1
|
Yuan L, Zhao N, Wang J, Liu Y, Meng L, Guo S, Wiemer EA, Chen Q, Mao Y, Ben J, Ma J. Major vault protein (MVP) negatively regulates osteoclastogenesis via calcineurin-NFATc1 pathway inhibition. Theranostics 2021; 11:7247-7261. [PMID: 34158848 PMCID: PMC8210610 DOI: 10.7150/thno.58468] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Rationale: Bone homeostasis is maintained by a balanced interplay of osteoblasts and osteoclasts. Osteoclasts are derived from monocyte/macrophage lineage. Major vault protein (MVP) is known to promote apoptosis and prevent metabolic diseases in macrophage. However, whether MVP is involved in osteoclastogenesis is unknown. Here, we identified an important function of MVP as a negative regulator of osteoclastogenesis and its therapeutic potential in preventing bone loss. Methods: Expression of MVP in osteoclasts was investigated in human tumor tissues with immunohistochemical staining. Next, we generated total body (Mvp-/- ) and monocyte-specific (Mvpf/fLyz2-Cre) MVP gene knockout mice to observe bone phenotype and osteoclastogenesis using micro-CT and bone histomorphometry. Moreover, we examined the effects of MVP on osteoclast differentiation, bone resorption, NFATc1 activation and calcium oscillations in vitro. Finally, we explored the clinical potential of targeting MVP in two osteoporosis mouse models and used an adeno-associated virus (AAV) gene to overexpress MVP locally in mice. Results: We found that Mvp-/- and Mvpf/fLyz2-Cre mice both exhibited osteoporosis-like phenotypes. MVP-deficiency also enhanced calcineurin-NFATc1 signaling and promoted NFATc1 activity, which led to enhanced osteoclastogenesis and bone resorption. Calcineurin inhibition using the small molecule inhibitor FK506 corrected the enhanced osteoclastogenesis in Mvpf/fLyz2-Cre group. Additionally, MVP reexpression in Mvpf/fLyz2-Cre group rescued calcineurin expression. MVP overexpression in wild-type mice prevented pathologic bone loss in mouse models of ovariectomized (OVX) and calvaria-adjacent lipopolysaccharide (LPS)-injected. Conclusions: Our data suggested that MVP negatively regulates osteoclast differentiation and bone resorption via inhibition of calcineurin-NFATc1 signaling. In osteoclast-related bone diseases such as osteoporosis, manipulation of MVP activity may be an attractive therapeutic target.
Collapse
|
2
|
Cody JD. The Consequences of Abnormal Gene Dosage: Lessons from Chromosome 18. Trends Genet 2020; 36:764-776. [PMID: 32660784 DOI: 10.1016/j.tig.2020.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022]
Abstract
Accurate interpretation of genomic copy number variation (CNV) remains a challenge and has important consequences for both congenital and late-onset disease. Hemizygosity dosage characterization of the genes on chromosome 18 reveals a spectrum of outcomes ranging from no clinical effect, to risk factors for disease, to both low- and high-penetrance disease. These data are important for accurate and predictive clinical management. Additionally, the potential mechanisms of reduced penetrance due to dosage compensation are discussed as a key to understanding avenues for potential treatment. This review describes the chromosome 18 findings, and discusses the molecular mechanisms that allow haploinsufficiency, reduced penetrance, and dosage compensation.
Collapse
Affiliation(s)
- Jannine DeMars Cody
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Chromosome 18 Registry and Research Society, San Antonio, TX 78229, USA.
| |
Collapse
|
3
|
Ma C, Geng B, Zhang X, Li R, Yang X, Xia Y. Fluid Shear Stress Suppresses Osteoclast Differentiation in RAW264.7 Cells through Extracellular Signal-Regulated Kinase 5 (ERK5) Signaling Pathway. Med Sci Monit 2020; 26:e918370. [PMID: 31914120 PMCID: PMC6977602 DOI: 10.12659/msm.918370] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Although extracellular signal-regulated kinase 5 (ERK5) is known to be critical for osteoclast differentiation, there are few studies on how fluid shear stress (FSS) regulates osteoclast differentiation through the ERK5 signaling pathway. We examined the expression of nuclear factor of activated T cells c1 (NFATc1) in RAW264.7 cells and its downstream factors, including cathepsin K (CTSK), tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinases-9 (MMP-9) and their relationship with ERK5. Material/Methods RAW264.7 cells were treated with RANKL, XMD8-92 (ERK5 inhibitor), and then loaded onto 12 dyn/cm2 FSS for 4 days. Endpoints measured were osteoclast differentiation, bone resorption, and TRAP activity. Cell viability was detected by using the Cell Counting Kit-8 (CCK-8) assay. Western blot was used to analyze protein expression of phosphorylated-ERK5 (p-ERK5), NFATc1, CTSK, TRAP, and MMP-9. Results FSS inhibited osteoclast differentiation and expression of NFATc1, CTSK, TRAP, and MMP-9; cell viability was not affected. ERK5 expression increased by FSS but not by RANKL, and it was blocked by XMD8-92. Furthermore, FSS suppressed osteoclast differentiation in RAW264.7 cells through ERK5 pathway. Conclusions Our findings demonstrated that FSS inhibited osteoclast differentiation in RAW264.7 cells via the ERK5 pathway through reduced NFATc1 expression and its downstream factors MMP-9, CTSK, and TRAP.
Collapse
Affiliation(s)
- Chongwen Ma
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Xiaohui Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Rui Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Xinxin Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| |
Collapse
|
4
|
Ding N, Geng B, Li Z, Yang Q, Yan L, Wan L, Zhang B, Wang C, Xia Y. Fluid shear stress promotes osteoblast proliferation through the NFATc1-ERK5 pathway. Connect Tissue Res 2019; 60:107-116. [PMID: 29609502 DOI: 10.1080/03008207.2018.1459588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Extracellular-regulated kinase 5 (ERK5) is thought to regulate osteoblast proliferation. To further understand how ERK5 signaling regulates osteoblast proliferation induced by fluid shear stress (FSS), we examined some potential signaling targets associated with ERK5 in MC3T3-E1 cells. METHODS MC3T3-E1 cells were treated with XMD8-92 (an ERK5 inhibitor) or Cyclosporin A (CsA, a nuclear factor of activated T cells (NFAT) c1 inhibitor) and/or exposed to 12 dyn/cm2 FSS. Phosphorylated-ERK5 (p-ERK5) and expression levels of NFATc1, ERK5, E2F2, and cyclin E1 were analyzed by western blot. The mRNA levels of genes associated with cell proliferation were analyzed by Polymerase Chain Reaction (PCR) array. Subcellular localization of p-ERK5 and NFATc1 were determined by immunofluorescence. Cell proliferation was evaluated by MTT assay. RESULTS NFATc1 expression was up-regulated by FSS. XMD8-92 only blocked ERK5 activation; however, CsA decreased NFATc1 and p-ERK5 levels, including after FSS stimulation. Exposure to NFATc1 inhibitor or ERK5 inhibitor resulted in decreased E2F2 and cyclin E1 expression and proliferation by proliferative MC3T3-E1 cells. Furthermore, immunofluorescence results illustrated that NFATc1 induced ERK5 phosphorylation, resulting in p-ERK5 translocation to the nucleus. CONCLUSIONS Our results reveal that NFATc1 acts as an intermediate to promote the phosphorylation of ERK5 induced by FSS. Moreover, activated NFATc1-ERK5 signaling up-regulates the expression of E2F2 and cyclin E1, which promote osteoblast proliferation.
Collapse
Affiliation(s)
- Ning Ding
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bin Geng
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Zhonghao Li
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Quanzeng Yang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Liang Yan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Lang Wan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bo Zhang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Cuifang Wang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Yayi Xia
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| |
Collapse
|
5
|
Watson JB, Bidgoli A, Johnston JM. B-precursor acute lymphoblastic leukemia complicating 18q deletion syndrome. Pediatr Blood Cancer 2018; 65:e27436. [PMID: 30188010 DOI: 10.1002/pbc.27436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 08/09/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Jordan B Watson
- Pediatric Hematology/Oncology, New York Medical College/Westchester Medical Center, Valhalla, New York
| | - Alan Bidgoli
- Department of Pediatrics, Memorial University Medical Center, Savannah, Georgia
| | - J Martin Johnston
- Hematology/Oncology, Department of Pediatrics, Renown Children's Hospital, University of Nevada, Reno, Nevada
| |
Collapse
|
6
|
Wu C, Liu X, Sun R, Qin Y, Liu Z, Yang S, Tang T, Zhu Z, Yu D, Liu F. Targeting Anion Exchange of Osteoclast, a New Strategy for Preventing Wear Particles Induced- Osteolysis. Front Pharmacol 2018; 9:1291. [PMID: 30459624 PMCID: PMC6232501 DOI: 10.3389/fphar.2018.01291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/22/2018] [Indexed: 12/30/2022] Open
Abstract
Joint replacement is essential for the treatment of serious joint disease. However, prosthetic failure remains an important clinical issue, with periprosthesis osteolysis (PO), caused by osteoclastic bone resorption induced by wear particles, being the leading cause of failure. Nuclear factor of activated T cells c1 (NFATc1) appears to play an important role in wear particle-induced osteoclastogenesis, with bicarbonate/chloride exchanger, solute carrier family 4, anion exchanger, member 2, (SLC4A2) being upregulated during osteoclastogenesis in an NFATc1-dependent manner. Anion exchange mediated by SLC4A2 in osteoclasts could affect the bone resorption activity by regulating pHi. This study investigated the role and mechanism of SLC4A2 in wear particle-induced osteoclast differentiation and function in vitro. The use of 4, 4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS), an anion exchange inhibitor, suppressed wear particle-induced PO in vivo. Furthermore, controlled release of DIDS from chitosan microspheres can strengthen the PO therapy effect. Therefore, anion exchange mediated by osteoclastic SLC4A2 may be a potential therapeutic target for the treatment of aseptic loosening of artificial joints.
Collapse
Affiliation(s)
- Chuanlong Wu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuqiang Liu
- Department of Orthopaedics, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Ruixin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunhao Qin
- Department of Orthopaedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqing Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenan Zhu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Degang Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengxiang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Li C, Zheng Z, Zhang X, Asatrian G, Chen E, Song R, Culiat C, Ting K, Soo C. Nfatc1 Is a Functional Transcriptional Factor Mediating Nell-1-Induced Runx3 Upregulation in Chondrocytes. Int J Mol Sci 2018; 19:ijms19010168. [PMID: 29316655 PMCID: PMC5796117 DOI: 10.3390/ijms19010168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 01/01/2023] Open
Abstract
Neural EGFL like 1 (Nell-1) is essential for chondrogenic differentiation, maturation, and regeneration. Our previous studies have demonstrated that Nell-1's pro-chondrogenic activities are predominantly reliant upon runt-related transcription factor 3 (Runx3)-mediated Indian hedgehog (Ihh) signaling. Here, we identify the nuclear factor of activated T-cells 1 (Nfatc1) as the key transcriptional factor mediating the Nell-1 → Runx3 signal transduction in chondrocytes. Using chromatin immunoprecipitation assay, we were able to determine that Nfatc1 binds to the -833--810 region of the Runx3-promoter in response to Nell-1 treatment. By revealing the Nell-1 → Nfatc1 → Runx3 → Ihh cascade, we demonstrate the involvement of Nfatc1, a nuclear factor of activated T-cells, in chondrogenesis, while providing innovative insights into developing a novel therapeutic strategy for cartilage regeneration and other chondrogenesis-related conditions.
Collapse
Affiliation(s)
- Chenshuang Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Greg Asatrian
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Richard Song
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Cymbeline Culiat
- NellOne Therapeutics, Inc., 99 Midway Ln # E, Oak Ridge, TN 37830, USA.
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery, the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
8
|
Cody JD, Sebold C, Heard P, Carter E, Soileau B, Hasi-Zogaj M, Hill A, Rupert D, Perry B, O'Donnell L, Gelfond J, Lancaster J, Fox PT, Hale DE. Consequences of chromsome18q deletions. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2015; 169:265-80. [PMID: 26235940 DOI: 10.1002/ajmg.c.31446] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 06/28/2015] [Indexed: 11/11/2022]
Abstract
Providing clinically relevant prognoses and treatment information for people with a chromsome18q deletion is particularly challenging because every unrelated person has a unique region of hemizygosity. The hemizygous region can involve almost any region of 18q including between 1 and 101 genes (30 Mb of DNA). Most individuals have terminal deletions, but in our cohort of over 350 individuals 23% have interstitial deletions. Because of this heterogeneity, we take a gene by gene approach to understanding the clinical consequences. There are 196 genes on 18q. We classified 133 of them as dosage insensitive, 15 (8%) as dosage sensitive leading to haploinsufficiency while another 10 (5%) have effects that are conditionally haploinsufficient and are dependent on another factor, genetic or environmental in order to cause an abnormal phenotype. Thirty-seven genes (19%) have insufficient information to classify their dosage effect. Phenotypes attributed to single genes include: congenital heart disease, minor bone morphology changes, central nervous system dysmyelination, expressive speech delay, vesicouretreral reflux, polyposis, Pitt-Hopkins syndrome, intellectual disability, executive function impairment, male infertility, aural atresia, and high frequency sensorineural hearing loss. Additionally, identified critical regions for other phenotypes include: adolescent idiopathic scoliosis and pectus excavatum, Virchow-Robin perivascular spaces, small corpus callosum, strabismus, atopic disorders, mood disorder, IgA deficiency, nystagmus, congenital heart disease, kidney malformation, vertical talus, CNS dysmyelination growth hormone deficiency and cleft palate. Together these findings make it increasingly feasible to compile an individualized syndrome description based on each person's individuated genotype. Future work will focus on understanding molecular mechanisms leading to treatment.
Collapse
|
9
|
Abkhezr M, Kim EY, Roshanravan H, Nikolos F, Thomas C, Hagmann H, Benzing T, Dryer SE. Pleiotropic signaling evoked by tumor necrosis factor in podocytes. Am J Physiol Renal Physiol 2015; 309:F98-108. [PMID: 26017975 DOI: 10.1152/ajprenal.00146.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/18/2015] [Indexed: 02/05/2023] Open
Abstract
TNF has been implicated in glomerular diseases, but its actions on podocytes are not well understood. Endogenous TNF expression is markedly increased in mouse podocytes exposed to sera from patients with recurrent focal segmental glomerulosclerosis, and TNF is able to increase its own expression in these cells. Exposure of podocytes to TNF increased phosphorylation of NF-κB p65-RelA followed by increased tyrosine phosphorylation of STAT3. STAT3 activation was blocked by the NF-κB inhibitor JSH-23 and by the STAT3 inhibitor stattic, whereas TNF-evoked NF-κB activation was not affected by stattic. TNF treatment increased nuclear accumulation of nuclear factor of activated T cells (NFAT)c1 in podocytes, a process that occurred downstream of STAT3 activation. TNF also increased expression of cyclin D1 but had no effect on cyclin-dependent kinase 4, p27(kip), or podocin. Despite its effects on cyclin D1, TNF treatment for up to 72 h did not cause podocytes to reenter the cell cycle. TNF increased total expression of transient receptor potential (TRP)C6 channels through a pathway dependent on NFATc1 and increased the steady-state expression of TRPC6 subunits on the podocyte cell surface. TNF effects on TRPC6 trafficking required ROS. Consistent with this, La(3+)-sensitive cationic currents activated by a diacylglycerol analog were increased in TNF-treated cells. The effects of TNF on NFATc1 and TRPC6 expression were blocked by cyclosporine A but were not blocked by the pan-TRP inhibitor SKF-96365. TNF therefore influences multiple pathways previously implicated in podocyte pathophysiology and is likely to sensitize these cells to other insults.
Collapse
Affiliation(s)
- Mousa Abkhezr
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Hila Roshanravan
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Fotis Nikolos
- Department of Biology and Biochemistry, University of Houston, Houston, Texas; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas; and
| | - Christoforos Thomas
- Department of Biology and Biochemistry, University of Houston, Houston, Texas; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas; and
| | - Henning Hagmann
- Department of Internal Medicine, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department of Internal Medicine, University of Cologne, Cologne, Germany
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas; Division of Nephrology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
10
|
Li S, Miller CH, Giannopoulou E, Hu X, Ivashkiv LB, Zhao B. RBP-J imposes a requirement for ITAM-mediated costimulation of osteoclastogenesis. J Clin Invest 2014; 124:5057-73. [PMID: 25329696 PMCID: PMC4347236 DOI: 10.1172/jci71882] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/11/2014] [Indexed: 01/07/2023] Open
Abstract
Osteoclastogenesis requires activation of RANK signaling as well as costimulatory signals from immunoreceptor tyrosine-based activation motif-containing (ITAM-containing) receptors/adaptors, predominantly tyrosine kinase-binding proteins DAP12 and FcRγ, in osteoclast precursors. It is not well understood how costimulatory signals are regulated and integrated with RANK signaling. Here, we found that osteopetrotic bone phenotypes in mice lacking DAP12 or DAP12 and FcRγ are mediated by the transcription factor RBP-J, as deletion of Rbpj in these mice substantially rescued the defects of bone remodeling. Using a TNF-α-induced model of inflammatory bone resorption, we determined that RBP-J deficiency enables TNF-α to induce osteoclast formation and bone resorption in DAP12-deficient animals. Thus, RBP-J imposes a requirement for ITAM-mediated costimulation of RANKL or TNF-α-induced osteoclastogenesis. Mechanistically, RBP-J suppressed induction of key osteoclastogenic factors NFATc1, BLIMP1, and c-FOS by inhibiting ITAM-mediated expression and function of PLCγ2 and activation of downstream calcium-CaMKK/PYK2 signaling. Moreover, RBP-J suppressed Plcg2 expression and downstream calcium oscillations indirectly by a TGF-β/PLCγ2/calcium axis. Together, our findings indicate that RBP-J suppresses ITAM-mediated costimulation, thereby limiting crosstalk between ITAM and RANK/TNFR signaling and allowing fine tuning of osteoclastogenesis during bone homeostasis and under inflammatory conditions. Furthermore, these data suggest that environmental cues that regulate RBP-J expression/function potentially modulate the requirement for costimulatory signaling for osteoclast differentiation and bone remodeling.
Collapse
Affiliation(s)
- Susan Li
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA. Biological Sciences Department, New York City College of Technology, City University of New York, New York, New York, USA. Department of Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Christine H. Miller
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA. Biological Sciences Department, New York City College of Technology, City University of New York, New York, New York, USA. Department of Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Eugenia Giannopoulou
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA. Biological Sciences Department, New York City College of Technology, City University of New York, New York, New York, USA. Department of Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Xiaoyu Hu
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA. Biological Sciences Department, New York City College of Technology, City University of New York, New York, New York, USA. Department of Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Lionel B. Ivashkiv
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA. Biological Sciences Department, New York City College of Technology, City University of New York, New York, New York, USA. Department of Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA. Biological Sciences Department, New York City College of Technology, City University of New York, New York, New York, USA. Department of Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| |
Collapse
|
11
|
Liu H, Zhao Q, Song Q, Zhou FH, Kang HJ, Pan L, Yao YM. Release of High Mobility Protein Box-1 is Greatly Regulated by Nuclear Factor of Activated T Cell-2 in Human Monocytes. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Close talk between inflammatory mediators and immunological cytokines has been discovered and reported. In this study, the role of nuclear factor of activated T cell-2 (NFAT2) in regulation of high mobility group box-1 (HMGB1) release was investigated. THP-1 cell and HEK293T cell were incubated and stimulated by lipopolysaccharide (LPS). Firstly, binding site between HMGB1 and NFAT2 was identified by co-immunoprecipitation (IP). Box A, Box B and CT domain of HMGB1 were constructed, as well as Rel-homology-domain (RHD), pre-RHD and pro-RHD of NFAT2. THP-1 cell was harvested, cell lysate and culture medium were collected at appointed times. Binding between HMGB1 and NFAT2 was measured, HMGB1 protein level in culture medium was analyzed at the same time. Secondly, the role of NFAT2 in regulating HMGB1 release was investigated. When THP-1 cell was cultured for 24 h, HMGB1 protein level was measured at appointed times with or without siRNA to inhibit NFAT2 expression. Our data show that HMGB1 bound to NFAT2 in THP-1 cell cytoplasm. Further experiments showed that box B domain of HMGB1 could bind to pre-RHD of NFAT2. After stimulation by LPS, interaction between HMGB1 and NFAT2 was discovered decreasing gradually. However, HMGB1 protein level increased in culture medium at the same time. Furthermore, HMGB1 release could be enhanced by NFAT2 inhibition. Taken together, release of HMGB1 could be regulated by NFAT2 in human monocytes.
Collapse
Affiliation(s)
- H. Liu
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Q. Zhao
- Gastroenterology Department of Nanlou, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Q. Song
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - F-H. Zhou
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - H-J. Kang
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - L. Pan
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Y-M. Yao
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital (formerly 304th Hospital), Beijing, People's Republic of China
| |
Collapse
|
12
|
Liu FX, Wu CL, Zhu ZA, Li MQ, Mao YQ, Liu M, Wang XQ, Yu DG, Tang TT. Calcineurin/NFAT pathway mediates wear particle-induced TNF-α release and osteoclastogenesis from mice bone marrow macrophages in vitro. Acta Pharmacol Sin 2013; 34:1457-66. [PMID: 24056707 DOI: 10.1038/aps.2013.99] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 07/08/2013] [Indexed: 12/29/2022] Open
Abstract
AIM To investigate the roles of the calcineurin/nuclear factor of activated T cells (NFAT) pathway in regulation of wear particles-induced cytokine release and osteoclastogenesis from mouse bone marrow macrophages in vitro. METHODS Osteoclasts were induced from mouse bone marrow macrophages (BMMs) in the presence of 100 ng/mL receptor activator of NF-κB ligand (RANKL). Acridine orange staining and MTT assay were used to detect the cell viability. Osteoclastogenesis was determined using TRAP staining and RT-PCR. Bone pit resorption assay was used to examine osteoclast phenotype. The expression and cellular localization of NFATc1 were examined using RT-PCR and immunofluorescent staining. The production of TNFα was analyzed with ELISA. RESULTS Titanium (Ti) or polymethylmethacrylate (PMMA) particles (0.1 mg/mL) did not significantly change the viability of BMMs, but twice increased the differentiation of BMMs into mature osteoclasts, and markedly increased TNF-α production. The TNF-α level in the PMMA group was significantly higher than in the Ti group (96 h). The expression of NFATc1 was found in BMMs in the presence of the wear particles and RANKL. In bone pit resorption assay, the wear particles significantly increased the resorption area and total number of resorption pits in BMMs-seeded ivory slices. Addition of 11R-VIVIT peptide (a specific inhibitor of calcineurin-mediated NFAT activation, 2.0 μmol/L) did not significantly affect the viability of BMMs, but abolished almost all the wear particle-induced alterations in BMMs. Furthermore, VIVIT reduced TNF-α production much more efficiently in the PMMA group than in the Ti group (96 h). CONCLUSION Calcineurin/NFAT pathway mediates wear particles-induced TNF-α release and osteoclastogenesis from BMMs. Blockade of this signaling pathway with VIVIT may provide a promising therapeutic modality for the treatment of periprosthetic osteolysis.
Collapse
|
13
|
Liu Z, Lee J, Krummey S, Lu W, Cai H, Lenardo MJ. The kinase LRRK2 is a regulator of the transcription factor NFAT that modulates the severity of inflammatory bowel disease. Nat Immunol 2011; 12:1063-70. [PMID: 21983832 PMCID: PMC4140245 DOI: 10.1038/ni.2113] [Citation(s) in RCA: 283] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 08/19/2011] [Indexed: 12/19/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been identified by genome-wide association studies as being encoded by a major susceptibility gene for Crohn's disease. Here we found that LRRK2 deficiency conferred enhanced susceptibility to experimental colitis in mice. Mechanistic studies showed that LRRK2 was a potent negative regulator of the transcription factor NFAT and was a component of a complex that included the large noncoding RNA NRON (an NFAT repressor). Furthermore, the risk-associated allele encoding LRRK2 Met2397 identified by a genome-wide association study for Crohn's disease resulted in less LRRK2 protein post-translationally. Severe colitis in LRRK2-deficient mice was associated with enhanced nuclear localization of NFAT1. Thus, our study defines a new step in the control of NFAT activation that involves an immunoregulatory function of LRRK2 and has important implications for inflammatory bowel disease.
Collapse
Affiliation(s)
- Zhihua Liu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinwoo Lee
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott Krummey
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Lu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaibin Cai
- Unit of Transgenesis; Laboratory of Neurogenetics; National Institute on Aging, National Institutes of Health; Bethesda, MD 20892 USA
| | - Michael J. Lenardo
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Malemud CJ. Suppression of Autoimmune Arthritis by Small Molecule Inhibitors of the JAK/STAT Pathway. Pharmaceuticals (Basel) 2010; 3:1446-1455. [PMID: 27713312 PMCID: PMC4033991 DOI: 10.3390/ph3051446] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 04/20/2010] [Accepted: 05/11/2010] [Indexed: 12/23/2022] Open
Abstract
A skewed ratio of pro-inflammatory to anti-inflammatory cytokines, elevated growth factor synthesis and T- and B-lymphocyte activation are 3 hallmarks of rheumatoid arthritis (RA) pathology. Interleukin-6 (IL-6), IL-7, IL-17, IL-12/IL-23 and growth factors, granulocyte macrophage-colony stimulating factor, IL-3, and erythropoietin activate the Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) pathway. Evidence showed that STAT protein phosphorylation (p-STAT) by activated JAKs is permissive for p-STAT to act as transcription factors by binding to STAT-responsive gene promoter sequences. This event is critical for perpetuating RA, in part, by up-regulating pro-inflammatory cytokine gene transcription. Activation of JAK/STAT by cytokines and growth factors can induce ‘cross-talk’ with other signaling pathways by which Stress-Activated Protein/Mitogen-Activated Protein Kinase (SAP/MAPK) and Phosphatidylinositide-3-Kinase (PI3K)-mediated signaling are also activated. JAK-specific small molecule inhibitors (SMIs) were developed to test whether JAK/STAT pathway blockade would regulate autoimmune-mediated inflammation. JAK-specific SMI blockade inhibited p-STAT induced by pro-inflammatory cytokines in vitro. Systemically administered JAK-specific SMI blockade also ameliorated biomarkers of inflammation in well-validated arthritis animal models. A few JAK-specific SMIs have made their way into RA clinical trials. In fact, the JAK3-specific SMI, CP-690,500 is the first JAK/STAT SMI to be assessed for clinical efficacy in a Phase III RA trial.
Collapse
Affiliation(s)
- Charles J Malemud
- Division of Rheumatic Diseases, Departments of Medicine & Anatomy, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|