic and
Translational Sciences, University of Pennsylvania, School of Dental
Medicine, Philadelphia, PA, USA,Center for Innovation &
Precision Dentistry, School of Dental Medicine, School of Engineering and
Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA,The Penn Center for
Musculoskeletal Disorders, School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA,S. Yang, Department of Basic and
Translational Sciences, University of Pennsylvania, School of Dental
Medicine, 240 S 40th Street, Philadelphia, PA 19104-6243, USA.
| |
Collapse
|
23
|
Liao J, Huang Y, Wang Q, Chen S, Zhang C, Wang D, Lv Z, Zhang X, Wu M, Chen G. Gene regulatory network from cranial neural crest cells to osteoblast differentiation and calvarial bone development. Cell Mol Life Sci 2022; 79:158. [PMID: 35220463 PMCID: PMC11072871 DOI: 10.1007/s00018-022-04208-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/02/2022] [Accepted: 02/14/2022] [Indexed: 11/03/2022]
Abstract
Calvarial bone is one of the most complex sequences of developmental events in embryology, featuring a uniquely transient, pluripotent stem cell-like population known as the cranial neural crest (CNC). The skull is formed through intramembranous ossification with distinct tissue lineages (e.g. neural crest derived frontal bone and mesoderm derived parietal bone). Due to CNC's vast cell fate potential, in response to a series of inductive secreted cues including BMP/TGF-β, Wnt, FGF, Notch, Hedgehog, Hippo and PDGF signaling, CNC enables generations of a diverse spectrum of differentiated cell types in vivo such as osteoblasts and chondrocytes at the craniofacial level. In recent years, since the studies from a genetic mouse model and single-cell sequencing, new discoveries are uncovered upon CNC patterning, differentiation, and the contribution to the development of cranial bones. In this review, we summarized the differences upon the potential gene regulatory network to regulate CNC derived osteogenic potential in mouse and human, and highlighted specific functions of genetic molecules from multiple signaling pathways and the crosstalk, transcription factors and epigenetic factors in orchestrating CNC commitment and differentiation into osteogenic mesenchyme and bone formation. Disorders in gene regulatory network in CNC patterning indicate highly close relevance to clinical birth defects and diseases, providing valuable transgenic mouse models for subsequent discoveries in delineating the underlying molecular mechanisms. We also emphasized the potential regenerative alternative through scientific discoveries from CNC patterning and genetic molecules in interfering with or alleviating clinical disorders or diseases, which will be beneficial for the molecular targets to be integrated for novel therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qiang Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Sisi Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Chenyang Zhang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Dan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhengbing Lv
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Mengrui Wu
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
24
|
Saito M, Hirano M, Izumi T, Mori Y, Ito K, Saitoh Y, Terada N, Sato T, Sukegawa J. Cytoskeletal Protein 4.1G Is Essential for the Primary Ciliogenesis and Osteoblast Differentiation in Bone Formation. Int J Mol Sci 2022; 23:ijms23042094. [PMID: 35216233 PMCID: PMC8878336 DOI: 10.3390/ijms23042094] [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: 12/28/2021] [Revised: 02/06/2022] [Accepted: 02/10/2022] [Indexed: 12/04/2022] Open
Abstract
The primary cilium is a hair-like immotile organelle with specific membrane receptors, including the receptor of Hedgehog signaling, smoothened. The cilium organized in preosteoblasts promotes differentiation of the cells into osteoblasts (osteoblast differentiation) by mediating Hedgehog signaling to achieve bone formation. Notably, 4.1G is a plasma membrane-associated cytoskeletal protein that plays essential roles in various tissues, including the peripheral nervous system, testis, and retina. However, its function in the bone remains unexplored. In this study, we identified 4.1G expression in the bone. We found that, in the 4.1G-knockout mice, calcium deposits and primary cilium formation were suppressed in the trabecular bone, which is preosteoblast-rich region of the newborn tibia, indicating that 4.1G is a prerequisite for osteoblast differentiation by organizing the primary cilia in preosteoblasts. Next, we found that the primary cilium was elongated in the differentiating mouse preosteoblast cell line MC3T3-E1, whereas the knockdown of 4.1G suppressed its elongation. Moreover, 4.1G-knockdown suppressed the induction of the cilia-mediated Hedgehog signaling and subsequent osteoblast differentiation. These results demonstrate a new regulatory mechanism of 4.1G in bone formation that promotes the primary ciliogenesis in the differentiating preosteoblasts and induction of cilia-mediated osteoblast differentiation, resulting in bone formation at the newborn stage.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
- Correspondence: ; Tel.: +81-22-717-8207
| | - Marina Hirano
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
- Department of Human Health and Nutrition, Shokei Gakuin University, Natori 981-1295, Japan;
| | - Tomohiro Izumi
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
| | - Yu Mori
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (Y.M.); (K.I.)
| | - Kentaro Ito
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (Y.M.); (K.I.)
| | - Yurika Saitoh
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo 120-0045, Japan;
| | - Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto 390-0802, Japan;
| | - Takeya Sato
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
| | - Jun Sukegawa
- Department of Human Health and Nutrition, Shokei Gakuin University, Natori 981-1295, Japan;
| |
Collapse
|
25
|
Yamaguchi H, Meyer MD, He L, Komatsu Y. Disruption of Trip11 in cranial neural crest cells is associated with increased ER and Golgi stress contributing to skull defects in mice. Dev Dyn 2022; 251:1209-1222. [PMID: 35147267 DOI: 10.1002/dvdy.461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Absence of Golgi microtubule-associated protein 210 (GMAP210), encoded by the TRIP11 gene, results in achondrogenesis. Although TRIP11 is thought to be specifically required for chondrogenesis, human fetuses with the mutation of TRIP11 also display bony skull defects where chondrocytes are usually not present. This raises an important question of how TRIP11 functions in bony skull development. RESULTS We disrupted Trip11 in neural crest-derived cell populations, which are critical for developing skull in mice. In Trip11 mutant skulls, expression levels of ER stress markers were increased compared to controls. Morphological analysis of electron microscopy data revealed swollen ER in Trip11 mutant skulls. Unexpectedly, we also found that Golgi stress increased in Trip11 mutant skulls, suggesting that both ER and Golgi stress-induced cell death may lead to osteopenia-like phenotypes in Trip11 mutant skulls. These data suggest that Trip11 plays pivotal roles in the regulation of ER and Golgi stress, which are critical for osteogenic cell survival. CONCLUSION We have recently reported that the molecular complex of ciliary protein and GMAP210 is required for collagen trafficking. In this paper, we further characterized the important role of Trip11 being possibly involved in the regulation of ER and Golgi stress during skull development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, Texas, USA
| | - Li He
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
26
|
Yamaguchi H, Kitami M, Uchima Koecklin KH, He L, Wang J, Lagor WR, Perrien DS, Komatsu Y. Temporospatial regulation of intraflagellar transport is required for the endochondral ossification in mice. Dev Biol 2021; 482:91-100. [PMID: 34929174 DOI: 10.1016/j.ydbio.2021.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/24/2021] [Accepted: 12/10/2021] [Indexed: 01/28/2023]
Abstract
Ciliogenic components, such as the family of intraflagellar transport (IFT) proteins, are recognized to play key roles in endochondral ossification, a critical process to form most bones. However, the unique functions and roles of each IFT during endochondral ossification remain unclear. Here, we show that IFT20 is required for endochondral ossification in mice. Utilizing osteo-chondrocyte lineage-specific Cre mice (Prx1-Cre and Col2-Cre), we deleted Ift20 to examine its function. Although chondrocyte-specific Ift20 deletion with Col2-Cre mice did not cause any overt skeletal defects, mesoderm-specific Ift20 deletion using Prx1-Cre (Ift20:Prx1-Cre) mice resulted in shortened limb outgrowth. Primary cilia were absent on chondrocytes of Ift20:Prx1-Cre mice, and ciliary-mediated Hedgehog signaling was attenuated in Ift20:Prx1-Cre mice. Interestingly, loss of Ift20 also increased Fgf18 expression in the perichondrium that sustained Sox9 expression, thus preventing endochondral ossification. Inhibition of enhanced phospho-ERK1/2 activation partially rescued defective chondrogenesis in Ift20 mutant cells, supporting an important role for FGF signaling. Our findings demonstrate that IFT20 is a critical regulator of temporospatial FGF signaling that is required for endochondral ossification.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, UTHealth, Houston, TX, 77030, USA
| | - Megumi Kitami
- Department of Pediatrics, McGovern Medical School, UTHealth, Houston, TX, 77030, USA
| | | | - Li He
- Department of Pediatrics, McGovern Medical School, UTHealth, Houston, TX, 77030, USA
| | - Jianbo Wang
- Department of Pediatrics, McGovern Medical School, UTHealth, Houston, TX, 77030, USA
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daniel S Perrien
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University, Atlanta, GA, 30232, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, UTHealth, Houston, TX, 77030, USA; Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center, UTHealth, Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Quidwai T, Wang J, Hall EA, Petriman NA, Leng W, Kiesel P, Wells JN, Murphy LC, Keighren MA, Marsh JA, Lorentzen E, Pigino G, Mill P. A WDR35-dependent coat protein complex transports ciliary membrane cargo vesicles to cilia. eLife 2021; 10:e69786. [PMID: 34734804 PMCID: PMC8754431 DOI: 10.7554/elife.69786] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Intraflagellar transport (IFT) is a highly conserved mechanism for motor-driven transport of cargo within cilia, but how this cargo is selectively transported to cilia is unclear. WDR35/IFT121 is a component of the IFT-A complex best known for its role in ciliary retrograde transport. In the absence of WDR35, small mutant cilia form but fail to enrich in diverse classes of ciliary membrane proteins. In Wdr35 mouse mutants, the non-core IFT-A components are degraded and core components accumulate at the ciliary base. We reveal deep sequence homology of WDR35 and other IFT-A subunits to α and ß' COPI coatomer subunits and demonstrate an accumulation of 'coat-less' vesicles that fail to fuse with Wdr35 mutant cilia. We determine that recombinant non-core IFT-As can bind directly to lipids and provide the first in situ evidence of a novel coat function for WDR35, likely with other IFT-A proteins, in delivering ciliary membrane cargo necessary for cilia elongation.
Collapse
Affiliation(s)
- Tooba Quidwai
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Jiaolong Wang
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Emma A Hall
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Narcis A Petriman
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Weihua Leng
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Petra Kiesel
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Jonathan N Wells
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Laura C Murphy
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Margaret A Keighren
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Esben Lorentzen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Gaia Pigino
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Human TechnopoleMilanItaly
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
28
|
Primary cilia in hard tissue development and diseases. Front Med 2021; 15:657-678. [PMID: 34515939 DOI: 10.1007/s11684-021-0829-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/13/2020] [Indexed: 10/20/2022]
Abstract
Bone and teeth are hard tissues. Hard tissue diseases have a serious effect on human survival and quality of life. Primary cilia are protrusions on the surfaces of cells. As antennas, they are distributed on the membrane surfaces of almost all mammalian cell types and participate in the development of organs and the maintenance of homeostasis. Mutations in cilium-related genes result in a variety of developmental and even lethal diseases. Patients with multiple ciliary gene mutations present overt changes in the skeletal system, suggesting that primary cilia are involved in hard tissue development and reconstruction. Furthermore, primary cilia act as sensors of external stimuli and regulate bone homeostasis. Specifically, substances are trafficked through primary cilia by intraflagellar transport, which affects key signaling pathways during hard tissue development. In this review, we summarize the roles of primary cilia in long bone development and remodeling from two perspectives: primary cilia signaling and sensory mechanisms. In addition, the cilium-related diseases of hard tissue and the manifestations of mutant cilia in the skeleton and teeth are described. We believe that all the findings will help with the intervention and treatment of related hard tissue genetic diseases.
Collapse
|
29
|
Yamaguchi H, Meyer MD, He L, Senavirathna L, Pan S, Komatsu Y. The molecular complex of ciliary and golgin protein is crucial for skull development. Development 2021; 148:270770. [PMID: 34128978 DOI: 10.1242/dev.199559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/27/2021] [Indexed: 01/13/2023]
Abstract
Intramembranous ossification, which consists of direct conversion of mesenchymal cells to osteoblasts, is a characteristic process in skull development. One crucial role of these osteoblasts is to secrete collagen-containing bone matrix. However, it remains unclear how the dynamics of collagen trafficking is regulated during skull development. Here, we reveal the regulatory mechanisms of ciliary and golgin proteins required for intramembranous ossification. During normal skull formation, osteoblasts residing on the osteogenic front actively secreted collagen. Mass spectrometry and proteomic analysis determined endogenous binding between ciliary protein IFT20 and golgin protein GMAP210 in these osteoblasts. As seen in Ift20 mutant mice, disruption of neural crest-specific GMAP210 in mice caused osteopenia-like phenotypes due to dysfunctional collagen trafficking. Mice lacking both IFT20 and GMAP210 displayed more severe skull defects compared with either IFT20 or GMAP210 mutants. These results demonstrate that the molecular complex of IFT20 and GMAP210 is essential for the intramembranous ossification during skull development.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX 77005, USA
| | - Li He
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Lakmini Senavirathna
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sheng Pan
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
30
|
Marquez J, Mann N, Arana K, Deniz E, Ji W, Konstantino M, Mis EK, Deshpande C, Jeffries L, McGlynn J, Hugo H, Widmeier E, Konrad M, Tasic V, Morotti R, Baptista J, Ellard S, Lakhani SA, Hildebrandt F, Khokha MK. DLG5 variants are associated with multiple congenital anomalies including ciliopathy phenotypes. J Med Genet 2021; 58:453-464. [PMID: 32631816 PMCID: PMC7785698 DOI: 10.1136/jmedgenet-2019-106805] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/01/2020] [Accepted: 05/25/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cilia are dynamic cellular extensions that generate and sense signals to orchestrate proper development and tissue homeostasis. They rely on the underlying polarisation of cells to participate in signalling. Cilia dysfunction is a well-known cause of several diseases that affect multiple organ systems including the kidneys, brain, heart, respiratory tract, skeleton and retina. METHODS Among individuals from four unrelated families, we identified variants in discs large 5 (DLG5) that manifested in a variety of pathologies. In our proband, we also examined patient tissues. We depleted dlg5 in Xenopus tropicalis frog embryos to generate a loss-of-function model. Finally, we tested the pathogenicity of DLG5 patient variants through rescue experiments in the frog model. RESULTS Patients with variants of DLG5 were found to have a variety of phenotypes including cystic kidneys, nephrotic syndrome, hydrocephalus, limb abnormalities, congenital heart disease and craniofacial malformations. We also observed a loss of cilia in cystic kidney tissue of our proband. Knockdown of dlg5 in Xenopus embryos recapitulated many of these phenotypes and resulted in a loss of cilia in multiple tissues. Unlike introduction of wildtype DLG5 in frog embryos depleted of dlg5, introduction of DLG5 patient variants was largely ineffective in restoring proper ciliation and tissue morphology in the kidney and brain suggesting that the variants were indeed detrimental to function. CONCLUSION These findings in both patient tissues and Xenopus shed light on how mutations in DLG5 may lead to tissue-specific manifestations of disease. DLG5 is essential for cilia and many of the patient phenotypes are in the ciliopathy spectrum.
Collapse
Affiliation(s)
- Jonathan Marquez
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nina Mann
- Division of Nephrology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kathya Arana
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Engin Deniz
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Weizhen Ji
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Monica Konstantino
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emily K Mis
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Lauren Jeffries
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Julie McGlynn
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hannah Hugo
- Division of Nephrology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eugen Widmeier
- Division of Nephrology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Martin Konrad
- Department of General Pediatrics, University Hospital Münster, Münster, Germany
| | - Velibor Tasic
- Department of Pediatric Nephrology, University Children's Hospital, Skopje, North Macedonia
| | - Raffaella Morotti
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Julia Baptista
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
- Institute of Biomedical & Clinical Science, College of Medicine and Health, Exeter, UK
| | - Sian Ellard
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
- Institute of Biomedical & Clinical Science, College of Medicine and Health, Exeter, UK
| | - Saquib Ali Lakhani
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Friedhelm Hildebrandt
- Division of Nephrology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
31
|
Jovasevic V, Zhang H, Sananbenesi F, Guedea AL, Soman KV, Wiktorowicz JE, Fischer A, Radulovic J. Primary cilia are required for the persistence of memory and stabilization of perineuronal nets. iScience 2021; 24:102617. [PMID: 34142063 PMCID: PMC8185192 DOI: 10.1016/j.isci.2021.102617] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/02/2021] [Accepted: 05/19/2021] [Indexed: 01/11/2023] Open
Abstract
It is well established that the formation of episodic memories requires multiple hippocampal mechanisms operating on different time scales. Early mechanisms of memory formation (synaptic consolidation) have been extensively characterized. However, delayed mechanisms, which maintain hippocampal activity as memories stabilize in cortical circuits, are not well understood. Here we demonstrate that contrary to the transient expression of early- and delayed-response genes, the expression of cytoskeleton- and extracellular matrix-associated genes remains dynamic even at remote time points. The most profound expression changes clustered around primary cilium-associated and collagen genes. These genes most likely contribute to memory by stabilizing perineuronal nets in the dorsohippocampal CA1 subfield, as revealed by targeted disruptions of the primary cilium or perineuronal nets. The findings show that nonsynaptic, primary cilium-mediated mechanisms are required for the persistence of context memory.
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Room 13-100, Montgomery Ward Memorial Building, Chicago, IL 60611, USA
| | - Hui Zhang
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Rose F. Kennedy Center, 1410 Pelham Parkway South, Room 115, Bronx, NY 10461, USA
| | | | - Anita L. Guedea
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL 60611, USA
| | - Kizhake V. Soman
- Division of Infectious Disease, Department of Internal Medicine, UTMB – Galveston, Galveston, TX 77555, USA
| | | | - Andre Fischer
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
| | - Jelena Radulovic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Room 13-100, Montgomery Ward Memorial Building, Chicago, IL 60611, USA
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Rose F. Kennedy Center, 1410 Pelham Parkway South, Room 115, Bronx, NY 10461, USA
| |
Collapse
|
32
|
A Neurotrophic Mechanism Directs Sensory Nerve Transit in Cranial Bone. Cell Rep 2021; 31:107696. [PMID: 32460020 PMCID: PMC7335423 DOI: 10.1016/j.celrep.2020.107696] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/17/2020] [Accepted: 05/06/2020] [Indexed: 11/21/2022] Open
Abstract
The flat bones of the skull are densely innervated during development, but little is known regarding their role during repair. We describe a neurotrophic mechanism that directs sensory nerve transit in the mouse calvaria. Patent cranial suture mesenchyme represents an NGF (nerve growth factor)-rich domain, in which sensory nerves transit. Experimental calvarial injury upregulates Ngf in an IL-1β/TNF-α-rich defect niche, with consequent axonal ingrowth. In calvarial osteoblasts, IL-1β and TNF-α stimulate Ngf and downstream NF-κB signaling. Locoregional deletion of Ngf delays defect site re-innervation and blunted repair. Genetic disruption of Ngf among LysM-expressing macrophages phenocopies these observations, whereas conditional knockout of Ngf among Pdgfra-expressing cells does not. Finally, inhibition of TrkA catalytic activity similarly delays re-innervation and repair. These results demonstrate an essential role of NGF-TrkA signaling in bone healing and implicate macrophage-derived NGF-induced ingrowth of skeletal sensory nerves as an important mediator of this repair.
Collapse
|
33
|
Douanne T, Stinchcombe JC, Griffiths GM. Teasing out function from morphology: Similarities between primary cilia and immune synapses. J Cell Biol 2021; 220:212075. [PMID: 33956049 PMCID: PMC8105739 DOI: 10.1083/jcb.202102089] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Immune synapses are formed between immune cells to facilitate communication and coordinate the immune response. The reorganization of receptors involved in recognition and signaling creates a transient area of plasma membrane specialized in signaling and polarized secretion. Studies on the formation of the immune synapse between cytotoxic T lymphocytes (CTLs) and their targets uncovered a critical role for centrosome polarization in CTL function and suggested a striking parallel between the synapse and primary cilium. Since these initial observations, a plethora of further morphological, functional, and molecular similarities have been identified between these two fascinating structures. In this review, we describe how advances in imaging and molecular techniques have revealed additional parallels as well as functionally significant differences and discuss how comparative studies continue to shed light on the molecular mechanisms underlying the functions of both the immune synapse and primary cilium.
Collapse
Affiliation(s)
- Tiphaine Douanne
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, University of Cambridge, Cambridge, England, UK
| | - Jane C Stinchcombe
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, University of Cambridge, Cambridge, England, UK
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, University of Cambridge, Cambridge, England, UK
| |
Collapse
|
34
|
Yamaguchi H, Kitami K, Wu X, He L, Wang J, Wang B, Komatsu Y. Alteration of DNA Damage Response Causes Cleft Palate. Front Physiol 2021; 12:649492. [PMID: 33854442 PMCID: PMC8039291 DOI: 10.3389/fphys.2021.649492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Cleft palate is one of the most common craniofacial birth defects, however, little is known about how changes in the DNA damage response (DDR) cause cleft palate. To determine the role of DDR during palatogenesis, the DDR process was altered using a pharmacological intervention approach. A compromised DDR caused by a poly (ADP-ribose) polymerase (PARP) enzyme inhibitor resulted in cleft palate in wild-type mouse embryos, with increased DNA damage and apoptosis. In addition, a mouse genetic approach was employed to disrupt breast cancer 1 (BRCA1) and breast cancer 2 (BRCA2), known as key players in DDR. An ectomesenchymal-specific deletion of Brca1 or Brca2 resulted in cleft palate due to attenuation of cell survival. This was supported by the phenotypes of the ectomesenchymal-specific Brca1/Brca2 double-knockout mice. The cleft palate phenotype was rescued by superimposing p53 null alleles, demonstrating that the BRCA1/2-p53 DDR pathway is critical for palatogenesis. Our study highlights the importance of DDR in palatogenesis.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Kohei Kitami
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Xiao Wu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Li He
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Jianbo Wang
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Bin Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Yoshihiro Komatsu
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
35
|
McGowan SE, McCoy DM. Neuropilin-1 directs PDGFRα-entry into lung fibroblasts and signaling from very early endosomes. Am J Physiol Lung Cell Mol Physiol 2021; 320:L179-L192. [PMID: 33174445 DOI: 10.1152/ajplung.00149.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/29/2020] [Accepted: 10/28/2020] [Indexed: 01/16/2023] Open
Abstract
Platelet-derived growth factor receptor-α (PDGFRα) is absolutely required for the development of secondary pulmonary alveolar septa. Our earlier observations indicated that PDGFRα resides intracellularly as well as on the plasma membrane of murine lung fibroblasts (LF). We have examined how neuropilin-1 (Nrp1), a surface receptor without kinase activity, regulates the intracellular trafficking of PDGFRα in LF obtained from mice, some bearing a targeted deletion of Nrp1 in myofibroblasts. Using the proximity ligation assay, we observed that PDGFRα and Nrp1 colocalized in both early antigen-1 (EEA1) containing sorting endosomes and with adaptor protein containing a pleckstrin homology domain and a phosphotyrosine-binding domain-1 (APPL1) in very early endosomes (VEE). These findings were confirmed using live-cell imaging, which demonstrated that recently internalized PDGFRα was observed in Rab5-containing vesicles residing within 100 nm of the plasma membrane. Nrp1 deletion reduced the phosphorylation of Akt (protein kinase B), the major downstream target of PDGFRα, and limited accumulation of inositol-3 phosphates in APPL1-containing endosomes after exposure to PDGFA. PDGFRα co-immunoprecipitated with APPL1, indicating that PDGFRα enters VEE. Targeted deletion of Nrp1 or APPL1-depletion in control LF reduced the activity of an Akt1 biosensor following stimulation with PDGFA. Our findings demonstrate that Nrp1 enhances the entry of PDGFRα into APPL1 containing VEE and that APPL1 enhances PDGFRα signaling. Therefore, Nrp1 promotes endosomal signaling by PDGFRα offering a potential mechanism to explain our prior observation that Nrp1 supports the formation of alveolar ducts and alveoli during secondary septation in mice.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
36
|
Corkins ME, Krneta-Stankic V, Kloc M, Miller RK. Aquatic models of human ciliary diseases. Genesis 2021; 59:e23410. [PMID: 33496382 PMCID: PMC8593908 DOI: 10.1002/dvg.23410] [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: 11/09/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 11/06/2022]
Abstract
Cilia are microtubule-based structures that either transmit information into the cell or move fluid outside of the cell. There are many human diseases that arise from malfunctioning cilia. Although mammalian models provide vital insights into the underlying pathology of these diseases, aquatic organisms such as Xenopus and zebrafish provide valuable tools to help screen and dissect out the underlying causes of these diseases. In this review we focus on recent studies that identify or describe different types of human ciliopathies and outline how aquatic organisms have aided our understanding of these diseases.
Collapse
Affiliation(s)
- Mark E. Corkins
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston Texas 77030
| | - Vanja Krneta-Stankic
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genes & Development, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics & Epigenetics, Houston, Texas 77030
| | - Malgorzata Kloc
- Houston Methodist, Research Institute, Houston Texas 77030
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston Texas 77030
| | - Rachel K. Miller
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics & Epigenetics, Houston, Texas 77030
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Biochemistry & Cell Biology, Houston Texas 77030
| |
Collapse
|
37
|
Paulson D, Harms R, Ward C, Latterell M, Pazour GJ, Fink DM. Loss of Primary Cilia Protein IFT20 Dysregulates Lymphatic Vessel Patterning in Development and Inflammation. Front Cell Dev Biol 2021; 9:672625. [PMID: 34055805 PMCID: PMC8160126 DOI: 10.3389/fcell.2021.672625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Microenvironmental signals produced during development or inflammation stimulate lymphatic endothelial cells to undergo lymphangiogenesis, in which they sprout, proliferate, and migrate to expand the vascular network. Many cell types detect changes in extracellular conditions via primary cilia, microtubule-based cellular protrusions that house specialized membrane receptors and signaling complexes. Primary cilia are critical for receipt of extracellular cues from both ligand-receptor pathways and physical forces such as fluid shear stress. Here, we report the presence of primary cilia on immortalized mouse and primary adult human dermal lymphatic endothelial cells in vitro and on both luminal and abluminal domains of mouse corneal, skin, and mesenteric lymphatic vessels in vivo. The purpose of this study was to determine the effects of disrupting primary cilia on lymphatic vessel patterning during development and inflammation. Intraflagellar transport protein 20 (IFT20) is part of the transport machinery required for ciliary assembly and function. To disrupt primary ciliary signaling, we generated global and lymphatic endothelium-specific IFT20 knockout mouse models and used immunofluorescence microscopy to quantify changes in lymphatic vessel patterning at E16.5 and in adult suture-mediated corneal lymphangiogenesis. Loss of IFT20 during development resulted in edema, increased and more variable lymphatic vessel caliber and branching, as well as red blood cell-filled lymphatics. We used a corneal suture model to determine ciliation status of lymphatic vessels during acute, recurrent, and tumor-associated inflammatory reactions and wound healing. Primary cilia were present on corneal lymphatics during all of the mechanistically distinct lymphatic patterning events of the model and assembled on lymphatic endothelial cells residing at the limbus, stalk, and vessel tip. Lymphatic-specific deletion of IFT20 cell-autonomously exacerbated acute corneal lymphangiogenesis resulting in increased lymphatic vessel density and branching. These data are the first functional studies of primary cilia on lymphatic endothelial cells and reveal a new dimension in regulation of lymphatic vascular biology.
Collapse
Affiliation(s)
- Delayna Paulson
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Rebecca Harms
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Cody Ward
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Mackenzie Latterell
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Darci M. Fink
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
- *Correspondence: Darci M. Fink,
| |
Collapse
|
38
|
Yamaguchi H, Terajima M, Kitami M, Wang J, He L, Saeki M, Yamauchi M, Komatsu Y. IFT20 is critical for collagen biosynthesis in craniofacial bone formation. Biochem Biophys Res Commun 2020; 533:739-744. [PMID: 32988591 DOI: 10.1016/j.bbrc.2020.09.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 10/23/2022]
Abstract
Intraflagellar transport (IFT) is essential for assembling primary cilia required for bone formation. Disruption of IFT frequently leads to bone defects in humans. While it has been well studied about the function of IFT in osteogenic cell proliferation and differentiation, little is known about its role in collagen biosynthesis during bone formation. Here we show that IFT20, the smallest IFT protein in the IFT-B complex, is important for collagen biosynthesis in mice. Deletion of Ift20 in craniofacial osteoblasts displayed bone defects in the face. While collagen protein levels are unaffected by loss of Ift20, collagen cross-linking was significantly altered. In both Ift20:Wnt1-Cre and Ift20:Ocn-Cre mice the bones exhibit increased hydroxylysine-aldehyde deived cross-linking, and decreased lysine-aldehyde derived cross-linking. To obtain insight into the molecular mechanisms, we examined the expression levels of telopeptidyl lysyl hydroxylase 2 (LH2), and associated chaperone complexes. The results demonstrated that, while LH2 levels were unaffected by loss of Ift20, its chaperone, FKBP65, was significantly increased in Ift20:Wnt1-Cre and Ift20:Ocn-Cre mouse calvaria as well as femurs. These results suggest that IFT20 plays a pivotal role in collagen biosynthesis by regulating, in part, telopeptidyl lysine hydroxylation and cross-linking in bone. To the best of our knowledge, this is the first to demonstrate that the IFT components control collagen post-translational modifications. This provides a novel insight into the craniofacial bone defects associated with craniofacial skeletal ciliopathies.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Masahiko Terajima
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Megumi Kitami
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan; Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan
| | - Jianbo Wang
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Li He
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Makio Saeki
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan
| | - Mitsuo Yamauchi
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Yoshihiro Komatsu
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA; Graduate Program in Genetics and Epigenetics, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
39
|
Barsch F, Niedermair T, Mamilos A, Schmitt VH, Grevenstein D, Babel M, Burgoyne T, Shoemark A, Brochhausen C. Physiological and Pathophysiological Aspects of Primary Cilia-A Literature Review with View on Functional and Structural Relationships in Cartilage. Int J Mol Sci 2020; 21:ijms21144959. [PMID: 32674266 PMCID: PMC7404129 DOI: 10.3390/ijms21144959] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cilia are cellular organelles that project from the cell. They occur in nearly all non-hematopoietic tissues and have different functions in different tissues. In mesenchymal tissues primary cilia play a crucial role in the adequate morphogenesis during embryological development. In mature articular cartilage, primary cilia fulfil chemo- and mechanosensitive functions to adapt the cellular mechanisms on extracellular changes and thus, maintain tissue homeostasis and morphometry. Ciliary abnormalities in osteoarthritic cartilage could represent pathophysiological relationships between ciliary dysfunction and tissue deformation. Nevertheless, the molecular and pathophysiological relationships of ‘Primary Cilia’ (PC) in the context of osteoarthritis is not yet fully understood. The present review focuses on the current knowledge about PC and provide a short but not exhaustive overview of their role in cartilage.
Collapse
Affiliation(s)
- Friedrich Barsch
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany and Institute of Exercise and Occupational Medicine, Department of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Tanja Niedermair
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
| | - Andreas Mamilos
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
| | - Volker H. Schmitt
- Cardiology I, Centre for Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55122 Mainz, Germany;
| | - David Grevenstein
- Department for Orthopedic and Trauma Surgery, University of Cologne, 50923 Köln, Germany;
| | - Maximilian Babel
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
| | - Thomas Burgoyne
- Royal Brompton Hospital and Harefield NHS Trust, SW3 6NP London and UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| | - Amelia Shoemark
- Royal Brompton Hospital and Harefield NHS Trust, University of Dundee, Dundee DD1 4HN, UK;
| | - Christoph Brochhausen
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (T.N.); (A.M.); (M.B.)
- Correspondence: ; Tel.: +49-941-944-6636
| |
Collapse
|
40
|
Shi W, Zhang Y, Chen K, He J, Feng X, Wei W, Hua J, Wang J. Primary cilia act as microgravity sensors by depolymerizing microtubules to inhibit osteoblastic differentiation and mineralization. Bone 2020; 136:115346. [PMID: 32240849 DOI: 10.1016/j.bone.2020.115346] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 11/18/2022]
Abstract
Microgravity-induced bone deterioration is a major challenge in long-term spaceflights since the underlying mechanisms remain elusive. Previously, we reported that primary cilia of osteoblasts gradually disappeared in microgravity conditions, and cilia abrogation was necessary for the inhibition of osteogenesis induced by microgravity. However, the precise roles of primary cilia have not been fully elucidated. Here, we report that microgravity depolymerizes the microtubule network of rat calvarial osteoblasts (ROBs) reversibly but has no effect on the architecture of actin filaments. Preventing primary ciliogenesis by chloral hydrate or a small interfering RNA sequence (siRNA) targeting intraflagellar transport protein 88 (IFT88) effectively relieves microgravity-induced microtubule depolymerization, whereas the stabilization of microtubules using pharmacological approaches cannot prevent the disappearance of primary cilia in microgravity conditions. Furthermore, quantification of the number of microtubules emerging from the ciliary base body shows that microgravity significantly decreases the number of basal microtubules, which is dependent on the existence of primary cilia. Finally, microgravity-induced repression of the differentiation, maturation, and mineralization of ROBs is abrogated by the stabilization of cytoplasmic microtubules. Taken together, these data suggest that primary cilia-dependent depolymerization of microtubules is responsible for the inhibition of osteogenesis induced by microgravity. Our study provides a new perspective regarding the mechanism of microgravity-induced bone loss, supporting the previously established role of primary cilia as a sensor in bone metabolism.
Collapse
Affiliation(s)
- Wengui Shi
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Yanan Zhang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Keming Chen
- Institute of Orthopaedics, Joint Logistic Support 940 Hospital of CPLA, Lanzhou 730050, China
| | - Jinpeng He
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Xiu Feng
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjun Wei
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Junrui Hua
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jufang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
41
|
Li X, Yang S, Han L, Mao K, Yang S. Ciliary IFT80 is essential for intervertebral disc development and maintenance. FASEB J 2020; 34:6741-6756. [PMID: 32227389 DOI: 10.1096/fj.201902838r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/19/2020] [Accepted: 03/14/2020] [Indexed: 12/24/2022]
Abstract
The intervertebral disc degeneration (IVDD)-related diseases occur in more than 90% of the population older than 50 years. Owing to the lack of understanding of the cellular mechanisms involved in IVDD formation effective treatment options are still unavailable. Primary cilia are microtubule-based organelles that play important roles in the organ development. Intraflagellar transport (IFT) proteins are essential for the assembly and bidirectional transport within the cilium. Role of cilia and IFT80 protein in intervertebral disc (IVD) development, maintenance, and degeneration are largely unknown. Using cilia-GFP mice, we found presence of cilia on growth plate (GP), cartilage endplate (EP) annulus fibrosus (AF), and nucleus pulposus (NP) with varying ciliary length. Cilia length in NP and AF during IVDD were significantly decreased. However, cilia numbers increased by 63% in AF during repair. Deletion of IFT80 in type II collagen-positive cells resulted in cilia loss in GP and EP, and disrupted IVD structure with disorganized and decreased GP, EP, and internal AF (IAF), and less compact and markedly decreased gel-like matrix in the NP. Deletion of IFT80 in type I collagen-positive cells led to a disorganized outer AF (OAF) with thinner, loosened, and disconnected fiber alignment. Mechanistic analyses showed that loss of IFT80 caused a significant increase in cell apoptosis in the IVD, and a marked decrease in expression of chondrogenic markers - type II collagen, sox9, aggrecan, and hedgehog (Hh) signaling components, including Gli1 and Patch1 in the IVD of IFT80fl/fl ; Col2-creERT mice, and Gli1 and Patch1 expression in the OAF of IFT80fl/fl ; Col1-creERT mice. Interestingly, Smoothened agonist-SAG rescued OAF cell proliferation and osteogenic differentiation. Our findings demonstrate that ciliary IFT80 is important for the maintenance of IVD cell organization and function through regulating the cell survival and Hh signaling.
Collapse
Affiliation(s)
- Xinhua Li
- Department of Basic and Translational Science, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Spinal Surgery, East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Shuting Yang
- Department of Basic and Translational Science, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Keya Mao
- Department of Orthopedics, Chinese PLA General Hospital (301 Hospital), Beijing, China
| | - Shuying Yang
- Department of Basic and Translational Science, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
42
|
Yang H, Huang K. Dissecting the Vesicular Trafficking Function of IFT Subunits. Front Cell Dev Biol 2020; 7:352. [PMID: 32010685 PMCID: PMC6974671 DOI: 10.3389/fcell.2019.00352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022] Open
Abstract
Intraflagellar transport (IFT) was initially identified as a transport machine with multiple protein subunits, and it is essential for the assembly, disassembly, and maintenance of cilium/flagellum, which serves as the nexus of extracellular-to-intracellular signal integration. To date, in addition to its well-established and indispensable roles in ciliated cells, most IFT subunits have presented more general functions of vesicular trafficking in the non-ciliated cells. Thus, this review aims to summarize the recent progress on the vesicular trafficking functions of the IFT subunits and to highlight the issues that may arise in future research.
Collapse
Affiliation(s)
- Huihui Yang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,Institute of Hydrobiology, University of Chinese Academy of Sciences, Beijing, China
| | - Kaiyao Huang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
43
|
Lim J, Li X, Yuan X, Yang S, Han L, Yang S. Primary cilia control cell alignment and patterning in bone development via ceramide-PKCζ-β-catenin signaling. Commun Biol 2020; 3:45. [PMID: 31988398 PMCID: PMC6985158 DOI: 10.1038/s42003-020-0767-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 12/16/2019] [Indexed: 02/01/2023] Open
Abstract
Intraflagellar transport (IFT) proteins are essential for cilia assembly and function. IFT protein mutations lead to ciliopathies, which manifest as variable skeletal abnormalities. However, how IFT proteins regulate cell alignment during bone development is unknown. Here, we show that the deletion of IFT20 in osteoblast lineage using Osterix-Cre and inducible type I Collagen-CreERT cause a compromised cell alignment and a reduced bone mass. This finding was validated by the disorganized collagen fibrils and decreased bone strength and stiffness in IFT20-deficient femurs. IFT20 maintains cilia and cell alignment in osteoblasts, as the concentric organization of three-dimensional spheroids was disrupted by IFT20 deletion. Mechanistically, IFT20 interacts with the ceramide-PKCζ complex to promote PKCζ phosphorylation in cilia and induce the apical localization of β-catenin in osteoblasts, both of which were disrupted in the absence of IFT20. These results reveal that IFT20 regulates polarity and cell alignment via ceramide-pPKCζ-β-catenin signaling during bone development.
Collapse
Affiliation(s)
- Jormay Lim
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Xinhua Li
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Xue Yuan
- Department of Oral Biology, State University of New York at Buffalo, School of Dental Medicine, Buffalo, NY, USA
| | - Shuting Yang
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Lin Han
- Department of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Shuying Yang
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA.
- Department of Oral Biology, State University of New York at Buffalo, School of Dental Medicine, Buffalo, NY, USA.
- The Penn Center for Musculoskeletal Disorders, University of Pennsylvania, School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
44
|
Regulation of the Extracellular Matrix by Ciliary Machinery. Cells 2020; 9:cells9020278. [PMID: 31979260 PMCID: PMC7072529 DOI: 10.3390/cells9020278] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/13/2020] [Accepted: 01/19/2020] [Indexed: 12/14/2022] Open
Abstract
The primary cilium is an organelle involved in cellular signalling. Mutations affecting proteins involved in cilia assembly or function result in diseases known as ciliopathies, which cause a wide variety of phenotypes across multiple tissues. These mutations disrupt various cellular processes, including regulation of the extracellular matrix. The matrix is important for maintaining tissue homeostasis through influencing cell behaviour and providing structural support; therefore, the matrix changes observed in ciliopathies have been implicated in the pathogenesis of these diseases. Whilst many studies have associated the cilium with processes that regulate the matrix, exactly how these matrix changes arise is not well characterised. This review aims to bring together the direct and indirect evidence for ciliary regulation of matrix, in order to summarise the possible mechanisms by which the ciliary machinery could regulate the composition, secretion, remodelling and organisation of the matrix.
Collapse
|
45
|
Intraflagellar transport 20: New target for the treatment of ciliopathies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118641. [PMID: 31893523 DOI: 10.1016/j.bbamcr.2019.118641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 11/22/2022]
Abstract
Cilia are ubiquitous in mammalian cells. The formation and assembly of cilia depend on the normal functioning of the ciliary transport system. In recent years, various proteins involved in the intracellular transport of the cilium have attracted attention, as many diseases are caused by disorders in cilia formation. Intraflagellar transport 20 (IFT20) is a subunit of IFT complex B, which contains approximately 20 protein particles. Studies have shown that defects in IFT20 are associated with numerous system -related diseases, such as those of the urinary system, cardiovascular system, skeletal system, nervous system, immune system, reproductive system, and respiratory system. This review summarizes current research on IFT20.We describe studies related to the role of IFT20 in cilia formation and discuss new targets for treating diseases associated with ciliary dysplasia.
Collapse
|
46
|
Abstract
Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.
Collapse
|
47
|
Chen G, Yao Y, Xu G, Zhang X. Regional difference in microRNA regulation in the skull vault. Dev Dyn 2019; 248:1009-1019. [PMID: 31397024 DOI: 10.1002/dvdy.97] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The murine calvaria has several membrane bones with different tissue origins (e.g., neural crest-derived frontal bone vs. mesoderm-derived parietal bone). Neural crest-derived frontal bone exhibits superior osteogenic activities and bone regeneration. MicroRNA (miRNA) has been emerged as a crucial regulator during organogenesis and is involved in a range of developmental processes. However, the underlying roles of miRNA regulation in frontal bone and parietal bone is unknown. RESULTS Total of 83 significantly expressed known miRNAs were identified in frontal bones versus parietal bones. The significantly enriched gene ontology and KEGG pathway that were predicted by the enrichment miRNAs were involved in several biological processes (cell differentiation, cell adhesion, and transcription), and multiple osteogenic pathways (e.g., focal adhesion, MAPK, VEGF, Wnt, and insulin signaling pathway. Focal adhesion and insulin signaling pathway were selected for target verification and functional analysis, and several genes were predicted to be targets genes by the differentially expressed miRNAs, and these targets genes were tested with significant expressions. CONCLUSIONS Our results revealed a novel pattern of miRNAs in murine calvaria with dual tissue origins, and explorations of these miRNAs will be valuable for the translational studies to enhance osteogenic potential and bone regeneration in the clinic.
Collapse
Affiliation(s)
- Guiqian Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, China
| | - Yifeng Yao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, China
| | - Guangtao Xu
- Department of Pathology and Molecular Medicine, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing University, Jiaxing, China
| | - Xingen Zhang
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
48
|
Yuan X, Liu M, Cao X, Yang S. Ciliary IFT80 regulates dental pulp stem cells differentiation by FGF/FGFR1 and Hh/BMP2 signaling. Int J Biol Sci 2019; 15:2087-2099. [PMID: 31592124 PMCID: PMC6775288 DOI: 10.7150/ijbs.27231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 02/19/2019] [Indexed: 01/09/2023] Open
Abstract
Primary cilia and intraflagellar transport (IFT) proteins control a wide variety of processes during development and tissue homeostasis. However, their potential roles in the regulation of stem cell differentiation and tooth development remain elusive. Here, we uncovered the critical roles of ciliary IFT80 in cilia formation and differentiation of dental pulp stem cells (DPSCs). IFT80-deficient DPSCs showed reduced fibroblast growth factor receptor 1 (FGFR1) expression, leading to the disruption of FGF2-FGFR1 signaling. We found, during DPSC differentiation, FGF2-FGFR1 signaling induces stress fiber rearrangement to promote cilia elongation, meanwhile stimulates PI3K-AKT signaling to aid Hh/bone morphogenetic protein 2 (BMP2) signaling activation. These signaling pathways and their coupling were disrupted in IFT80-deficient DPSCs, causing impaired differentiation. Our findings revealed a novel mechanism that ciliary protein regulates the odontogenic differentiation of DPSCs through FGF/FGFR1 and Hh/BMP2 signaling.
Collapse
Affiliation(s)
- Xue Yuan
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, United States
| | - Min Liu
- Department of Anatomy & Cell Biology, School of Dental Medicine, University of Pennsylvania, PA, United States
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, United States
- Department of Anatomy & Cell Biology, School of Dental Medicine, University of Pennsylvania, PA, United States
| |
Collapse
|
49
|
Cassioli C, Baldari CT. A Ciliary View of the Immunological Synapse. Cells 2019; 8:E789. [PMID: 31362462 PMCID: PMC6721628 DOI: 10.3390/cells8080789] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 12/28/2022] Open
Abstract
The primary cilium has gone from being a vestigial organelle to a crucial signaling hub of growing interest given the association between a group of human disorders, collectively known as ciliopathies, and defects in its structure or function. In recent years many ciliogenesis proteins have been observed at extraciliary sites in cells and likely perform cilium-independent functions ranging from regulation of the cytoskeleton to vesicular trafficking. Perhaps the most striking example is the non-ciliated T lymphocyte, in which components of the ciliary machinery are repurposed for the assembly and function of the immunological synapse even in the absence of a primary cilium. Furthermore, the specialization traits described at the immunological synapse are similar to those seen in the primary cilium. Here, we review common regulators and features shared by the immunological synapse and the primary cilium that document the remarkable homology between these structures.
Collapse
Affiliation(s)
- Chiara Cassioli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
50
|
Lowe M. The Physiological Functions of the Golgin Vesicle Tethering Proteins. Front Cell Dev Biol 2019; 7:94. [PMID: 31316978 PMCID: PMC6611411 DOI: 10.3389/fcell.2019.00094] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/16/2019] [Indexed: 01/02/2023] Open
Abstract
The golgins comprise a family of vesicle tethering proteins that act in a selective manner to tether transport vesicles at the Golgi apparatus. Tethering is followed by membrane fusion to complete the delivery of vesicle-bound cargo to the Golgi. Different golgins are localized to different regions of the Golgi, and their ability to selectively tether transport vesicles is important for the specificity of vesicle traffic in the secretory pathway. In recent years, our mechanistic understanding of golgin-mediated tethering has greatly improved. We are also beginning to appreciate how the loss of golgin function can impact upon physiological processes through the use of animal models and the study of human disease. These approaches have revealed that loss of a golgin causes tissue-restricted phenotypes, which can vary in severity and the cell types affected. In many cases, it is possible to attribute these phenotypes to a defect in vesicular traffic, although why certain tissues are sensitive to loss of a particular golgin is still, in most cases, unclear. Here, I will summarize recent progress in our understanding of golgins, focusing on the physiological roles of these proteins, as determined from animal models and the study of disease in humans. I will describe what these in vivo analyses have taught us, as well as highlight less understood aspects, and areas for future investigations.
Collapse
Affiliation(s)
- Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|