1
|
Shin SY, Kang IS, Kim C. ERK inhibits osteoclast differentiation in RAW 264.7 cells through the osteoprotegerin-mediated autophagy. Bone 2025; 193:117424. [PMID: 39947572 DOI: 10.1016/j.bone.2025.117424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Osteoclasts (OCs) are bone-resorbing cells derived from the monocyte/macrophage lineage. The extracellular signal-regulated kinase (ERK) pathway controls cellular responses such as proliferation, differentiation, and survival, including those of OCs. In the present study, ERK inhibitors reduced the proliferation of bone marrow-derived macrophages (BMMs) and RAW 264.7 cells. However, ERK inhibitors decreased OC differentiation in BMMs but increased it in RAW 264.7 cells. ERK downregulation using small interfering RNA transfection also increased the OC differentiation and the expression of receptor activator of nuclear factor-κB, OC-specific markers, and OC-associated transcription factors in RAW 264.7 cells. These findings suggest ERK regulates OC differentiation in RAW 264.7 cells differently than in BMMs. Thus, we further investigated the mechanism by which ERK negatively regulates OC differentiation in RAW 264.7 cells. ERK inhibition decreased the expression of osteoprotegerin (OPG), a negative regulator of OC differentiation. OPG knockdown increased OC formation. ERK inhibitors activated the Akt/mammalian target of the rapamycin (mTOR) signaling pathway while inhibiting unc-51-like autophagy activating kinase 1 (ULK1). This resulted in decreased levels of microtubule-associated protein 1A/1B-light chain 3-II (LC3-II) and increased levels of p62, thereby reducing autophagy. In addition, OPG knockdown reduced autophagy by activating Akt/mTOR and inhibiting ULK1, resulting in decreased LC3-II and accumulated p62. Therefore, ERK inhibition promoted OC differentiation by downregulating OPG-mediated inhibition of osteoclastogenesis and autophagy in RAW 264.7 cells. These findings highlight ERK's complex role in OC differentiation and suggest that understanding ERK's dual impact on OC differentiation can provide insights into novel treatment strategies for bone-related disorders.
Collapse
Affiliation(s)
- Soo-Young Shin
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea; BK21, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - In-Soon Kang
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea; BK21, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
2
|
Modgil S, Walker CL, Chrenek MA, Grossniklaus HE, McDonald FE, Iuvone PM. Long-Term Functional Rescue of Trauma-Induced Vision Loss by a Novel, Small Molecule TrkB Activator. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638863. [PMID: 40027732 PMCID: PMC11870575 DOI: 10.1101/2025.02.18.638863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Brain-derived neurotrophic factor (BDNF) signaling through the tropomyosin-related kinase B (TrkB) receptor promotes neuronal growth and survival following an injury. However, its short half-life and pleiotropic effects limit the clinical use of BDNF as a therapy in neurodegenerative disorders. Identification of novel and selective TrkB activators may ameliorate the damage caused to retinal neurons during eye-related injuries, and may reduce adverse visual outcomes associated with visual trauma. We previously described a selective TrkB agonist, N-[2-(5-hydroxy-1H-indol-3-yl) ethyl]-2-oxopiperidine-3-carboxamide (HIOC), that reduces the decline in visual function in a mouse model of ocular trauma (1). Using the lead optimization approach, we subsequently synthesized a fluoropyridine analog of HIOC, 2-fluoro-N-(2-(5-hydroxy-1H-indol-3-yl) ethyl) nicotinamide (HIFN), which also successfully activates TrkB. HIFN is a more potent TrkB activator than the parent compound, HIOC. Further, treatment with HIFN demonstrated neuroprotection in an animal model of overpressure ocular blast injury, ameliorating blast-related visual functional decline. Mice treated with HIFN had better visual acuity, contrast sensitivity, and retinal function supported by enhanced survival of retinal ganglion cells compared to vehicle-treated animals. Moreover, HIFN exhibited better protective effects than HIOC. The therapeutic effects of HIFN were attributed to TrkB activation, as blocking the receptor with a selective receptor antagonist (ANA-12) abrogated the neuroprotection. Together, our results identify HIFN, a novel TrkB receptor activator, as a strategy for decreasing retinal degeneration and progressive vision loss associated with traumatic ocular injury. In addition, this compound may have broader applications treating other diseases with altered TrkB activity.
Collapse
Affiliation(s)
- Shweta Modgil
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA 30322
| | | | - Micah A. Chrenek
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA 30322
| | - Hans E. Grossniklaus
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA 30322
| | | | - P. Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
3
|
Ayyasamy R, Fan S, Czernik P, Lecka-Czernik B, Chattopadhyay S, Chakravarti R. 14-3-3ζ suppresses RANKL signaling by destabilizing TRAF6. J Biol Chem 2024; 300:107487. [PMID: 38908751 PMCID: PMC11331427 DOI: 10.1016/j.jbc.2024.107487] [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: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/24/2024] Open
Abstract
Macrophages are essential regulators of inflammation and bone loss. Receptor activator of nuclear factor-κβ ligand (RANKL), a pro-inflammatory cytokine, is responsible for macrophage differentiation to osteoclasts and bone loss. We recently showed that 14-3-3ζ-knockout (YwhazKO) rats exhibit increased bone loss in the inflammatory arthritis model. 14-3-3ζ is a cytosolic adaptor protein that actively participates in many signaling transductions. However, the role of 14-3-3ζ in RANKL signaling or bone remodeling is unknown. We investigated how 14-3-3ζ affects osteoclast activity by evaluating its role in RANKL signaling. We utilized 14-3-3ζ-deficient primary bone marrow-derived macrophages obtained from wildtype and YwhazKO animals and RAW264.7 cells generated using CRISPR-Cas9. Our results showed that 14-3-3ζ-deficient macrophages, upon RANKL stimulation, have bigger and stronger tartrate-resistant acid phosphatase-positive multinucleated cells and increased bone resorption activity. The presence of 14-3-3ζ suppressed RANKL-induced MAPK and AKT phosphorylation, transcription factors (NFATC1 and p65) nuclear translocation, and subsequently, gene induction (Rank, Acp5, and Ctsk). Mechanistically, 14-3-3ζ interacts with TRAF6, an essential component of the RANKL receptor complex. Upon RANKL stimulation, 14-3-3ζ-TRAF6 interaction was increased, while RANK-TRAF6 interaction was decreased. Importantly, 14-3-3ζ supported TRAF6 ubiquitination and degradation by the proteasomal pathway, thus dampening the downstream RANKL signaling. Together, we show that 14-3-3ζ regulates TRAF6 levels to suppress inflammatory RANKL signaling and osteoclast activity. To the best of our knowledge, this is the first report on 14-3-3ζ regulation of RANKL signaling and osteoclast activation.
Collapse
Affiliation(s)
- R Ayyasamy
- Department of Physiology & Pharmacology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - S Fan
- Department of Medical Microbiology & Immunology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - P Czernik
- Department of Orthopedics, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - B Lecka-Czernik
- Department of Orthopedics, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - S Chattopadhyay
- Department of Medical Microbiology & Immunology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA; Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - R Chakravarti
- Department of Physiology & Pharmacology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA.
| |
Collapse
|
4
|
Tenshin H, Delgado-Calle J, Windle JJ, Roodman GD, Chirgwin JM, Kurihara N. Osteocytes and Paget's Disease of Bone. Curr Osteoporos Rep 2024; 22:266-272. [PMID: 38457001 PMCID: PMC11060996 DOI: 10.1007/s11914-024-00863-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW To describe the contributions of osteocytes to the lesions in Paget's disease, which are characterized by locally overactive bone resorption and formation. RECENT FINDINGS Osteocytes, the most abundant cells in bone, are altered in Paget's disease lesions, displaying increased size, decreased canalicular length, incomplete differentiation, and less sclerostin expression compared to controls in both patients and mouse models. Pagetic lesions show increased senescent osteocytes that express RANK ligand, which drives osteoclastic bone resorption. Abnormal osteoclasts in Paget's disease secrete abundant IGF1, which enhances osteocyte senescence, contributing to lesion formation. Recent data suggest that osteocytes contribute to lesion formation in Paget's disease by responding to high local IGF1 released from abnormal osteoclasts. Here we describe the characteristics of osteocytes in Paget's disease and their role in bone lesion formation based on recent results with mouse models and supported by patient data.
Collapse
Affiliation(s)
- Hirofumi Tenshin
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jesus Delgado-Calle
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - G David Roodman
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - John M Chirgwin
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
- Research Service, Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Noriyoshi Kurihara
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
5
|
Chen X, Ye F, He H, Chen G, Chen Z, Ye E, He B, Yang Y, Zhang J. Denosumab Induces Neoplastic Stromal Cell Apoptosis Via p62 Downregulation Dependent on Autophagy Pathway in Giant Cell Tumour of Bone. Curr Cancer Drug Targets 2024; 24:565-578. [PMID: 37961860 DOI: 10.2174/0115680096265253231022185008] [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: 06/02/2023] [Revised: 07/13/2023] [Accepted: 08/30/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND As the only humanized monoclonal antibody against receptor activator of nuclear factor-κB ligand (RANKL) for giant cell tumour of bone (GCTB) therapy, denosumab has limited antitumour effect on neoplastic stromal cells. Nevertheless, its mechanism of action has not yet been clarified. A previous study has revealed that p62 may play an important role in the antitumour activity of denosumab. OBJECTIVE The study aimed to investigate if the mechanism by which denosumab inhibits GCTB neoplastic stromal cells growth is via p62 modulation and other related mechanisms. METHODS p62 expression before and after denosumab therapy was analysed by RT‒qPCR, western blot, ELISA, and immunohistochemical assays. Two primary neoplastic stromal cells were isolated from fresh GCTB tumour tissue (L cell) and metastatic tissue (M cell). Cell proliferation, migration, apoptosis, and autophagy were investigated in p62 knockdown neoplastic stromal cells transfected by short hairpin RNA lentivirus in vitro. Tumor growth was evaluated in the chick chorioallantoic membrane model in vivo. RESULTS p62 expression was found to be downregulated following denosumab therapy. The patients with a decrease in p62 expression had lower recurrence-free survival rates. The proliferation of M cells was not inhibited by denosumab therapy, but it was restored by p62 knockdown. Moreover, p62 knockdown inhibited tumour growth in vivo. Denosumab induced M cell apoptosis and arrested the cell cycle at the G1/G0 transition and these effects were also enhanced by p62 knockdown. Autophagic flux assays revealed p62 modulation to be dependent on autophagy following denosumab incubation. CONCLUSION Denosumab induced neoplastic stromal cells apoptosis via p62 downregulation dependent on autophagy pathway. The combination of p62 and RANKL knockdown might be a better strategy than RANKL knockdown alone for GCTB targeted therapy.
Collapse
Affiliation(s)
- Xianwei Chen
- Department of Orthopaedics, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Fan Ye
- Department of Orthopaedics, The First Affiliated Hospital of Nanyang Medical College, Nangyang, Henan, 473000, China
| | - Hao He
- Department of Orthopaedics, The People's Hospital of Guang'an, Guang'an, Sichuan, 638000, China
| | - Gong Chen
- Department of Orthopaedics, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Zhifu Chen
- Department of Orthopaedics, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - En Ye
- Department of Pathology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Bingjan He
- Department of Orthopaedics, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Yuqi Yang
- Department of Orthopaedics, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Jing Zhang
- Department of Orthopaedics, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| |
Collapse
|
6
|
Budnik A, Palewski M, Michnowska-Kobylińska M, Lisowski Ł, Łapińska M, Stachurska Z, Szpakowicz A, Konstantynowicz J, Kamiński K, Konopińska J. The prevalence of age-related macular degeneration and osteoporosis in the older Polish population: Is there a link? PLoS One 2023; 18:e0293143. [PMID: 37856460 PMCID: PMC10586687 DOI: 10.1371/journal.pone.0293143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Age-related macular degeneration is the primary cause of irreversible blindness in developed countries, whereas the global prevalence of osteoporosis-a major public health problem-is 19.7%. Both diseases may coincide in populations aged >50 years, leading to serious health deterioration and decreased quality of life. OBJECTIVES This study aimed to analyze the relationship between age-related macular degeneration and osteopenia, defined as decreased bone mineral density, in the Polish population. METHODS Participants were derived from the population-based Bialystok PLUS Study. Randomized individuals were stratified into two groups, those with age-related macular degeneration (AMD-1 group) or without age-related macular degeneration (AMD-0 group). Using a cutoff value of -1.0 to identify low bone mass, participants with femoral bone mineral density T-scores above -1.0 were assigned to the normal reference, and those with T-scores below -1.0 were assigned to the osteopenia category. Among 436 Caucasian participants aged 50-80 years (252 women, 184 men), the prevalence of age-related macular degeneration was 9.9% in women and 12.0% in men. Decreased bone mineral density based on T-scores was observed in 36.9% of women and in 18.9% of men. Significant differences in femoral bone mineral density between the AMD-0 and AMD-1 groups were detected only in men (mean difference [95% confidence interval] = 0.11 (0.02; 0.13); p = 0.012 for femoral bone mineral density, and 0.73 [0.015; 0.94]; p = 0.011 for the femoral T-score). No associations were observed between bone mineral density and age-related macular degeneration in women. CONCLUSION Decreased femoral bone mineral density may be associated with a higher risk of age-related macular degeneration in men, but a causal link remains unclear.
Collapse
Affiliation(s)
- Agnieszka Budnik
- Department of Ophthalmology, Medical University of Białystok, Białystok, Poland
| | - Marcin Palewski
- Department of Ophthalmology, Medical University of Białystok, Białystok, Poland
| | | | - Łukasz Lisowski
- Department of Ophthalmology, Medical University of Białystok, Białystok, Poland
| | - Magda Łapińska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Zofia Stachurska
- Population Research Centre, Medical University of Białystok, Białystok, Poland
| | - Anna Szpakowicz
- Department of Cardiology, Medical University of Białystok, Białystok, Poland
| | - Jerzy Konstantynowicz
- Department of Pediatrics, Rheumatology, Immunology and Metabolic Bone Diseases, Medical University of Białystok, Białystok, Poland
| | - Karol Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Joanna Konopińska
- Department of Ophthalmology, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
7
|
Arnst J, Jing Z, Cohen C, Ha SW, Viggeswarapu M, Beck GR. Bioactive silica nanoparticles target autophagy, NF-κB, and MAPK pathways to inhibit osteoclastogenesis. Biomaterials 2023; 301:122238. [PMID: 37441901 PMCID: PMC10530178 DOI: 10.1016/j.biomaterials.2023.122238] [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: 11/03/2022] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023]
Abstract
Spherical 50 nm silica-based nanoparticles (SiNPs) promote healthy bone homeostasis and maintenance by supporting bone forming osteoblast lineage cells while simultaneously inhibiting the differentiation of bone resorbing osteoclasts. Previous work demonstrated that an intraperitoneal injection of SiNPs in healthy mice - both young and old - increased bone density and quality, suggesting the possibility that SiNPs represent a dual action therapeutic. However, the underlying mechanisms governing the osteoclast response to SiNPs have yet to be fully explored and defined. Therefore, the goals of this study were to investigate the cellular and molecular mechanisms by which SiNPs inhibit osteoclastogenesis. SiNPs strongly inhibited RANKL-induced osteoclast differentiation within the first hours and concomitantly inhibited early transcriptional regulators such as Nfatc1. SiNPs simultaneously stimulated expression of autophagy related genes p62 and LC3β dependent on ERK1/2 signaling pathway. Intriguingly, SiNPs were found to stimulate autophagosome formation while inhibiting the autophagic flux necessary for RANKL-stimulated osteoclast differentiation, resulting in the inhibition of both the canonical and non-canonical NF-κB signaling pathways and stabilizing TRAF3. These results suggest a model in which SiNPs inhibit osteoclastogenesis by inhibiting the autophagic machinery and RANKL-dependent functionality. This mechanism of action defines a novel therapeutic strategy for inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Jamie Arnst
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Zhaocheng Jing
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA; The Second Hospital of Shandong University, Department of Orthopedics, Jinan, Shandong, 250033, China
| | - Cameron Cohen
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Shin-Woo Ha
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Manjula Viggeswarapu
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - George R Beck
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, 30033, USA; Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA; The Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Agas D, Marchegiani A, Laus F, Gabai V, Sufianov AA, Shneider A, Sabbieti MG. p62/SQSTM1 indirectly mediates remote multipotent mesenchymal cells and rescues bone loss and bone marrow integrity in ovariectomized rats. J Cell Physiol 2023; 238:407-419. [PMID: 36565474 DOI: 10.1002/jcp.30937] [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: 07/30/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Intramuscular administration of p62/SQSTM1 (sequestosome1)-encoding plasmid demonstrated an anticancer effect in rodent models and dogs as well as a high safety profile and the first evidence of clinical benefits in humans. Also, an anti-inflammatory effect of the plasmid was reported in several rodent disease models. Yet, the mechanisms of action for the p62 plasmid remain unknown. Here, we tested a hypothesis that the p62-plasmid can act through the modulation of bone marrow multipotent mesenchymal cells (MSCs). We demonstrated that a p62 plasmid can affect MSCs indirectly by stimulating p62-transfected cells to secrete an active ingredient(s) sensed by untransfected MSCs. When we transfected MSCs with the p62-plasmid, collected their supernatant, and added it to an untransfected MSCs culture, it switched the differentiation state and prompt osteogenic responses of the untransfected MSCs. According to an accepted viewpoint, ovariectomy leads to bone pathology via dysregulation of MSCs, and restoring the MSC homeostasis would restore ovariectomy-induced bone damage. To validate our in vitro observations in a clinically relevant in vivo model, we administered the p62 plasmid to ovariectomized rats. It partially reversed bone loss and notably reduced adipogenesis with concurrent reestablishing of the MSC subpopulation pool within the bone marrow. Overall, our study suggests that remote modulation of progenitor MSCs via administering a p62-encoding plasmid may constitute a mechanism for its previously reported effects and presents a feasible disease-preventing and/or therapeutic strategy.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | - Fulvio Laus
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | | | - Albert A Sufianov
- Federal Center of Neurosurgery, Tyumen, Russian Federation.,Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Alexander Shneider
- CureLab Oncology Inc., Dedham, Massachusetts, USA.,Department of Molecular Biology, Ariel University, Ariel, Israel
| | | |
Collapse
|
9
|
Yang J, Tong T, Zhu C, Zhou M, Jiang Y, Chen H, Que L, Liu L, Zhu G, Ha T, Chen Q, Li C, Xu Y, Li J, Li Y. Peli1 contributes to myocardial ischemia/reperfusion injury by impairing autophagy flux via its E3 ligase mediated ubiquitination of P62. J Mol Cell Cardiol 2022; 173:30-46. [PMID: 36179399 DOI: 10.1016/j.yjmcc.2022.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 01/18/2023]
Abstract
Autophagy flux is impaired during myocardial ischemia/reperfusion (M-I/R) via the accumulation of autophagosome and insufficient clearance, which exacerbates cardiomyocyte death. Peli1 (Pellion1) is a RING finger domain-containing ubiquitin E3 ligase that could catalyze the polyubiquitination of substrate proteins. Peli1 has been demonstrated to play an important role in ischemic cardiac diseases. However, little is known about whether Peli1 is involved in the regulation of autophagy flux during M-I/R. The present study investigated whether M-I/R induced impaired autophagy flux could be mediated through Peli1 dependent mechanisms. We induced M-I/R injury in wild type (WT) and Peli1 knockout mice and observed that M-I/R significantly decreased cardiac function that was associated with increased cardiac Peli1 expression and upregulated autophagy-associated protein LC3II and P62. In contrast, Peli1 knockout mice exhibited significant improvement of M-I/R induced cardiac dysfunction and decreased LC3II and P62 expression. Besides, inhibitors of autophagy also increased the infarct size in Peli1 knockout mice after 24 h of reperfusion. Mechanistic studies demonstrated that in vivo I/R or in vitro hypoxia/reoxygenation (H/R) markedly increased the Peli1 E3 ligase activity which directly promoted the ubiquitination of P62 at lysine(K)7 via K63-linkage to inhibit its dimerization and autophagic degradation. Co-immunoprecipitation and GST-pull down assay indicated that Peli1 interacted with P62 via the Ring domain. In addition, Peli1 deficiency also decreased cardiomyocyte apoptosis. Together, our work demonstrated a critical link between increased expression and activity of Peli1 and autophagy flux blockage in M-I/R injury, providing insight into a promising strategy for treating myocardium M-I/R injury.
Collapse
Affiliation(s)
- Jie Yang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tingting Tong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Chenghao Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Miao Zhou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yuqing Jiang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Hao Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Pathology, Wannan Medical College, Wuhu 241002, Anhui, China
| | - Linli Que
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Li Liu
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Guoqing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tuanzhu Ha
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN 37614-0575, USA
| | - Qi Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN 37614-0575, USA
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute of Biomedical Research, Liaocheng University, Liaocheng 252000, Shandong, China
| | - Jiantao Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
10
|
Luo ML, Huang W, Zhu HP, Peng C, Zhao Q, Han B. Advances in indole-containing alkaloids as potential anticancer agents by regulating autophagy. Biomed Pharmacother 2022; 149:112827. [PMID: 35316753 DOI: 10.1016/j.biopha.2022.112827] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer is a leading cause of death worldwide, and cancer development is often associated with disturbances in the autophagy process. Autophagy is a catabolic process involved in many physiological processes, crucial for cell growth and survival. It is an intracellular lysosomal/vacuolar degradation system. In this system, inner cytoplasmic cell membrane is degraded by lysosomal hydrolases, and the products are released back into the cytoplasm. Indole alkaloids are natural products extensively found in nature and have been proven to possess various pharmacological activities. In recent years, pharmacological studies have demonstrated another potential of indole alkaloids, autophagy regulation. The regulation may contribute to the efficacy of indole alkaloids in preventing and treating cancer. This review summarizes the current understanding of indole alkaloids' effect on tumor cells and autophagy. Then, we focus on mechanisms by which indole alkaloids can target the autophagy process associated with cancer, including the PI3K/Akt/mTOR signaling pathway, MAPK signaling pathway, ROS signaling pathway, Beclin-1, and so on. Literature has been surveyed primarily from 2009 to Nov. 2021, and some semisynthetic or fully synthetic indole derivatives are also discussed.
Collapse
Affiliation(s)
- Meng-Lan Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Keller C, Yorgan TA, Rading S, Schinke T, Karsak M. Impact of the Endocannabinoid System on Bone Formation and Remodeling in p62 KO Mice. Front Pharmacol 2022; 13:858215. [PMID: 35392569 PMCID: PMC8980328 DOI: 10.3389/fphar.2022.858215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/28/2022] [Indexed: 11/15/2022] Open
Abstract
Several studies have shown that the G-protein coupled cannabinoid receptor CB2 and its interaction partner p62 are molecularly involved in bone remodeling processes. Pharmacological activation of the CB2 receptor enhanced bone volume in postmenopausal osteoporosis and arthritis models in rodents, whereas knockout or mutation of the p62 protein in aged mice led to Paget’s disease of bone-like conditions. Studies of pharmacological CB2 agonist effects on bone metabolism in p62 KO mice have not been performed to date. Here, we assessed the effect of the CB2-specific agonist JWH133 after a short-term (5 days in 3-month-old mice) or long-term (4 weeks in 6-month-old mice) treatment on structural, dynamic, and cellular bone morphometry obtained by μCT of the femur and histomorphometry of the vertebral bodies in p62 KO mice and their WT littermates in vivo. A genotype-independent stimulatory effect of CB2 on bone formation, trabecular number, and trabecular thickness after short-term treatment and on tissue mineral density after long-term treatment was detected, indicating a weak osteoanabolic function of this CB2 agonist. Moreover, after short-term systemic CB2 receptor activation, we found significant differences at the cellular level in the number of osteoblasts and osteoclasts only in p62 KO mice, together with a weak increase in trabecular number and a decrease in trabecular separation. Long-term treatment showed an opposite JWH133 effect on osteoclasts in WT versus p62 KO animals and decreased cortical thickness only in treated p62 KO mice. Our results provide new insights into CB2 receptor signaling in vivo and suggest that CB2 agonist activity may be regulated by the presence of its macromolecular binding partner p62.
Collapse
Affiliation(s)
- Christina Keller
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Rading
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meliha Karsak
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Tong X, Yu G, Liu Q, Zhang X, Bian J, Liu Z, Gu J. Puerarin alleviates cadmium-induced oxidative damage to bone by reducing autophagy in rats. ENVIRONMENTAL TOXICOLOGY 2022; 37:720-729. [PMID: 34897960 DOI: 10.1002/tox.23437] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/28/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
Autophagy is a regulatory mechanism involved in cadmium (Cd)-induced bone toxicity and is suppressed by various stimuli, including oxidative stress. Puerarin is an isoflavonoid compound isolated from Pueraria, a plant used in traditional Chinese medicine. The underlying mechanisms of action of puerarin remain unclear. The objective of this study was to explore the mitigating effects of puerarin on cadmium-induced oxidative damage in the bones of rats. Cadmium exposure increased oxidative damage in rat bones; this was markedly decreased by puerarin treatment, as demonstrated by changes in the activity of antioxidative enzymes. Cadmium-induced blockage of the expression of key bone regulatory proteins, autophagy-related markers, and signaling molecules was also alleviated by puerarin treatment. Additionally, cadmium reduced expression of the autophagic protein Rab7 and of late endosomal/lysosomal adaptor and MAPK and mTOR activator 1 (LAMTOR1); the decrease in these proteins was not restored by puerarin treatment. We speculate that puerarin relieves the inhibition of fusion of autophagosomes with lysosomes that is induced by cadmium; however, this specific effect of puerarin and downstream effects on bone regulatory mechanisms require further investigation. In conclusion, puerarin alleviates cadmium-induced oxidative damage in the bones of rats by attenuating autophagy, which is likely associated with the antioxidant activity of puerarin.
Collapse
Affiliation(s)
- Xishuai Tong
- Institutes of Agricultural Science and Technology Development, Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu, P. R. China
| | - Gengsheng Yu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
| | - Qingyang Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
| | - Xueqing Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu, P. R. China
| | - Zongping Liu
- Institutes of Agricultural Science and Technology Development, Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu, P. R. China
| | - Jianhong Gu
- Institutes of Agricultural Science and Technology Development, Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu, P. R. China
| |
Collapse
|
13
|
Xu S, Li S, Liu X, Tan K, Zhang J, Li K, Bai X, Zhang Y. Rictor Is a Novel Regulator of TRAF6/TRAF3 in Osteoclasts. J Bone Miner Res 2021; 36:2053-2064. [PMID: 34155681 DOI: 10.1002/jbmr.4398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/27/2022]
Abstract
Tumor necrosis factor receptor-associated factors (TRAFs) are crucial for receptor activator of nuclear factor-κB (RANK) activation in osteoclasts. However, the upstream mechanisms of TRAF members in the osteoclastic lineage remain largely unknown. Here, we demonstrated that Rictor, a key component of mechanistic target of rapamycin complex 2 (mTORC2), was crucial for TRAF6/TRAF3 expression in osteoclasts. Our ex vivo and in vivo studies showed that Rictor ablation from the osteoclastic lineage reduced osteoclast numbers and increased bone mass in mice. Mechanistically, we found that Rictor ablation restricted osteoclast formation, which disrupted TRAF6 stability and caused autophagy block in a manner distinct from mTORC1, resulting in reduced TRAF3 degradation. Boosting TRAF6 expression or knockdown of TRAF3 levels in Rictor-deficient cells could both overcome the defect. Moreover, Rictor could interact with TRAF6 upon RANK ligand (RANKL) stimulation and loss of Rictor impaired TRAF6 stability and promoted its ubiquitinated degradation. These findings established an innovative link between Rictor, TRAF protein levels, and autophagic block. More importantly, mTOR complexes in the osteoclastic lineage are likely switches for coordinating TRAF6 and TRAF3 protein levels, and Rictor may function as an essential upstream regulator of TRAF6/TRAF3 that is partially independent of mTORC1 activity. Inhibitors targeting Rictor may therefore be valuable for preventing or treating osteoclast-related diseases. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Song Xu
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China.,Department of Arthroplasty, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shihai Li
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Xianming Liu
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Kang Tan
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Jiahuan Zhang
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Kai Li
- Academy of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Yue Zhang
- Department of Cell Biology, School of Basic Medical Science, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Gong L, Wang K, Wang M, Hu R, Li H, Gao D, Lin M. CUL5-ASB6 Complex Promotes p62/SQSTM1 Ubiquitination and Degradation to Regulate Cell Proliferation and Autophagy. Front Cell Dev Biol 2021; 9:684885. [PMID: 34164402 PMCID: PMC8215545 DOI: 10.3389/fcell.2021.684885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/06/2021] [Indexed: 12/30/2022] Open
Abstract
p62/SQSTM1 (sequestosome-1) is a key protein involved in multiple cellular bioprocesses including autophagy, nutrient sensing, cell growth, cell death, and survival. Therefore, it is implicated in human diseases such as obesity and cancer. Here, we show that the CUL5–ASB6 complex is a ubiquitin E3 ligase complex mediating p62 ubiquitination and degradation. Depletion of CUL5 or ASB6 induced p62 accumulation, and overexpression of ASB6 promoted ubiquitination and degradation of p62. Functionally, ASB6 overexpression can inhibit the proliferation of MEF and hepatocellular carcinoma cells by reducing p62 protein level, and impair the occurrence of autophagy. Overall, our study identified a new molecular mechanism regulating p62 stability, which may provide additional insights for understanding the delicate control of p62 and cell proliferation–autophagy control in physiological and pathological settings.
Collapse
Affiliation(s)
- Liyan Gong
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kaihua Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mengcheng Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ronggui Hu
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Huaguang Li
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Daming Gao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Moubin Lin
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Jia ZK, Fu CX, Wang AL, Yao K, Chen XJ. Cataract-causing allele in CRYAA (Y118D) proceeds through endoplasmic reticulum stress in mouse model. Zool Res 2021; 42:300-309. [PMID: 33929105 PMCID: PMC8175955 DOI: 10.24272/j.issn.2095-8137.2020.354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
As small heat shock proteins, α-crystallins function as molecular chaperones and inhibit the misfolding and aggregation of β/γ-crystallins. Genetic mutations of CRYAA are associated with protein aggregation and cataract occurrence. One possible process underlying cataract formation is that endoplasmic reticulum stress (ERS) induces the unfolded protein response (UPR), leading to apoptosis. However, the pathogenic mechanism related to this remains unexplored. Here, we successfully constructed a cataract-causing CRYAA (Y118D) mutant mouse model, in which the lenses of the CRYAA-Y118D mutant mice showed severe posterior rupture, abnormal morphological changes, and aberrant arrangement of crystallin fibers. Histological analysis was consistent with the clinical pathological characteristics. We also explored the pathogenic factors involved in cataract development through transcriptome analysis. In addition, based on key pathway analysis, up-regulated genes in CRYAA-Y118D mutant mice were implicated in the ERS-UPR pathway. This study showed that prolonged activation of the UPR pathway and severe stress response can cause proteotoxic and ERS-induced cell death in CRYAA-Y118D mutant mice.
Collapse
Affiliation(s)
- Zhe-Kun Jia
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Chen-Xi Fu
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Ai-Ling Wang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Ke Yao
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China. E-mail:
| | - Xiang-Jun Chen
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China. E-mail:
| |
Collapse
|
16
|
Oh SY, Kang N, Kang JY, Kim KW, Choi JH, Yang YM, Shin DM. Sestrin2 Regulates Osteoclastogenesis via the p62-TRAF6 Interaction. Front Cell Dev Biol 2021; 9:646803. [PMID: 33842470 PMCID: PMC8033026 DOI: 10.3389/fcell.2021.646803] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/09/2021] [Indexed: 12/26/2022] Open
Abstract
The receptor activator of nuclear factor-kappa B ligand (RANKL) mediates osteoclast differentiation and functions by inducing Ca2+ oscillations, activating mitogen-activated protein kinases (MAPKs), and activating nuclear factor of activated T-cells type c1 (NFATc1) via the RANK and tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) interaction. Reactive oxygen species (ROS) also plays an important role during osteoclastogenesis and Sestrin2, an antioxidant, maintains cellular homeostasis upon stress injury via regulation of ROS, autophagy, and inflammation. However, the role of Sestrin2 in osteoclastogenesis remains unknown. In this study, we investigated the role of Sestrin2 in the RANKL-RANK-TRAF6 signaling pathway during osteoclast differentiation. Deletion of Sestrin2 (Sesn2) increased bone mass and reduced the number of multinucleated osteoclasts on bone surfaces. RANKL-induced osteoclast differentiation and function decreased in Sesn2 knockout (KO) bone marrow-derived monocytes/macrophages (BMMs) due to inhibition of NFATc1 expression, but osteoblastogenesis was not affected. mRNA expression of RANKL-induced specific osteoclastogenic genes and MAPK protein expression were lower in Sesn2 KO BMMs than wild-type (WT) BMMs after RANKL treatment. However, the Sesn2 deletion did not affect ROS generation or intracellular Ca2+ oscillations during osteoclastogenesis. In contrast, the interaction between TRAF6 and p62 was reduced during osteoclasts differentiation in Sesn2 KO BMMs. The reduction in the TRAF6/p62 interaction and TRAP activity in osteoclastogenesis in Sesn2 KO BMMs was recovered to the WT level upon expression of Flag-Sesn2 in Sesn2 KO BMMs. These results suggest that Sestrin2 has a novel role in bone homeostasis and osteoclasts differentiation through regulation of NFATc1 and the TRAF6/p62 interaction.
Collapse
Affiliation(s)
- Sue Young Oh
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Namju Kang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.,BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jung Yun Kang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.,BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, South Korea.,Department of Dental Hygiene, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Ki Woo Kim
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jong-Hoon Choi
- Department of Orofacial Pain & Oral Medicine, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yu-Mi Yang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Dong Min Shin
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.,BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, South Korea
| |
Collapse
|
17
|
Florencio-Silva R, Sasso GRDS, Sasso-Cerri E, Simões MDJ, Cerri PS. Immunoexpression pattern of autophagy mediators in alveolar bone osteoclasts following estrogen withdrawal in female rats. J Mol Histol 2021; 52:321-333. [PMID: 33409945 DOI: 10.1007/s10735-020-09953-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 12/28/2020] [Indexed: 12/30/2022]
Abstract
It is known that estrogen deficiency increases osteoclast formation and activity. Autophagy, a cell survival pathway, has been shown to be crucial for osteoclast function. However, little is known about the effects of estrogen depletion on osteoclast autophagy. Here, we evaluated the effects of estrogen deficiency in the immunoexpression of autophagy mediators in alveolar bone osteoclasts of ovariectomized rats. Twelve adult female rats were ovariectomized (OVX-group) or SHAM-operated (SHAM-group). After three weeks, the rats were euthanized and maxillary fragments containing alveolar bone of the first molars were processed for light microscopy or transmission electron microscopy (TEM). Paraffin-sections were subjected to the TRAP method (osteoclast marker) or to the immunohistochemical detections of beclin-1, LC3α, and p62 (autophagy mediators); araldite-sections were processed for TEM. The number of TRAP-positive osteoclasts and the number of immunolabeled-multinucleated cells (MNCs) along the alveolar bone surface of the first molar were computed. The number of TRAP-positive osteoclasts and the number of beclin-1-, LC3α- and p62-immunolabelled osteoclasts were significantly higher in OVX-group than the SHAM-group. MNCs were frequently located juxtaposed to Howship lacunae along the alveolar bone surface, indicating that these cells are osteoclasts. TEM revealed osteoclasts exhibiting autophagosomes. Our data indicate that autophagy plays an important role during estrogen deficiency-induced osteoclastogenesis. Thus, our results contribute to a better understanding on the role of autophagy on osteoclasts under estrogenic deficiency, and reinforce the idea that modulation of autophagy may be a useful tool to inhibit excessive oral bone resorption in post-menopausal women.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina - EPM, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brasil.
| | - Gisela Rodrigues da Silva Sasso
- Departamento de Ginecologia, Escola Paulista de Medicina - EPM, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brasil
| | - Estela Sasso-Cerri
- Araraquara - Laboratory of Histology and Embryology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brasil
| | - Manuel de Jesus Simões
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina - EPM, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brasil
| | - Paulo Sérgio Cerri
- Araraquara - Laboratory of Histology and Embryology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brasil
| |
Collapse
|
18
|
Usategui-Martín R, Lendinez-Tortajada V, Pérez-Castrillón JL, Briongos-Figuero L, Abadía-Otero J, Martín-Vallejo J, Lara-Hernandez F, Chaves FJ, García-Garcia AB, Martín-Escudero JC. Polymorphisms in genes involved in inflammation, the NF-kB pathway and the renin-angiotensin-aldosterone system are associated with the risk of osteoporotic fracture. The Hortega Follow-up Study. Bone 2020; 138:115477. [PMID: 32535289 DOI: 10.1016/j.bone.2020.115477] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023]
Abstract
Osteoporosis is the most common bone disorder worldwide and is associated with a reduced quality of life with important clinical and economic consequences. The most widely accepted etiopathogenic hypothesis on the origin of osteoporosis and its complications is that they are a consequence of the synergic action of environmental and genetic factors. Bone is constantly being remodelled through anabolic and catabolic pathways in which inflammation, the NF-kB pathway and the renin-angiotensin-aldosterone system (RAAS) are crucial. The aim of our study was to determine whether polymorphisms in genes implicated in inflammation, the NF-kB pathway and RAAS modified the risk of osteoporotic fracture. We analysed 221 patients with osteoporotic fracture and 354 controls without fracture from the HORTEGA sample after 12-14 years of follow up. In addition, we studied the genotypic distribution of 230 single nucleotide polymorphisms (SNPs) in genes involved in inflammation, NF-kB pathway and RAAS. Our results showed that be carrier of the C allele of the rs2228145 IL6R polymorphism was the principal genetic risk factor associated with osteoporotic fracture. The results also showed that variant genotypes of the rs4762 AGT, rs4073 IL8, rs2070699 END1 and rs4291 ACE polymorphisms were important genetic risk factors for fracture. The study provides information about the genetic factors associated with inflammation, the NF-kB pathway and RAAS, which are involved in the risk of osteoporotic fracture and reinforces the hypothesis that genetic factors are crucial in the etiopathogenesis of osteoporosis and its complications.
Collapse
Affiliation(s)
| | - Verónica Lendinez-Tortajada
- Genomic and Genetic Diagnosis Unit, Instituto de Investigación Sanitaria Clínico de Valencia, INCLIVA, Valencia, Spain.
| | - José Luis Pérez-Castrillón
- Department of Internal Medicine, Rio Hortega Universitary Hospital, Valladolid, Spain; Department of Medicine, Faculty of Medicine, University of Valladolid, Valladolid, Spain.
| | | | - Jesica Abadía-Otero
- Department of Internal Medicine, Rio Hortega Universitary Hospital, Valladolid, Spain; Department of Medicine, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| | - Javier Martín-Vallejo
- Department of Statistics, University of Salamanca, Salamanca Biomedical Research Institute (IBSAL), Salamanca, Spain.
| | - Francisco Lara-Hernandez
- Genomic and Genetic Diagnosis Unit, Instituto de Investigación Sanitaria Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Felipe J Chaves
- Genomic and Genetic Diagnosis Unit, Instituto de Investigación Sanitaria Clínico de Valencia, INCLIVA, Valencia, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain.
| | - Ana B García-Garcia
- Genomic and Genetic Diagnosis Unit, Instituto de Investigación Sanitaria Clínico de Valencia, INCLIVA, Valencia, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain.
| | - Juan Carlos Martín-Escudero
- Department of Internal Medicine, Rio Hortega Universitary Hospital, Valladolid, Spain; Department of Medicine, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| |
Collapse
|
19
|
Vacher J, Bruccoleri M, Pata M. Ostm1 from Mouse to Human: Insights into Osteoclast Maturation. Int J Mol Sci 2020; 21:ijms21165600. [PMID: 32764302 PMCID: PMC7460669 DOI: 10.3390/ijms21165600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
The maintenance of bone mass is a dynamic process that requires a strict balance between bone formation and resorption. Bone formation is controlled by osteoblasts, while osteoclasts are responsible for resorption of the bone matrix. The opposite functions of these cell types have to be tightly regulated not only during normal bone development, but also during adult life, to maintain serum calcium homeostasis and sustain bone integrity to prevent bone fractures. Disruption of the control of bone synthesis or resorption can lead to an over accumulation of bone tissue in osteopetrosis or conversely to a net depletion of the bone mass in osteoporosis. Moreover, high levels of bone resorption with focal bone formation can cause Paget’s disease. Here, we summarize the steps toward isolation and characterization of the osteopetrosis associated trans-membrane protein 1 (Ostm1) gene and protein, essential for proper osteoclast maturation, and responsible when mutated for the most severe form of osteopetrosis in mice and humans.
Collapse
Affiliation(s)
- Jean Vacher
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
- Departement de Medecine, Universite de Montreal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3A 1A3, Canada
- Correspondence:
| | - Michael Bruccoleri
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
- Departement de Medecine, Universite de Montreal, Montreal, QC H2W 1R7, Canada
| | - Monica Pata
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
| |
Collapse
|
20
|
Rajapakse D, Peterson K, Mishra S, Fan J, Lerner J, Campos M, Wistow G. Amelotin is expressed in retinal pigment epithelium and localizes to hydroxyapatite deposits in dry age-related macular degeneration. Transl Res 2020; 219:45-62. [PMID: 32160961 PMCID: PMC7197213 DOI: 10.1016/j.trsl.2020.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/28/2022]
Abstract
Deposition of hydroxyapatite (HAP) basal to the retinal pigment epithelium (RPE) is linked to the progression of age-related macular degeneration (AMD). Serum-deprivation of RPE cells in culture mimics some features of AMD. We now show that serum-deprivation also leads to the induction of amelotin (AMTN), a protein involved in hydroxyapatite mineralization in enamel. HAP is formed in our culture model and is blocked by siRNA inhibition of AMTN expression. In situ hybridization and immunofluorescence imaging of human eye tissue show that AMTN is expressed in RPE of donor eyes with geographic atrophy ("dry" AMD) in regions with soft drusen containing HAP spherules or nodules. AMTN is not found in hard drusen, normal RPE, or donor eyes diagnosed with wet AMD. These findings suggest that AMTN is involved in formation of HAP spherules or nodules in AMD, and as such provides a new therapeutic target for slowing disease progression.
Collapse
Affiliation(s)
- Dinusha Rajapakse
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Katherine Peterson
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Sanghamitra Mishra
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Jianguo Fan
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Joshua Lerner
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Campos
- Histopathology Core Facility, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Graeme Wistow
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
21
|
Usategui-Martín R, Gestoso-Uzal N, Calero-Paniagua I, De Pereda JM, Del Pino-Montes J, González-Sarmiento R. A mutation in p62 protein (p. R321C), associated to Paget's disease of bone, causes a blockade of autophagy and an activation of NF-kB pathway. Bone 2020; 133:115265. [PMID: 32036052 DOI: 10.1016/j.bone.2020.115265] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 12/13/2022]
Abstract
Paget's disease of bone (PDB) is a bone disorder characterized by an increase in bone turnover in a disorganized way with a large increase in bone resorption followed by bone formation. The most important known genetic factor predisposing to PDB is mutation in Sequestosome1 (SQSTM1) gene. We have studied the prevalence of SQSTM1 mutations and examined genotype-phenotype correlations in a Spanish cohort of PDB patients. Also, we have characterized three PDB patients that carry the c.961C>T SQSTM1 gene mutation that it is localized in exon 6 of SQSTM1 gene and it causes the p. R321C mutation. This mutation has been reported in patients with amyotrophic lateral sclerosis and frontotemporal dementia but in our knowledge this is the first time that p62 p. R321C mutation is associated to PDB. We show that p62 p.R321C mutation could induce blockage of autophagy and cell proliferation through NF-kB pathway. These results reinforce the hypothesis of autophagy involvement in Paget's disease of bone.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain; Salamanca Institute of Biomedical Research (IBSAL), University Hospital of Salamanca, University of Salamanca, Salamanca-CSIC, Spain
| | - Nerea Gestoso-Uzal
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain; Salamanca Institute of Biomedical Research (IBSAL), University Hospital of Salamanca, University of Salamanca, Salamanca-CSIC, Spain
| | - Ismael Calero-Paniagua
- Salamanca Institute of Biomedical Research (IBSAL), University Hospital of Salamanca, University of Salamanca, Salamanca-CSIC, Spain; Rheumatology Service, University Hospital of Salamanca, Salamanca, Spain
| | - José María De Pereda
- Salamanca Institute of Biomedical Research (IBSAL), University Hospital of Salamanca, University of Salamanca, Salamanca-CSIC, Spain; Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain.
| | - Javier Del Pino-Montes
- Salamanca Institute of Biomedical Research (IBSAL), University Hospital of Salamanca, University of Salamanca, Salamanca-CSIC, Spain; Rheumatology Service, University Hospital of Salamanca, Salamanca, Spain.
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain; Salamanca Institute of Biomedical Research (IBSAL), University Hospital of Salamanca, University of Salamanca, Salamanca-CSIC, Spain; Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain.
| |
Collapse
|
22
|
Lycorine Attenuates Autophagy in Osteoclasts via an Axis of mROS/TRPML1/TFEB to Reduce LPS-Induced Bone Loss. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8982147. [PMID: 31687088 PMCID: PMC6800915 DOI: 10.1155/2019/8982147] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
Lycorine, a plant alkaloid, exhibits anti-inflammatory activity by acting in macrophages that share precursor cells with osteoclasts (OCs). We hypothesized that lycorine might decrease bone loss by acting in OCs after lipopolysaccharide (LPS) stimulation, since OCs play a main role in LPS-induced bone loss. Microcomputerized tomography (μCT) analysis revealed that lycorine attenuated LPS-induced bone loss in mice. In vivo tartrate-resistant acid phosphatase (TRAP) staining showed that increased surface area and number of OCs in LPS-treated mice were also decreased by lycorine treatment, suggesting that OCs are responsible for the bone-sparing effect of lycorine. In vitro, the increased number and activity of OCs induced by LPS were reduced by lycorine. Lycorine also decreased LPS-induced autophagy in OCs by evaluation of decreased lipidated form of microtubule-associated proteins 1A/1B light chain 3B (LC3) (LC3II) and increased sequestosome 1 (p62). Lycorine attenuated oxidized transient receptor potential cation channel, mucolipin subfamily (TRPML1) by reducing mitochondrial reactive oxygen species (mROS) and decreased transcription factor EB (TFEB) nuclear translocation. Lycorine reduced the number and activity of OCs by decreasing autophagy in OCs via an axis of mROS/TRPML1/TFEB. Collectively, lycorine protected against LPS-induced bone loss by acting in OCs. Our data highlight the therapeutic potential of lycorine for protection against inflammatory bone loss.
Collapse
|
23
|
Ethiraj P, Sambandam Y, Hathaway-Schrader JD, Haque A, Novince CM, Reddy SV. RANKL triggers resistance to TRAIL-induced cell death in oral squamous cell carcinoma. J Cell Physiol 2019; 235:1663-1673. [PMID: 31309556 DOI: 10.1002/jcp.29086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Oral squamous cell carcinoma (OSCC) occurs as a malignancy of the oral cavity. RANK ligand (RANKL) is essential for osteoclast formation/bone resorption. Recently, we showed autoregulation of receptor activator of nuclear factor-κB ligand (RANKL) stimulates OSCC cell proliferation. OSCC cells show resistance to tumor necrosis factor related apoptosis inducing ligand (TRAIL) treatment. Therefore, we hypothesize that RANKL promotes resistance for TRAIL induction of OSCC apoptotic cell death. In this study, SCC14A and SCC74A cells cultured with TRAIL revealed high-level expression of RANKL which increased resistance to TRAIL inhibition of tumor cell proliferation. RANKL stimulation inhibited terminal deoxynucleotidyl transferase dUTP nick end labeling positive staining in TRAIL-treated cells. CRISPR/Cas-9 knockout of RANKL (RANKL-KO) increased caspase-9, caspase-3 activity and cytochrome c release in OSCC cells. RANKL inhibited proapoptotic proteins BAD and BAX expression. TRAIL treatment suppressed the SQSTM1/p62 and RANKL restored the expression. Interestingly, RANKL alone significantly increased proteasome activity. RANKL-KO in OSCC cells inhibited autophagic activity as evidenced by decreased light chain 3B-II and beclin-1 expression. Thus, RANKL stimulation of OSCC tumor cells triggered resistance for TRAIL-induced OSCC cell death. Taken together, blockade of RANKL may inhibit OSCC tumor progression and enhance the potential of TRAIL induced OSCC tumor cell apoptosis.
Collapse
Affiliation(s)
- Purushoth Ethiraj
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Yuvaraj Sambandam
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sakamuri V Reddy
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
24
|
Thoenen E, Curl A, Iwakuma T. TP53 in bone and soft tissue sarcomas. Pharmacol Ther 2019; 202:149-164. [PMID: 31276706 DOI: 10.1016/j.pharmthera.2019.06.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022]
Abstract
Genomic and functional study of existing and emerging sarcoma targets, such as fusion proteins, chromosomal aberrations, reduced tumor suppressor activity, and oncogenic drivers, is broadening our understanding of sarcomagenesis. Among these mechanisms, the tumor suppressor p53 (TP53) plays significant roles in the suppression of bone and soft tissue sarcoma progression. Although mutations in TP53 were thought to be relatively low in sarcomas, modern techniques including whole-genome sequencing have recently illuminated unappreciated alterations in TP53 in osteosarcoma. In addition, oncogenic gain-of-function activities of missense mutant p53 (mutp53) have been reported in sarcomas. Moreover, new targeting strategies for TP53 have been discovered: restoration of wild-type p53 (wtp53) activity through inhibition of TP53 negative regulators, reactivation of the wtp53 activity from mutp53, depletion of mutp53, and targeting of vulnerabilities in cells with TP53 deletions or mutations. These discoveries enable development of novel therapeutic strategies for therapy-resistant sarcomas. We have outlined nine bone and soft tissue sarcomas for which TP53 plays a crucial tumor suppressive role. These include osteosarcoma, Ewing sarcoma, chondrosarcoma, rhabdomyosarcoma (RMS), leiomyosarcoma (LMS), synovial sarcoma, liposarcoma (LPS), angiosarcoma, and undifferentiated pleomorphic sarcoma (UPS).
Collapse
Affiliation(s)
- Elizabeth Thoenen
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66010, USA
| | - Amanda Curl
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66010, USA
| | - Tomoo Iwakuma
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66010, USA; Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66010, USA; Translational Laboratory Oncology Research, Children's Mercy Research Institute, Kansas City, MO 64108, USA.
| |
Collapse
|
25
|
Ivanova S, Polajnar M, Narbona-Perez AJ, Hernandez-Alvarez MI, Frager P, Slobodnyuk K, Plana N, Nebreda AR, Palacin M, Gomis RR, Behrends C, Zorzano A. Regulation of death receptor signaling by the autophagy protein TP53INP2. EMBO J 2019; 38:embj.201899300. [PMID: 30979779 DOI: 10.15252/embj.201899300] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 02/15/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
TP53INP2 positively regulates autophagy by binding to Atg8 proteins. Here, we uncover a novel role of TP53INP2 in death-receptor signaling. TP53INP2 sensitizes cells to apoptosis induced by death receptor ligands. In keeping with this, TP53INP2 deficiency in cultured cells or mouse livers protects against death receptor-induced apoptosis. TP53INP2 binds caspase-8 and the ubiquitin ligase TRAF6, thereby promoting the ubiquitination and activation of caspase-8 by TRAF6. We have defined a TRAF6-interacting motif (TIM) and a ubiquitin-interacting motif in TP53INP2, enabling it to function as a scaffold bridging already ubiquitinated caspase-8 to TRAF6 for further polyubiquitination of caspase-8. Mutations of key TIM residues in TP53INP2 abrogate its interaction with TRAF6 and caspase-8, and subsequently reduce levels of death receptor-induced apoptosis. A screen of cancer cell lines showed that those with higher protein levels of TP53INP2 are more prone to TRAIL-induced apoptosis, making TP53INP2 a potential predictive marker of cancer cell responsiveness to TRAIL treatment. These findings uncover a novel mechanism for the regulation of caspase-8 ubiquitination and reveal TP53INP2 as an important regulator of the death receptor pathway.
Collapse
Affiliation(s)
- Saška Ivanova
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain .,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.,Departament de Bioquimica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Mira Polajnar
- Institute of Biochemistry II, Goethe University School of Medicine, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Munich Cluster for System Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Alvaro Jesus Narbona-Perez
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Maria Isabel Hernandez-Alvarez
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Departament de Bioquimica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.,Hospital Universitari de Tarragona Joan XXIII, Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - Petra Frager
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Konstantin Slobodnyuk
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natalia Plana
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,ICREA, Insitució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Manuel Palacin
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.,CIBER de Enfermedades Raras, Barcelona, Spain
| | - Roger R Gomis
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,ICREA, Insitució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.,CIBERONC, Barcelona, Spain.,Departament de Medicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Christian Behrends
- Institute of Biochemistry II, Goethe University School of Medicine, Frankfurt am Main, Germany.,Munich Cluster for System Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain .,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.,Departament de Bioquimica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Sul OJ, Sung YB, Rajasekaran M, Ke K, Yu R, Back SH, Choi HS. MicroRNA-155 induces autophagy in osteoclasts by targeting transforming growth factor β-activated kinase 1-binding protein 2 upon lipopolysaccharide stimulation. Bone 2018; 116:279-289. [PMID: 30144578 DOI: 10.1016/j.bone.2018.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/30/2022]
Abstract
The autophagy pathway has been suggested to influence skeletal structure by modulating bone metabolism. Recent findings suggest that microRNAs (miR) play a critical role in autophagy. We hypothesized that inflammation induces miR-155, which enhances autophagy in osteoclasts (OC), leading to inflammatory bone loss. The expression of miR-155 was elevated in tibiae from LPS-injected mice and in OC stimulated by lipopolysaccharide (LPS) compared with vehicle treatment. Overexpression of miR-155 enhanced autophagy as well as differentiation in OC, whereas inhibition of endogenous miR-155 decreased both. Transforming growth factor β-activated kinase 1-binding protein 2 (TAB2) was identified as a target gene of miR-155 via binding to the 3'-UTR of TAB2, which directly interacts with BECLIN1. BECLIN1 was dissociated from TAB2, which started to associate with TAK1 when autophagy was induced. Our data demonstrate that LPS-induced miR-155 promoted autophagy to increase OC formation via decreased TAB2.
Collapse
Affiliation(s)
- Ok-Joo Sul
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - You-Bin Sung
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Monisha Rajasekaran
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Ke Ke
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Sung-Hoon Back
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea.
| |
Collapse
|
27
|
Rab GTPases in Osteoclastic Endomembrane Systems. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4541538. [PMID: 30186859 PMCID: PMC6114073 DOI: 10.1155/2018/4541538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
Osteoclasts (OCs) are bone-resorbing cells that maintain bone homeostasis. OC differentiation, survival, and activity are regulated by numerous small GTPases, including those of the Rab family, which are involved in plasma membrane delivery and lysosomal and autophagic degradation pathways. In resorbing OCs, polarized vesicular trafficking pathways also result in formation of the ruffled membrane, the resorbing organelle, and in transcytosis.
Collapse
|
28
|
Tsai CH, Hsu MH, Huang PH, Hsieh CT, Chiu YM, Shieh DC, Lee YJ, Tsay GJ, Wu YY. A paeonol derivative, YPH-PA3 promotes the differentiation of monocyte/macrophage lineage precursor cells into osteoblasts and enhances their autophagy. Eur J Pharmacol 2018; 832:104-113. [PMID: 29782859 DOI: 10.1016/j.ejphar.2018.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022]
Abstract
Previous studies have indicated that paeonol inhibits RANKL-induced osteoclastogenesis by inhibiting the ERK, p38, and NF-κB pathway. We modified paeonol to form a new compound, YPH-PA3, and found that it promoted osteoclastogenesis rather than inhibited it the way paeonol does. The aim of this study is to investigate the mechanisms involved in YPH-PA3-promoted osteoclastogenesis. YPH-PA3-promoted differentiation of RAW264.7 cells (human monocytes) into osteoclasts is activated through ERK/p38/JNK phosphorylation, affecting c-FOS, NF-κB, and NFATc2. Real-time quantitative PCR and western blot revealed an increased expression of autophagy-related markers during YPH-PA3-induced osteoclastogenesis. We also demonstrated the relationship between p62/LC3 localization and F-actin ring formation by double-labeling immunofluorescence. Knockdown of p62 small-interfering RNA (siRNA) attenuated YPH-PA3-induced expression of autophagy-related genes. Our study results indicated that p62 may play a role in YPH-PA3-induced autophagy and osteoclastogenesis, which may help to develop a novel therapeutic strategy against osteoclastogenesis-related diseases.
Collapse
Affiliation(s)
- Chun-Hao Tsai
- Department of Orthopedics, School of Medicine, China Medical University Hospital and China Medical University, Taichung, Taiwan
| | - Ming-Hua Hsu
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Po-Hao Huang
- Department of Internal Medicine, School of Medicine, China Medical University Hospital and China Medical University, Taichung, Taiwan
| | - Chin-Tung Hsieh
- Department of Pediatrics, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, I-Lan, Taiwan
| | - Ying-Ming Chiu
- Division of Allergy, Immunology & Rheumatology, Changhua Christian Hospital, Changhua, Taiwan; Department of Nursing, College of Medicine & Nursing, Hung Kuang University, Taichung, Taiwan
| | - Dong-Chen Shieh
- Department of Nursing, College of Medicine & Nursing, Hung Kuang University, Taichung, Taiwan
| | - Yi-Ju Lee
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Gregory J Tsay
- Department of Internal Medicine, School of Medicine, China Medical University Hospital and China Medical University, Taichung, Taiwan; Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Ying Wu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
29
|
Islam MA, Sooro MA, Zhang P. Autophagic Regulation of p62 is Critical for Cancer Therapy. Int J Mol Sci 2018; 19:ijms19051405. [PMID: 29738493 PMCID: PMC5983640 DOI: 10.3390/ijms19051405] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/30/2018] [Accepted: 05/03/2018] [Indexed: 12/13/2022] Open
Abstract
Sequestosome1 (p62/SQSTM 1) is a multidomain protein that interacts with the autophagy machinery as a key adaptor of target cargo. It interacts with phagophores through the LC3-interacting (LIR) domain and with the ubiquitinated protein aggregates through the ubiquitin-associated domain (UBA) domain. It sequesters the target cargo into inclusion bodies by its PB1 domain. This protein is further the central hub that interacts with several key signaling proteins. Emerging evidence implicates p62 in the induction of multiple cellular oncogenic transformations. Indeed, p62 upregulation and/or reduced degradation have been implicated in tumor formation, cancer promotion as well as in resistance to therapy. It has been established that the process of autophagy regulates the levels of p62. Autophagy-dependent apoptotic activity of p62 is recently being reported. It is evident that p62 plays a critical role in both autophagy and apoptosis. Therefore in this review we discuss the role of p62 in autophagy, apoptosis and cancer through its different domains and outline the importance of modulating cellular levels of p62 in cancer therapeutics.
Collapse
Affiliation(s)
- Md Ariful Islam
- Jiangsu Key Laboratory of New Drug Screening & Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| | - Mopa Alina Sooro
- Jiangsu Key Laboratory of New Drug Screening & Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| | - Pinghu Zhang
- Institute of Translational Medicine & Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
30
|
4-Phenylbutyric acid protects against lipopolysaccharide-induced bone loss by modulating autophagy in osteoclasts. Biochem Pharmacol 2018; 151:9-17. [DOI: 10.1016/j.bcp.2018.02.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 02/14/2018] [Indexed: 11/20/2022]
|
31
|
Adamik J, Silbermann R, Marino S, Sun Q, Anderson JL, Zhou D, Xie XQ, Roodman GD, Galson DL. XRK3F2 Inhibition of p62-ZZ Domain Signaling Rescues Myeloma-Induced GFI1-Driven Epigenetic Repression of the Runx2 Gene in Pre-osteoblasts to Overcome Differentiation Suppression. Front Endocrinol (Lausanne) 2018; 9:344. [PMID: 30008697 PMCID: PMC6033965 DOI: 10.3389/fendo.2018.00344] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023] Open
Abstract
Multiple myeloma bone disease (MMBD) is characterized by non-healing lytic bone lesions that persist even after a patient has achieved a hematologic remission. We previously reported that p62 (sequestosome-1) in bone marrow stromal cells (BMSC) is critical for the formation of MM-induced signaling complexes that mediate OB suppression. Importantly, XRK3F2, an inhibitor of the p62-ZZ domain, blunted MM-induced Runx2 suppression in vitro, and induced new bone formation and remodeling in the presence of tumor in vivo. Additionally, we reported that MM cells induce the formation of repressive chromatin on the Runx2 gene in BMSC via direct binding of the transcriptional repressor GFI1, which recruits the histone modifiers, histone deacetylase 1 (HDAC1) and Enhancer of zeste homolog 2 (EZH2). In this study we investigated the mechanism by which blocking p62-ZZ domain-dependent signaling prevents MM-induced suppression of Runx2 in BMSC. XRK3F2 prevented MM-induced upregulation of Gfi1 and repression of the Runx2 gene when present in MM-preOB co-cultures. We also show that p62-ZZ-domain blocking by XRK3F2 also prevented MM conditioned media and TNF plus IL7-mediated Gfi1 mRNA upregulation and the concomitant Runx2 repression, indicating that XRK3F2's prevention of p62-ZZ domain signaling within preOB is involved in the response. Chromatin immunoprecipitation (ChIP) analyses revealed that XRK3F2 decreased MM-induced GFI1 occupancy at the Runx2-P1 promoter and prevented recruitment of HDAC1, thus preserving the transcriptionally permissive chromatin mark H3K9ac on Runx2 and allowing osteogenic differentiation. Furthermore, treatment of MM-exposed preOB with XRK3F2 after MM removal decreased GFI1 enrichment at Runx2-P1 and rescued MM-induced suppression of Runx2 mRNA and its downstream osteogenic gene targets together with increased osteogenic differentiation. Further, primary BMSC (hBMSC) from MM patients (MM-hBMSC) had little ability to increase H3K9ac on the Runx2 promoter in osteogenic conditions when compared to hBMSC from healthy donors (HD). XRK3F2 treatment enriched Runx2 gene H3K9ac levels in MM-hBMSC to the level observed in HD-hBMSC, but did not alter HD-hBMSC H3K9ac. Importantly, XRK3F2 treatment of long-term MM-hBMSC cultures rescued osteogenic differentiation and mineralization. Our data show that blocking p62-ZZ domain-dependent signaling with XRK3F2 can reverse epigenetic-based mechanisms of MM-induced Runx2 suppression and promote osteogenic differentiation.
Collapse
Affiliation(s)
- Juraj Adamik
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rebecca Silbermann
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Silvia Marino
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Quanhong Sun
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Judith L. Anderson
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Dan Zhou
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - G. David Roodman
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Deborah L. Galson
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- *Correspondence: Deborah L. Galson ;
| |
Collapse
|
32
|
Wu DJ, Adamopoulos IE. Loss of WDFY3 ameliorates severity of serum transfer-induced arthritis independently of autophagy. Cell Immunol 2017; 316:61-69. [PMID: 28449847 DOI: 10.1016/j.cellimm.2017.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/26/2017] [Accepted: 04/10/2017] [Indexed: 11/16/2022]
Abstract
WDFY3 is a master regulator of selective autophagy that we recently showed to interact with TRAF6 and augment RANKL-induced osteoclastogenesis in vitro and in vivo via the NF-κB pathway. Since the NF-κB pathway plays a major role in inflammation herein, we investigate the role of WDFY3 in an arthritis animal model. Our data show that WDFY3 conditional knockout mice (Wdfy3loxP/loxP-LysM-Cre+) were protected in the K/BxN serum transfer-induced arthritis animal model. These effects were independent of alterations in starvation-induced autophagy as evidenced by Western blot analysis of the autophagy marker LC3, autophagosome formation in osteoclast precursors and lysosome formation in osteoclasts derived from WDFY3-cKO mice compared to controls. Moreover, we demonstrate by immunofluorescence and co-immunoprecipitation that WDFY3 interacts with SQSTM1 in macrophages and osteoclasts. Collectively, our data suggest that loss of WDFY3 in myeloid cells leads to reduced severity of inflammatory arthritis independently of WDFY3 function in starvation-induced autophagy.
Collapse
Affiliation(s)
- Dennis J Wu
- Graduate Group in Immunology, University of California at Davis, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, USA
| | - Iannis E Adamopoulos
- Graduate Group in Immunology, University of California at Davis, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, USA.
| |
Collapse
|
33
|
Li C, Siragy HM. Autophagy upregulates (pro)renin receptor expression via reduction of P62/SQSTM1 and activation of ERK1/2 signaling pathway in podocytes. Am J Physiol Regul Integr Comp Physiol 2017; 313:R58-R64. [PMID: 28450279 DOI: 10.1152/ajpregu.00088.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/19/2022]
Abstract
Autophagy plays a major role in podocytes health and disease. P62, also known as sequestosome-1 (SQSTM1), is a marker for autophagic activity and is required for the formation and degradation of ubiquitnated protein by autophagy. Knockout of p62 enhanced extracellular signal-regulated kinases (ERK1/2) activity. (pro)renin receptor (PRR) is expressed in podocytes where it contributes to the homeostasis of these cells. The influence of autophagy on PRR expression is unknown. We hypothesized that in podocytes, upregulation of autophagic activity increases PRR expression via reduction of p62 and stimulation of ERK1/2 signaling pathway. Cultured mouse podocytes were treated with the autophagy activators, rapamycin or Earle's balanced salt solution (EBSS), for 48 h. Both rapamycin and EBSS significantly decreased p62 protein levels, increased ERK1/2 activation by phosphorylating pTpY185/187, and increased mRNA and protein expressions of PRR. Utilizing confocal microscopy demonstrated that rapamycin and EBSS significantly decreased p62/SQSTM1 and increased PRR protein expressions. Similarly, by enhancing autophagic activity by transfection with autophagy-related 5 (ATG5) cDNA or ATG7 cDNA, results similar to those observed with rapamycin and EBSS treatments were produced. Inhibition of autophagic flux with bafilomycin A1 reversed the effects of rapamycin. ERK1/2 inhibitor U0126 significantly attenuated mRNA and protein expressions of PRR in podocytes treated with rapamycin. In conclusion, upregulation of autophagy enhanced PRR expression through reduction of p62 and stimulation of ERK1/2 activity signaling pathway.
Collapse
Affiliation(s)
- Caixia Li
- Division of Endocrinology and Metabolism, University of Virginia Health System, Charlottesville, Virginia
| | - Helmy M Siragy
- Division of Endocrinology and Metabolism, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
34
|
Natural uranium impairs the differentiation and the resorbing function of osteoclasts. Biochim Biophys Acta Gen Subj 2017; 1861:715-726. [PMID: 28089586 DOI: 10.1016/j.bbagen.2017.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/13/2016] [Accepted: 01/05/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Uranium is a naturally occurring radionuclide ubiquitously present in the environment. The skeleton is the main site of uranium long-term accumulation. While it has been shown that natural uranium is able to perturb bone metabolism through its chemical toxicity, its impact on bone resorption by osteoclasts has been poorly explored. Here, we examined for the first time in vitro effects of natural uranium on osteoclasts. METHODS The effects of uranium on the RAW 264.7 monocyte/macrophage mouse cell line and primary murine osteoclastic cells were characterized by biochemical, molecular and functional analyses. RESULTS We observed a cytotoxicity effect of uranium on osteoclast precursors. Uranium concentrations in the μM range are able to inhibit osteoclast formation, mature osteoclast survival and mineral resorption but don't affect the expression of the osteoclast gene markers Nfatc1, Dc-stamp, Ctsk, Acp5, Atp6v0a3 or Atp6v0d2 in RAW 274.7 cells. Instead, we observed that uranium induces a dose-dependent accumulation of SQSTM1/p62 during osteoclastogenesis. CONCLUSIONS We show here that uranium impairs osteoclast formation and function in vitro. The decrease in available precursor cells, as well as the reduced viability of mature osteoclasts appears to account for these effects of uranium. The SQSTM1/p62 level increase observed in response to uranium exposure is of particular interest since this protein is a known regulator of osteoclast formation. A tempting hypothesis discussed herein is that SQSTM1/p62 dysregulation contributes to uranium effects on osteoclastogenesis. GENERAL SIGNIFICANCE We describe cellular and molecular effects of uranium that potentially affect bone homeostasis.
Collapse
|
35
|
Taniguchi K, Yamachika S, He F, Karin M. p62/SQSTM1-Dr. Jekyll and Mr. Hyde that prevents oxidative stress but promotes liver cancer. FEBS Lett 2016; 590:2375-97. [PMID: 27404485 DOI: 10.1002/1873-3468.12301] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/08/2016] [Accepted: 07/09/2016] [Indexed: 12/17/2022]
Abstract
p62/SQSTM1 is a multifunctional signaling hub and autophagy adaptor with many binding partners, which allow it to activate mTORC1-dependent nutrient sensing, NF-κB-mediated inflammatory responses, and the NRF2-activated antioxidant defense. p62 recognizes polyubiquitin chains via its C-terminal domain and binds to LC3 via its LIR motif, thereby promoting the autophagic degradation of ubiquitinated cargos. p62 accumulates in many human liver diseases, including nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC), where it is a component of Mallory-Denk bodies and intracellular hyaline bodies. Chronic p62 elevation contributes to HCC development by preventing oncogene-induced senescence and death of cancer-initiating cells and enhancing their proliferation. In this review, we discuss p62-mediated signaling pathways and their roles in liver pathophysiology, especially NASH and HCC.
Collapse
Affiliation(s)
- Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA.,Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Yamachika
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA
| | - Feng He
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
36
|
McManus S, Bisson M, Chamberland R, Roy M, Nazari S, Roux S. Autophagy and 3-Phosphoinositide-Dependent Kinase 1 (PDK1)-Related Kinome in Pagetic Osteoclasts. J Bone Miner Res 2016; 31:1334-43. [PMID: 26848537 DOI: 10.1002/jbmr.2806] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/24/2016] [Accepted: 02/03/2016] [Indexed: 01/30/2023]
Abstract
In Paget's disease of bone (PDB), a major contributory factor are osteoclasts (OCs) that are larger, more numerous, resistant to apoptosis, and hyperactive. The aim of this human in vitro study was to identify kinase cascades involved in the OC phenotype and to determine their impact on downstream processes. Basal phosphorylation levels of Akt and ERK were found to be elevated in PDB OCs. Given our previous findings that 3-phosphoinositide-dependent protein kinase 1 (PDK1) associates with the crucial adaptor p62 in OCs, we hypothesized that PDK1 may play an important role in OC-related kinome regulation. The increased phosphorylation of Akt and its substrate GSK3β observed in PDB OCs was reduced significantly upon PDK1 inhibition, as well as that of 4EBP1 and Raptor. This suggests a PDK1/Akt-dependent activation of mammalian target of rapamycin complex 1 (mTORC1) in PDB OCs. The resistance to apoptosis and the bone resorption were also overcome upon PDK1 inhibition. Studying autophagy by LC3B expression, we found a less inducible autophagy compared with control cells, which was reversed by PDK1 inhibition. In addition, PBD OCs exhibited higher LC3B-II/LC3B-I ratios and numbers of p62 and LC3B puncta per OC area, which did not further increase in the presence of lysosomal protease inhibitors, suggesting an accumulation of non-degradative autophagosomes. Together these results indicate a strong potential regulatory role for PDK1 in OC stimulatory pathways (Akt, ERK) and autophagy induction (via mTORC1), which may contribute to the OC phenotype in PDB. We also identified defects in late autophagosome maturation in these cells, the mechanism of which remains to be determined. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stephen McManus
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada
| | - Martine Bisson
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada
| | - Richard Chamberland
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada
| | - Michèle Roy
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada
| | - Shekeba Nazari
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada
| | - Sophie Roux
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
37
|
Zhang J, Yang Z, Dong J. P62: An emerging oncotarget for osteolytic metastasis. J Bone Oncol 2016; 5:30-7. [PMID: 26998424 PMCID: PMC4782024 DOI: 10.1016/j.jbo.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/18/2016] [Accepted: 01/30/2016] [Indexed: 12/13/2022] Open
Abstract
Bone metastasis occurs in the majority of late-stage tumors with poor prognosis. It is mainly classified as osteoblastic metastasis and osteolytic metastasis. The pathogenesis of osteolytic metastasis is a “vicious cycle” between tumor cells and bone cells (primarily the osteoclasts), which is mediated by secretory factors. The P62 adapter protein is a versatile multitasker between tumor cells and bone cells. The overexpression of P62 has been detected among a variety of tumors, playing positive roles in both tumorigenesis and metastasis. Moreover, P62 is an important modulator of the osteoclastogenesis pathway. Therefore, the ability of P62 to modulate tumors and osteoclasts suggests that it may be a feasible oncotarget for bone metastasis, especially for osteolytic metastasis. Recent research has shown that a P62 DNA vaccine triggered effective anti-tumor, anti-metastatic and anti-osteoporotic activities. Growing lines of evidence point to P62 as an emerging oncotarget for osteolytic metastasis. In this review, we outline the different roles of P62 in tumor cells and osteoclasts, focusing on the P62-related signaling pathway in key steps of osteolytic metastasis, including tumorigenesis, metastasis and osteoclastogenesis. Finally, we discuss the newest observations on P62 as an oncotarget for osteolytic metastasis treatment.
Collapse
Affiliation(s)
- Jing Zhang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Jian Dong
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China; Stem Cell Therapy Technical of Clinical Transformation and Basic Research Key Laboratory of Yunnan Province, Kunming, Yunnan 650118, PR China
| |
Collapse
|
38
|
Shi J, Wang L, Zhang H, Jie Q, Li X, Shi Q, Huang Q, Gao B, Han Y, Guo K, Liu J, Yang L, Luo Z. Glucocorticoids: Dose-related effects on osteoclast formation and function via reactive oxygen species and autophagy. Bone 2015; 79:222-32. [PMID: 26115910 DOI: 10.1016/j.bone.2015.06.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 01/09/2023]
Abstract
Whether glucocorticoids directly enhance or interrupt osteoclastogenesis is still a controversial subject. In this study, we ascertained the dose-dependent positive effects of glucocorticoids on osteoclastogenesis in vivo and in vitro as well as investigated the mechanism in vitro. As the dose of glucocorticoids increased, osteoclastogenesis was stimulated at 0.1 μM, a peak was achieved at 1 μM and a corresponding decrease occurred at 10 μM. Reactive oxygen species (ROS), which play a crucial role in osteoclastogenesis, and autophagy flux activity, a cellular recycling process, were consistently up-regulated along with the dose-dependent effects of the glucocorticoids on osteoclast formation and function. N-acetyl-cysteine (NAC), a ROS scavenger, abrogated the effects of the glucocorticoids on autophagy and osteoclastogenesis. Moreover, 3-methyladenine (3-MA), an autophagy inhibitor, interrupted osteoclastogenesis stimulation by the glucocorticoids. These results implied that with glucocorticoid administration, ROS and autophagy, as a downstream factor of ROS, played vital roles in osteoclast formation and function. 3-MA administration did not enhance ROS accumulation, so that autophagy had no effect on ROS induced by glucocorticoids. Our investigation demonstrated that glucocorticoids had dose-dependent positive effects on osteoclast formation and function via ROS and autophagy. These results provide support for ROS and autophagy as therapeutic targets in glucocorticoid-related bone loss diseases such as glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Jun Shi
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Long Wang
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Hongyang Zhang
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Qiang Jie
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Xiaojie Li
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Qiyue Shi
- Institute of Orthopedics, Xi'an Chinese Medicine Hospital, China
| | - Qiang Huang
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Bo Gao
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Yuehu Han
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Kai Guo
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China
| | - Jian Liu
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China.
| | - Liu Yang
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China.
| | - Zhuojing Luo
- Institute of Orthopedics, Xijing Hospital, Fourth Military Medical University, China.
| |
Collapse
|
39
|
Xi G, Shen X, Wai C, Vilas CK, Clemmons DR. Hyperglycemia stimulates p62/PKCζ interaction, which mediates NF-κB activation, increased Nox4 expression, and inflammatory cytokine activation in vascular smooth muscle. FASEB J 2015; 29:4772-82. [PMID: 26231202 DOI: 10.1096/fj.15-275453] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/20/2015] [Indexed: 12/20/2022]
Abstract
Hyperglycemia leads to vascular smooth muscle cell (VSMC) dedifferentiation and enhances responses to IGF-I. Prior studies showed that hyperglycemia stimulated NADPH oxidase 4 (Nox4) synthesis, and IGF-I facilitated its recruitment to a signaling complex where it oxidized src, leading to AKT and MAPK activation. To determine the mechanism that led to these changes, we analyzed the roles of p62 (sequestrosome1) and PKCζ. Hyperglycemia induced a 4.9 ± 1.0-fold increase in p62/PKCζ association, and disruption of PKCζ/p62 using a peptide inhibitor or p62 knockdown reduced PKCζ activation (78 ± 6%). 3-Phosphoinoside-dependent protein kinase 1 was also recruited to the p62 complex and directly phosphorylated PKCζ, leading to its activation (3.1 ± 0.4-fold). Subsequently, activated PKCζ phosphorylated p65 rel, which led to increased Nox4 synthesis. Studies in diabetic mice confirmed these findings (6.0 ± 0.4-fold increase in p62/PKCζ) and their disruption of attenuated Nox4 synthesis (76 ± 9% reduction). PKCζ/p62 activation stimulated inflammatory cytokine production and enhanced IGF-I-stimulated VSMC proliferation. These results define the molecular mechanism by which PKCζ is activated in response to hyperglycemia and suggest that this could be a mechanism by which other stimuli such as cytokines or metabolic stress function to stimulate NF-κB activation, thereby altering VSMC sensitivity to IGF-I.
Collapse
Affiliation(s)
- Gang Xi
- *Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina, USA; and College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Xinchun Shen
- *Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina, USA; and College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Christine Wai
- *Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina, USA; and College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Caroline K Vilas
- *Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina, USA; and College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - David R Clemmons
- *Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina, USA; and College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| |
Collapse
|
40
|
Klinck R, Laberge G, Bisson M, McManus S, Michou L, Brown JP, Roux S. Alternative splicing in osteoclasts and Paget's disease of bone. BMC MEDICAL GENETICS 2014; 15:98. [PMID: 25115182 PMCID: PMC4143580 DOI: 10.1186/s12881-014-0098-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 08/07/2014] [Indexed: 01/12/2023]
Abstract
Background Mutations in the SQSTM1/p62 gene have been reported in Paget’s disease of bone (PDB), but they are not sufficient to induce the pagetic osteoclast (OC) phenotype. We hypothesized that specific RNA isoforms of OC-related genes may contribute to the overactivity of pagetic OCs, along with other genetic predisposing factors. Methods Alternative splicing (AS) events were studied using a PCR-based screening strategy in OC cultures from 29 patients with PDB and 26 healthy donors (HD), all genotyped for the p62P392L mutation. Primer pairs targeting 5223 characterized AS events were used to analyze relative isoform ratios on pooled cDNA from samples of the four groups (PDB, PDBP392L, HD, HDP392L). Of the 1056 active AS events detected in the screening analysis, 192 were re-analyzed on non-amplified cDNA from each subject of the whole cohort. Results This analysis led to the identification of six AS events significantly associated with PDB, but none with p62P392L. The corresponding genes included LGALS8, RHOT1, CASC4, USP4, TBC1D25, and PIDD. In addition, RHOT1 and LGALS8 genes were upregulated in pagetic OCs, as were CASC4 and RHOT1 genes in the presence of p62P392L. Finally, we showed that the proteins encoded by LGALS8, RHOT1, USP4, TBC1D25, and PIDD were expressed in human OCs. Conclusion This study allowed the identification of hitherto unknown players in OC biology, and our findings of a differential AS in pagetic OCs may generate new concepts in the pathogenesis of PDB.
Collapse
|
41
|
Pandey P, Mersha MD, Dhillon HS. A synergistic approach towards understanding the functional significance of dopamine receptor interactions. J Mol Signal 2013; 8:13. [PMID: 24308343 PMCID: PMC3878971 DOI: 10.1186/1750-2187-8-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/18/2013] [Indexed: 11/10/2022] Open
Abstract
The importance of the neurotransmitter dopamine (DA) in the nervous system is underscored by its role in a wide variety of physiological and neural functions in both vertebrates and invertebrates. Binding of dopamine to its membrane receptors initiates precise signaling cascades that result in specific cellular responses. Dopamine receptors belong to a super-family of G-protein coupled receptors (GPCRs) that are characterized by seven trans-membrane domains. In mammals, five dopamine receptors have been identified which are grouped into two different categories D1- and D2-like receptors. The interactions of DA receptors with other proteins including specific Gα subunits are critical in deciding the fate of downstream molecular events carried out by effector proteins. In this mini-review we provide a synopsis of known protein-protein interactions of DA receptors and a perspective on the potential synergistic utility of Caenorhabditis elegans as a model eukaryote with a comparatively simpler nervous system to gain insight on the neuronal and behavioral consequences of the receptor interactions.
Collapse
Affiliation(s)
| | | | - Harbinder S Dhillon
- Department of Biological Sciences, Center for Neuroscience Research, Delaware State University, Dover, DE 19901, USA.
| |
Collapse
|
42
|
Ishii T, Warabi E, Siow RCM, Mann GE. Sequestosome1/p62: a regulator of redox-sensitive voltage-activated potassium channels, arterial remodeling, inflammation, and neurite outgrowth. Free Radic Biol Med 2013; 65:102-116. [PMID: 23792273 DOI: 10.1016/j.freeradbiomed.2013.06.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
Abstract
Sequestosome1/p62 (SQSTM1) is an oxidative stress-inducible protein regulated by the redox-sensitive transcription factor Nrf2. It is not an antioxidant but known as a multifunctional regulator of cell signaling with an ability to modulate targeted or selective degradation of proteins through autophagy. SQSTM1 implements these functions through physical interactions with different types of proteins including atypical PKCs, nonreceptor-type tyrosine kinase p56(Lck) (Lck), polyubiquitin, and autophagosomal factor LC3. One of the notable physiological functions of SQSTM1 is the regulation of redox-sensitive voltage-gated potassium (Kv) channels which are composed of α and β subunits: (Kvα)4 (Kvβ)4. Previous studies have established that SQSTM1 scaffolds PKCζ, enhancing phosphorylation of Kvβ which induces inhibition of pulmonary arterial Kv1.5 channels under acute hypoxia. Recent studies reveal that Lck indirectly interacts with Kv1.3 α subunits and plays a key role in acute hypoxia-induced Kv1.3 channel inhibition in T lymphocytes. Kv1.3 channels provide a signaling platform to modulate the migration and proliferation of arterial smooth muscle cells and activation of T lymphocytes, and hence have been recognized as a therapeutic target for treatment of restenosis and autoimmune diseases. In this review, we focus on the functional interactions of SQSTM1 with Kv channels through two key partners aPKCs and Lck. Furthermore, we provide molecular insights into the functions of SQSTM1 in suppression of proliferation of arterial smooth muscle cells and neointimal hyperplasia following carotid artery ligation, in T lymphocyte differentiation and activation, and in NGF-induced neurite outgrowth in PC12 cells.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8575, Japan
| | - Richard C M Siow
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, London SE1 9NH, UK
| | - Giovanni E Mann
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, London SE1 9NH, UK
| |
Collapse
|
43
|
Ochotny N, Voronov I, Owen C, Aubin JE, Manolson MF. The R740S mutation in the V-ATPase a3 subunit results in osteoclast apoptosis and defective early-stage autophagy. J Cell Biochem 2013; 114:2823-33. [DOI: 10.1002/jcb.24630] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 07/22/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Noelle Ochotny
- Faculty of Dentistry; Dental Research Institute; University of Toronto; Toronto; Ontario; Canada
| | - Irina Voronov
- Faculty of Dentistry; Dental Research Institute; University of Toronto; Toronto; Ontario; Canada
| | - Celeste Owen
- Centre for Modeling Human Disease; Samuel Lunenfeld Research Institute; Mt. Sinai Hospital; Toronto; Ontario; Canada
| | | | - Morris F. Manolson
- Faculty of Dentistry; Dental Research Institute; University of Toronto; Toronto; Ontario; Canada
| |
Collapse
|
44
|
Manley S, Williams JA, Ding WX. Role of p62/SQSTM1 in liver physiology and pathogenesis. Exp Biol Med (Maywood) 2013; 238:525-38. [PMID: 23856904 DOI: 10.1177/1535370213489446] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
p62/sequestosome-1/A170/ZIP (hereafter referred to as p62) is a scaffold protein that has multiple functions, such as signal transduction, cell proliferation, cell survival, cell death, inflammation, tumourigenesis and oxidative stress response. While p62 is an autophagy substrate and is degraded by autophagy, p62 serves as an autophagy receptor for selective autophagic clearance of protein aggregates and organelles. Moreover, p62 functions as a signalling hub for various signalling pathways, including NF-κB, Nrf2 and mTOR. In this review, we discuss the pathophysiological role of p62 in the liver, including formation of hepatic inclusion bodies, cholestasis, obesity, insulin resistance, liver cell death and tumourigenesis.
Collapse
Affiliation(s)
- Sharon Manley
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, USA
| | | | | |
Collapse
|
45
|
Roy B. Biomolecular basis of the role of diabetes mellitus in osteoporosis and bone fractures. World J Diabetes 2013; 4:101-113. [PMID: 23961320 PMCID: PMC3746082 DOI: 10.4239/wjd.v4.i4.101] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/04/2013] [Accepted: 07/18/2013] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis has become a serious health problem throughout the world which is associated with an increased risk of bone fractures and mortality among the people of middle to old ages. Diabetes is also a major health problem among the people of all age ranges and the sufferers due to this abnormality increasing day by day. The aim of this review is to summarize the possible mechanisms through which diabetes may induce osteoporosis. Diabetes mellitus generally exerts its effect on different parts of the body including bone cells specially the osteoblast and osteoclast, muscles, retina of the eyes, adipose tissue, endocrine system specially parathyroid hormone (PTH) and estrogen, cytokines, nervous system and digestive system. Diabetes negatively regulates osteoblast differentiation and function while positively regulates osteoclast differentiation and function through the regulation of different intermediate factors and thereby decreases bone formation while increases bone resorption. Some factors such as diabetic neuropathy, reactive oxygen species, Vitamin D, PTH have their effects on muscle cells. Diabetes decreases the muscle strength through regulating these factors in various ways and ultimately increases the risk of fall that may cause bone fractures.
Collapse
|
46
|
Epigenetic upregulation of lncRNAs at 13q14.3 in leukemia is linked to the In Cis downregulation of a gene cluster that targets NF-kB. PLoS Genet 2013; 9:e1003373. [PMID: 23593011 PMCID: PMC3616974 DOI: 10.1371/journal.pgen.1003373] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 01/28/2013] [Indexed: 01/07/2023] Open
Abstract
Non-coding RNAs are much more common than previously thought. However, for the vast majority of non-coding RNAs, the cellular function remains enigmatic. The two long non-coding RNA (lncRNA) genes DLEU1 and DLEU2 map to a critical region at chromosomal band 13q14.3 that is recurrently deleted in solid tumors and hematopoietic malignancies like chronic lymphocytic leukemia (CLL). While no point mutations have been found in the protein coding candidate genes at 13q14.3, they are deregulated in malignant cells, suggesting an epigenetic tumor suppressor mechanism. We therefore characterized the epigenetic makeup of 13q14.3 in CLL cells and found histone modifications by chromatin-immunoprecipitation (ChIP) that are associated with activated transcription and significant DNA-demethylation at the transcriptional start sites of DLEU1 and DLEU2 using 5 different semi-quantitative and quantitative methods (aPRIMES, BioCOBRA, MCIp, MassARRAY, and bisulfite sequencing). These epigenetic aberrations were correlated with transcriptional deregulation of the neighboring candidate tumor suppressor genes, suggesting a coregulation in cis of this gene cluster. We found that the 13q14.3 genes in addition to their previously known functions regulate NF-kB activity, which we could show after overexpression, siRNA-mediated knockdown, and dominant-negative mutant genes by using Western blots with previously undescribed antibodies, by a customized ELISA as well as by reporter assays. In addition, we performed an unbiased screen of 810 human miRNAs and identified the miR-15/16 family of genes at 13q14.3 as the strongest inducers of NF-kB activity. In summary, the tumor suppressor mechanism at 13q14.3 is a cluster of genes controlled by two lncRNA genes that are regulated by DNA-methylation and histone modifications and whose members all regulate NF-kB. Therefore, the tumor suppressor mechanism in 13q14.3 underlines the role both of epigenetic aberrations and of lncRNA genes in human tumorigenesis and is an example of colocalization of a functionally related gene cluster.
Collapse
|
47
|
Notas G, Kampa M, Pelekanou V, Troullinaki M, Jacquot Y, Leclercq G, Castanas E. Whole transcriptome analysis of the ERα synthetic fragment P295-T311 (ERα17p) identifies specific ERα-isoform (ERα, ERα36)-dependent and -independent actions in breast cancer cells. Mol Oncol 2013; 7:595-610. [PMID: 23474223 DOI: 10.1016/j.molonc.2013.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 02/06/2013] [Accepted: 02/07/2013] [Indexed: 02/07/2023] Open
Abstract
ERα17p is a peptide corresponding to the sequence P295LMIKRSKKNSLALSLT311 of the estrogen receptor alpha (ERα) and initially found to interfere with ERα-related calmodulin binding. ERα17p was subsequently found to elicit estrogenic responses in E2-deprived ERα-positive breast cancer cells, increasing proliferation and ERE-dependent gene transcription. Surprisingly, in E2-supplemented media, ERα17p-induced apoptosis and modified the actin network, influencing cell motility. Here, we report that ERα17p internalizes in breast cancer cells (T47D, MDA-MB-231, SKBR3) and induces a massive early (3 h) transcriptional activity. Remarkably, about 75% of significantly modified transcripts were also modified by E2, confirming the pro-estrogenic profile of ERα17p. The different ER spectra of the used cell lines allowed us to identify a specific ERα17p signature related to ERα as well as its variant ERα36. With respect to ERα, the peptide activates nuclear (cell cycle, cell proliferation, nucleic acid and protein synthesis) and extranuclear signaling pathways. In contrast, through ERα36, it mainly triggers inhibitory actions on inflammation. This is the first work reporting a detailed ERα36-specific transcriptional signature. In addition, we report that ERα17p-induced transcripts related to apoptosis and actin modifying effects of the peptide are independent from its estrogen receptor(s)-related actions. We discuss our findings in view of the potential use of ERα17p as a selective peptidomimetic estrogen receptor modulator (PERM).
Collapse
Affiliation(s)
- George Notas
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion 71003, Greece
| | | | | | | | | | | | | |
Collapse
|
48
|
Rosenfeld JA, Kim KH, Angle B, Troxell R, Gorski JL, Westemeyer M, Frydman M, Senturias Y, Earl D, Torchia B, Schultz RA, Ellison JW, Tsuchiya K, Zimmerman S, Smolarek TA, Ballif BC, Shaffer LG. Further Evidence of Contrasting Phenotypes Caused by Reciprocal Deletions and Duplications: Duplication of NSD1 Causes Growth Retardation and Microcephaly. Mol Syndromol 2013; 3:247-54. [PMID: 23599694 DOI: 10.1159/000345578] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2012] [Indexed: 12/15/2022] Open
Abstract
Microduplications of the Sotos syndrome region containing NSD1 on 5q35 have recently been proposed to cause a syndrome of microcephaly, short stature and developmental delay. To further characterize this emerging syndrome, we report the clinical details of 12 individuals from 8 families found to have interstitial duplications involving NSD1, ranging in size from 370 kb to 3.7 Mb. All individuals are microcephalic, and height and childhood weight range from below average to severely restricted. Mild-to-moderate learning disabilities and/or developmental delay are present in all individuals, including carrier family members of probands; dysmorphic features and digital anomalies are present in a majority. Craniosynostosis is present in the individual with the largest duplication, though the duplication does not include MSX2, mutations of which can cause craniosynostosis, on 5q35.2. A comparison of the smallest duplication in our cohort that includes the entire NSD1 gene to the individual with the largest duplication that only partially overlaps NSD1 suggests that whole-gene duplication of NSD1 in and of itself may be sufficient to cause the abnormal growth parameters seen in these patients. NSD1 duplications may therefore be added to a growing list of copy number variations for which deletion and duplication of specific genes have contrasting effects on body development.
Collapse
Affiliation(s)
- J A Rosenfeld
- Signature Genomic Laboratories, PerkinElmer, Inc., Spokane, Wash., USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kim SE, Suh DH, Yun YP, Lee JY, Park K, Chung JY, Lee DW. Local delivery of alendronate eluting chitosan scaffold can effectively increase osteoblast functions and inhibit osteoclast differentiation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:2739-2749. [PMID: 22850978 DOI: 10.1007/s10856-012-4729-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 07/23/2012] [Indexed: 06/01/2023]
Abstract
The aim of this study was to investigate the effect of alendronate released from chitosan scaffolds on enhancement of osteoblast functions and inhibition of osteoclast differentiation in vitro. The surface and cell morphologies of chitosan scaffolds and alendronate-loaded chitosan scaffolds were characterized by variable pressure field emission scanning electron microscope (VP-FE-SEM). Alendronate was released in a sustained manner. For evaluating osteoblast functions in MG-63 cells, we investigated cell proliferation, alkaline phosphatase (ALP) activity, and calcium deposition. Furthermore, for evaluating inhibition of osteoclast differentiation in RAW 264.7 cells, we investigated tartrate-resistant acid phosphatase (TRAP) activity, TRAP staining, and gene expressions. The in vitro studies revealed that osteoblasts grown on alendronate-loaded chitosan scaffold showed a significant increment in cell proliferation, ALP activity, and calcium deposition as compared to those grown on chitosan scaffolds. In addition, the in vitro study showed that osteoclast differentiation in RAW 264.7 cells cultured on alendronate-loaded chitosan scaffolds was greatly inhibited as compared to those cultured on chitosan scaffolds by the results of TRAP activity, TRAP staining, and gene expressions. Taken together, alendronate-loaded chitosan scaffolds could achieve the dual functions of improvement in osteoblast functions and inhibition of osteoclast differentiation. Thus, alendronate-eluting chitosan substrates are promising materials for enhancing osteoblast functions and inhibiting osteoclast differentiation in orthopedic and dental fields.
Collapse
Affiliation(s)
- Sung Eun Kim
- Department of Orthopedic Surgery and Rare Diseases Institute, Korea University Medical College, Guro Hospital, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Imbalances between bone resorption and formation lie at the root of disorders such as osteoporosis, Paget's disease of bone (PDB), and osteopetrosis. Recently, genetic and functional studies have implicated proteins involved in autophagic protein degradation as important mediators of bone cell function in normal physiology and in pathology. Autophagy is the conserved process whereby aggregated proteins, intracellular pathogens, and damaged organelles are degraded and recycled. This process is important both for normal cellular quality control and in response to environmental or internal stressors, particularly in terminally-differentiated cells. Autophagic structures can also act as hubs for the spatial organization of recycling and synthetic process in secretory cells. Alterations to autophagy (reduction, hyperactivation, or impairment) are associated with a number of disorders, including neurodegenerative diseases and cancers, and are now being implicated in maintenance of skeletal homoeostasis. Here, we introduce the topic of autophagy, describe the new findings that are starting to emerge from the bone field, and consider the therapeutic potential of modifying this pathway for the treatment of age-related bone disorders.
Collapse
Affiliation(s)
- Lynne J Hocking
- Musculoskeletal Research Programme, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK.
| | | | | |
Collapse
|