1
|
Wajda A, Bogucka D, Stypińska B, Radkowski MJ, Targowski T, Dudek E, Kmiołek T, Modzelewska E, Paradowska-Gorycka A. Expression of Prostaglandin Genes and β-Catenin in Whole Blood as Potential Markers of Muscle Degeneration. Int J Mol Sci 2023; 24:12885. [PMID: 37629065 PMCID: PMC10454559 DOI: 10.3390/ijms241612885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/09/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Prostaglandin signaling pathways are closely related to inflammation, but also muscle regeneration and processes associated with frailty and sarcopenia, whereas β-catenin (CTNNB1 gene) as a part of Wnt signaling is also involved in the differentiation of muscle cells and fibrosis. The present study analyzed the association between selected prostaglandin pathway genes and clinical parameters in patients with sarcopenia and frailty syndrome. The present study was conducted on patients with sarcopenia, frailty syndrome, and control older patients (N = 25). Additionally, two healthy controls at the age of 25-30 years (N = 51) and above 50 years old (N = 42) were included. The expression of the PTRGER4, PTGES2 (COX2), PTGS2, and CTNNB1 genes in whole blood was checked by the qPCR method. The serum cytokine levels (IL-10, TNFα, IFN-y, IL-1α, IL-1β) in patients and controls were checked by the Q-Plex Human Cytokine Panel. The results showed a significant effect of age on PTGER4 gene expression (p = 0.01). A negative trend between the appendicular skeletal muscle mass parameter (ASSM) and the expression of PTGER4 has been noted (r = -0.224, p = 0.484). PTGES2 and PTGS2 expressions negatively correlated with creatine phosphokinase (r = -0.71, p = 0.009; r = -0.58, p = 0.047) and positively with the functional mobility test timed up and go scale (TUG) (r = 0.61, p = 0.04; r = 0.63, p = 0.032). In the older control group, a negative association between iron levels and the expression of PTGS2 (r = -0.47, p = 0.017) was observed. A similar tendency was noted in patients with sarcopenia (r = -0.112, p = 0.729). A negative trend between appendicular skeletal muscle mass (ASMM) and PTGER4 seems to confirm the impairment of muscle regeneration associated with sarcopenia. The expression of the studied genes revealed a trend in associations with the clinical picture of muscular dystrophy and weakening patients. Perhaps PTGS2 and PTGES2 is in opposition to the role of the PTGER4 receptor in muscle physiology. Nevertheless, further, including functional studies is needed.
Collapse
Affiliation(s)
- Anna Wajda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Diana Bogucka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Barbara Stypińska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Marcin Jerzy Radkowski
- Department of Geriatrics, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (M.J.R.); (T.T.)
| | - Tomasz Targowski
- Department of Geriatrics, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (M.J.R.); (T.T.)
| | - Ewa Dudek
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Tomasz Kmiołek
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Ewa Modzelewska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Agnieszka Paradowska-Gorycka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| |
Collapse
|
2
|
Rey F, Messa L, Maghraby E, Casili G, Ottolenghi S, Barzaghini B, Raimondi MT, Cereda C, Cuzzocrea S, Zuccotti G, Esposito E, Paterniti I, Carelli S. Oxygen Sensing in Neurodegenerative Diseases: Current Mechanisms, Implication of Transcriptional Response, and Pharmacological Modulation. Antioxid Redox Signal 2023; 38:160-182. [PMID: 35793106 DOI: 10.1089/ars.2022.0046] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Significance: Oxygen (O2) sensing is the fundamental process through which organisms respond to changes in O2 levels. Complex networks exist allowing the maintenance of O2 levels through the perception, capture, binding, transport, and delivery of molecular O2. The brain extreme sensitivity to O2 balance makes the dysregulation of related processes crucial players in the pathogenesis of neurodegenerative diseases (NDs). In this study, we wish to review the most relevant advances in O2 sensing in relation to Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Recent Advances: Over the years, it has been clarified that most NDs share common pathways, a great number of which are in relation to O2 imbalance. These include hypoxia, hyperoxia, reactive oxygen species production, metabolism of metals, protein misfolding, and neuroinflammation. Critical Issues: There is still a gap in knowledge concerning how O2 sensing plays a role in the above indicated neurodegenerations. Specifically, O2 concentrations are perceived in body sites that are not limited to the brain, but primarily reside in other organs. Moreover, the mechanisms of O2 sensing, gene expression, and signal transduction seem to correlate with neurodegeneration, but many aspects are mechanistically still unexplained. Future Directions: Future studies should focus on the precise characterization of O2 level disruption and O2 sensing mechanisms in NDs. Moreover, advances need to be made also concerning the techniques used to assess O2 sensing dysfunctions in these diseases. There is also the need to develop innovative therapies targeting this precise mechanism rather than its secondary effects, as early intervention is necessary. Antioxid. Redox Signal. 38, 160-182.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy.,Department of Biomedical and Clinical Sciences, Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milano, Milano, Italy
| | - Letizia Messa
- Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy.,Department of Biomedical and Clinical Sciences, Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milano, Milano, Italy
| | - Erika Maghraby
- Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy.,Department of Biomedical and Clinical Sciences, Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milano, Milano, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Sara Ottolenghi
- Department of Medicine and Surgery, University of Milano Bicocca, Milano, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta," Politecnico di Milano, Milano, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta," Politecnico di Milano, Milano, Italy
| | - Cristina Cereda
- Department of Women, Mothers and Neonatal Care, Children's Hospital "V. Buzzi," Milano, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy.,Department of Biomedical and Clinical Sciences, Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milano, Milano, Italy.,Department of Pediatrics, Children's Hospital "V. Buzzi," Milano, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy.,Department of Biomedical and Clinical Sciences, Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milano, Milano, Italy
| |
Collapse
|
3
|
A functional role of S100A4/non-muscle myosin IIA axis for pro-tumorigenic vascular functions in glioblastoma. Cell Commun Signal 2022; 20:46. [PMID: 35392912 PMCID: PMC8991692 DOI: 10.1186/s12964-022-00848-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive form of brain tumor and has vascular-rich features. The S100A4/non-muscle myosin IIA (NMIIA) axis contributes to aggressive phenotypes in a variety of human malignancies, but little is known about its involvement in GBM tumorigenesis. Herein, we examined the role of the S100A4/NMIIA axis during tumor progression and vasculogenesis in GBM. METHODS We performed immunohistochemistry for S100A4, NMIIA, and two hypoxic markers, hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase 9 (CA9), in samples from 94 GBM cases. The functional impact of S100A4 knockdown and hypoxia were also assessed using a GBM cell line. RESULTS In clinical GBM samples, overexpression of S100A4 and NMIIA was observed in both non-pseudopalisading (Ps) and Ps (-associated) perinecrotic lesions, consistent with stabilization of HIF-1α and CA9. CD34(+) microvascular densities (MVDs) and the interaction of S100A4 and NMIIA were significantly higher in non-Ps perinecrotic lesions compared to those in Ps perinecrotic areas. In non-Ps perinecrotic lesions, S100A4(+)/HIF-1α(-) GBM cells were recruited to the surface of preexisting host vessels in the vascular-rich areas. Elevated vascular endothelial growth factor A (VEGFA) mRNA expression was found in S100A4(+)/HIF-1α(+) GBM cells adjacent to the vascular-rich areas. In addition, GBM patients with high S100A4 protein expression had significantly worse OS and PFS than did patients with low S100A4 expression. Knockdown of S100A4 in the GBM cell line KS-1 decreased migration capability, concomitant with decreased Slug expression; the opposite effects were elicited by blebbistatin-dependent inhibition of NMIIA. CONCLUSION S100A4(+)/HIF-1α(-) GBM cells are recruited to (and migrate along) preexisting vessels through inhibition of NMIIA activity. This is likely stimulated by extracellular VEGF that is released by S100A4(+)/HIF-1α(+) tumor cells in non-Ps perinecrotic lesions. In turn, these events engender tumor progression via acceleration of pro-tumorigenic vascular functions. Video abstract.
Collapse
|
4
|
The hypoxia-inducible factor prolyl hydroxylase inhibitor FG4592 promotes natriuresis through upregulation of COX2 in the renal medulla. Hypertens Res 2022; 45:814-823. [PMID: 35304594 DOI: 10.1038/s41440-022-00889-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/03/2021] [Accepted: 01/09/2022] [Indexed: 11/08/2022]
Abstract
The renal medulla is a key site for the regulation of renal sodium excretion. However, the molecular mechanism remains unclear. Cyclooxygenase 2 (COX2) is specifically expressed in the renal medulla and contributes to the maintenance of the electrolyte/water balance in the body. Hypoxia-inducible factors (HIFs) have also been found to be expressed in the renal medulla, probably owing to the hypoxic conditions in the renal medulla. This study was designed to test the effects of HIF activation on renal sodium handling and renal medullary COX2 expression. Our data showed that HIF activation by the prolyl hydroxylase inhibitor (PHI) FG4592 enhanced natriuresis in mice challenged with a high-salt diet. In addition, FG4592 upregulated the expression of COX2 in the renal medulla. An in vitro study further supported the finding that HIF can induce the expression of COX2 and that this induction is mediated through direct binding to the promoter region of the Cox2 gene, facilitating its transcription. In addition, the COX2 inhibitor celecoxib diminished the natriuretic effect of FG4592. Together, these results suggest that HIF activation promotes sodium excretion through upregulation of COX2 in the renal medulla and therefore maintains sodium homeostasis in the body.
Collapse
|
5
|
Borkowski LF, Keilholz AN, Smith CL, Canda KA, Nichols NL. Nonsteroidal anti-inflammatory drug (ketoprofen) delivery differentially impacts phrenic long-term facilitation in rats with motor neuron death induced by intrapleural CTB-SAP injections. Exp Neurol 2022; 347:113892. [PMID: 34634309 PMCID: PMC10805451 DOI: 10.1016/j.expneurol.2021.113892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 09/05/2021] [Accepted: 10/05/2021] [Indexed: 11/22/2022]
Abstract
Intrapleural injections of cholera toxin B conjugated to saporin (CTB-SAP) selectively eliminates respiratory (e.g., phrenic) motor neurons, and mimics motor neuron death and respiratory deficits observed in rat models of neuromuscular diseases. Additionally, microglial density increases in the phrenic motor nucleus following CTB-SAP. This CTB-SAP rodent model allows us to study the impact of motor neuron death on the output of surviving phrenic motor neurons, and the underlying mechanisms that contribute to enhancing or constraining their output at 7 days (d) or 28d post-CTB-SAP injection. 7d CTB-SAP rats elicit enhanced phrenic long-term facilitation (pLTF) through the Gs-pathway (inflammation-resistant in naïve rats), while pLTF is elicited though the Gq-pathway (inflammation-sensitive in naïve rats) in control and 28d CTB-SAP rats. In 7d and 28d male CTB-SAP rats and controls, we evaluated the effect of cyclooxygenase-1/2 enzymes on pLTF by delivery of the nonsteroidal anti-inflammatory drug, ketoprofen (IP), and we hypothesized that pLTF would be unaffected by ketoprofen in 7d CTB-SAP rats, but pLTF would be enhanced in 28d CTB-SAP rats. In anesthetized, paralyzed and ventilated rats, pLTF was surprisingly attenuated in 7d CTB-SAP rats and enhanced in 28d CTB-SAP rats (both p < 0.05) following ketoprofen delivery. Additionally in CTB-SAP rats: 1) microglia were more amoeboid in the phrenic motor nucleus; and 2) cervical spinal inflammatory-associated factor expression (TNF-α, BDNF, and IL-10) was increased vs. controls in the absence of ketoprofen (p < 0.05). Following ketoprofen delivery, TNF-α and IL-10 expression was decreased back to control levels, while BDNF expression was differentially affected over the course of motor neuron death in CTB-SAP rats. This study furthers our understanding of factors (e.g., cyclooxygenase-1/2-induced inflammation) that contribute to enhancing or constraining pLTF and its implications for breathing following respiratory motor neuron death.
Collapse
Affiliation(s)
- Lauren F Borkowski
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Amy N Keilholz
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Catherine L Smith
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Kaylie A Canda
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Nicole L Nichols
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
6
|
Wang W, Tang J, Zhong M, Chen J, Li T, Dai Y. HIF-1 α may play a role in late pregnancy hypoxia-induced autism-like behaviors in offspring rats. Behav Brain Res 2021; 411:113373. [PMID: 34048873 DOI: 10.1016/j.bbr.2021.113373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that can be caused by various factors. The present study aimed to determine whether prenatal hypoxia can lead to ASD and the role of hypoxia-inducible factor-1α (HIF-1α) in this process. We constructed a prenatal hypoxia model of pregnant rats by piping nitrogen and oxygen mixed gas, with an oxygen concentration of 10 ± 0.5 %, into the self-made hypoxia chamber. Rats were subjected to different extents of hypoxia treatments at different points during pregnancy. The results showed that hypoxia for 6 h on the 17th gestation day is most likely to lead to autistic behavior in offspring rats, including social deficits, repetitive behaviors, and impaired learning and memory. The mRNA expression level of TNF-α also increased in hypoxia-induced autism group and valproic acid (VPA) group. Western blotting analysis showed increased levels of hypoxia inducible factor 1 alpha (HIF-1α) and decreased levels of phosphatase and tensin homolog (PTEN) in the hypoxic-induced autism group. Meanwhile, N-methyl d-aspartate receptor subtype 2 (NR2A) and glutamate ionotropic receptor AMPA type subunit 2 (GluR2) were upregulated in the hypoxic-induced autism group. HIF-1α might play a role in hypoxia-caused autism-like behavior and its regulatory effect is likely to be achieved by regulating synaptic plasticity.
Collapse
Affiliation(s)
- Weiyu Wang
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jinghua Tang
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Min Zhong
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jie Chen
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Ying Dai
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
| |
Collapse
|
7
|
Zhang Z, Song Y, Wang SI, Ha SH, Jang KY, Park BH, Moon YJ, Kim JR. Osteoblasts/Osteocytes sirtuin6 Is Vital to Preventing Ischemic Osteonecrosis Through Targeting VDR-RANKL Signaling. J Bone Miner Res 2021; 36:579-590. [PMID: 33147653 DOI: 10.1002/jbmr.4207] [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: 04/06/2020] [Revised: 10/20/2020] [Accepted: 11/01/2020] [Indexed: 12/15/2022]
Abstract
Ischemic osteonecrosis (ION) can produce permanent deformity and osteoarthritis in the femoral head and other joints. No biologic treatment has been established, and the molecular mechanisms involved in the pathogenesis of ION have not been elucidated. In this work, we found that treatment with sirtuin6 (Sirt6) suppressed inflammatory cytokines, bone resorption, progression of osteoarthritis, and reduced bone deformity in an ION mouse model. We used a deacetylase mutant adenovirus to confirm that those effects were caused by the deacetylase function of Sirt6. Among the osteoclastogenic factors of osteoblasts, only the receptor activator of NF-κb ligand (RANKL) level changed in response to Sirt6 knockout in primary osteoblasts. In particular, the vitamin D receptor physically interacted with Sirt6 and induced recruitment of Sirt6 around RANKL promoters. Finally, Tg mice overexpressing Sirt6 resisted osteocyte death, bone resorption, and progression of osteoarthritis after ischemic surgery, whereas osteoblast/osteocyte-specific Sirt6 knockout mice showed aggravated bone loss and severe deformity. Our findings demonstrate that administration of Sirt6 prevents bone loss and osteoarthritis in ischemic conditions. Activation of Sirt6 in osteoblasts/osteocytes could be a new therapeutic approach to treating ION of the femoral head and other bone regions. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Zhongkai Zhang
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Yiping Song
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Sung Il Wang
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Sang Hoon Ha
- Division of Biotechnology, Chonbuk National University, Iksan, Republic of Korea
| | - Kyu Yun Jang
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea.,Department of Pathology, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Young Jae Moon
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea.,Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Republic of Korea.,Department of Orthopaedic Surgery, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Jung Ryul Kim
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Jeonju, Republic of Korea.,Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| |
Collapse
|
8
|
Ha JS, Choi HR, Kim IS, Kim EA, Cho SW, Yang SJ. Hypoxia-Induced S100A8 Expression Activates Microglial Inflammation and Promotes Neuronal Apoptosis. Int J Mol Sci 2021; 22:1205. [PMID: 33530496 PMCID: PMC7866104 DOI: 10.3390/ijms22031205] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/06/2021] [Accepted: 01/18/2021] [Indexed: 12/31/2022] Open
Abstract
S100 calcium-binding protein A8 (S100A8), a danger-associated molecular pattern, has emerged as an important mediator of the pro-inflammatory response. Some S100 proteins play a prominent role in neuroinflammatory disorders and increase the secretion of pro-inflammatory cytokines in microglial cells. The aim of this study was to determine whether S100A8 induced neuronal apoptosis during cerebral hypoxia and elucidate its mechanism of action. In this study, we reported that the S100A8 protein expression was increased in mouse neuronal and microglial cells when exposed to hypoxia, and induced neuroinflammation and neuronal apoptosis. S100A8, secreted from neurons under hypoxia, activated the secretion of tumor necrosis factor (TNF-α) and interleukin-6 (IL-6) through phosphorylation of extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in microglia. Also, phosphorylation of ERK via the TLR4 receptor induced the priming of the NLRP3 inflammasome. The changes in Cyclooxygenase-2 (COX-2) expression, a well-known inflammatory activator, were regulated by the S100A8 expression in microglial cells. Knockdown of S100A8 levels by using shRNA revealed that microglial S100A8 expression activated COX-2 expression, leading to neuronal apoptosis under hypoxia. These results suggested that S100A8 may be an important molecule for bidirectional microglia-neuron communication and a new therapeutic target for neurological disorders caused by microglial inflammation during hypoxia.
Collapse
Affiliation(s)
- Ji Sun Ha
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (J.S.H.); (H.-R.C.)
| | - Hye-Rim Choi
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (J.S.H.); (H.-R.C.)
| | - In Sik Kim
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Uijeongbu 11759, Korea;
| | - Eun-A Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (J.S.H.); (H.-R.C.)
| |
Collapse
|
9
|
Yang HZ, Zhou XH. Mechanism for hypoxia inducible factor-1α to promote immune escape and therapeutic tolerance in hepatocellular carcinoma under hypoxic microenvironment. Shijie Huaren Xiaohua Zazhi 2020; 28:904-913. [DOI: 10.11569/wcjd.v28.i18.904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The liver is the largest immune organ in the body, and immunologic tolerance and escape mechanisms play an important role in hepatocellular carcinoma (HCC) development. HCC has a complex tumor microenvironment (TME), and it is necessary to study the mechanism that causes HCC cells to escape the body immune surveillance and produce therapeutic resistance in HCC clinical treatment. Hypoxia inducible factor-1α (HIF-1α) is a transcription factor that contains α subunits regulated by hypoxia. Tumor cells highly express HIF-1α in a hypoxic environment, which participates in the processes of tumor cell proliferation and metastasis, microvascular production, immune escape, and therapeutic tolerance, ultimately promoting tumorigenesis and development. In this paper, we will elaborate on the mechanisms by which HCC cells activate HIF-1α expression to promote hypoxic adaptation in cancer cells and regulate immune escape and treatment tolerance in hypoxic TME.
Collapse
Affiliation(s)
- Huan-Zhen Yang
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xi-Han Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical College Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
10
|
Geng C, Li X, Li Y, Song S, Wang C. Nonsteroidal anti-inflammatory drugs alleviate severity of post-endoscopic retrograde cholangiopancreatography pancreatitis by inhibiting inflammation and reducing apoptosis. J Gastroenterol Hepatol 2020; 35:896-904. [PMID: 32064683 DOI: 10.1111/jgh.15012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/08/2020] [Accepted: 02/14/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIM The prophylactic effect of nonselective nonsteroidal anti-inflammatory drugs on post-ERCP (endoscopic retrograde cholangiopancreatography) pancreatitis has been observed for a long time. However, whether the selective nonsteroidal anti-inflammatory drugs possess similar abilities and the mechanisms by which nonsteroidal anti-inflammatory drugs work remain unclear. The present study aimed to determine the protective effects of nonsteroidal anti-inflammatory drugs on post-ERCP pancreatitis in a rat model and examine underlying mechanisms. METHODS Thirty-two female rats were equally and randomly divided into four groups: the sham group, post-ERCP pancreatitis model group, indomethacin-pretreated group, and parecoxib-pretreated group. Indomethacin or parecoxib was delivered 30 min prior to surgery; 24 h after post-ERCP pancreatitis establishment, the rats were sacrificed. Serum amylase and lipase activities, inflammatory cytokine release, pancreatic histopathological scores, neutrophil infiltration, and the expression pattern cyclooxygenase at the protein level and pancreatic apoptosis were quantified and analyzed. RESULTS Both indomethacin and parecoxib inhibited the activities of serum amylase and lipase and reduced the severity of pancreatic histopathology. Mechanistically, both drugs decreased the expression level of cyclooxygenase 2; however, they had no influence on the cyclooxygenase 1 protein level. Moreover, they reduced inflammatory cytokine release, neutrophil infiltration into the pancreas, and NF-κB p65 activation. Notably, we found that apoptotic cells in the pancreas were remarkably diminished after the administration of both nonsteroidal anti-inflammatory drugs. CONCLUSIONS Both selective and nonselective nonsteroidal anti-inflammatory drugs exert protective effects against post-ERCP pancreatitis by restraining inflammation and reducing acinar cell apoptosis through the inhibition of cyclooxygenase 2.
Collapse
Affiliation(s)
- Chong Geng
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiao Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Yanni Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Shuailing Song
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chunhui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part II - Modulation of angiogenesis. Clin Hemorheol Microcirc 2020; 73:409-438. [PMID: 31177206 DOI: 10.3233/ch-199103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The treatment of critical-size bone defects following complicated fractures, infections or tumor resections is a major challenge. The same applies to fractures in patients with impaired bone healing due to systemic inflammatory and metabolic diseases. Despite considerable progress in development and establishment of new surgical techniques, design of bone graft substitutes and imaging techniques, these scenarios still represent unresolved clinical problems. However, the development of new active substances offers novel potential solutions for these issues. This work discusses therapeutic approaches that influence angiogenesis or hypoxic situations in healing bone and surrounding tissue. In particular, literature on sphingosine-1-phosphate receptor modulators and nitric oxide (NO•) donors, including bi-functional (hybrid) compounds like NO•-releasing cyclooxygenase-2 inhibitors, was critically reviewed with regard to their local and systemic mode of action.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
12
|
Lewis A, Elks PM. Hypoxia Induces Macrophage tnfa Expression via Cyclooxygenase and Prostaglandin E2 in vivo. Front Immunol 2019; 10:2321. [PMID: 31611882 PMCID: PMC6776637 DOI: 10.3389/fimmu.2019.02321] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/13/2019] [Indexed: 01/25/2023] Open
Abstract
Macrophage phenotypes are poorly characterized in disease systems in vivo. Appropriate macrophage activation requires complex coordination of local microenvironmental cues and cytokine signaling. If the molecular mechanisms underpinning macrophage activation were better understood, macrophages could be pharmacologically tuned during disease situations. Here, using zebrafish tnfa:GFP transgenic lines as in vivo readouts, we show that physiological hypoxia and stabilization of Hif-1α promotes macrophage tnfa expression. We demonstrate a new mechanism of Hif-1α-induced macrophage tnfa expression via a cyclooxygenase/prostaglandin E2 axis. These findings uncover a macrophage HIF/COX/TNF axis that links microenvironmental cues to macrophage phenotype, with important implications during inflammation, infection, and cancer, where hypoxia is a common microenvironmental feature and where cyclooxygenase and TNF are major mechanistic players.
Collapse
Affiliation(s)
| | - Philip M. Elks
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
13
|
Lin CH, Liao LY, Yang TY, Chang YJ, Tung CW, Hsu SL, Hsueh CM. Microglia-Derived Adiposomes are Potential Targets for the Treatment of Ischemic Stroke. Cell Mol Neurobiol 2019; 39:591-604. [PMID: 30852719 DOI: 10.1007/s10571-019-00665-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 02/19/2019] [Indexed: 12/28/2022]
Abstract
It is known that cerebral ischemia can cause brain inflammation and adiposome can serve as a depot of inflammatory mediators. In the study, the pro-inflammatory and pro-death role of adiposome in ischemic microglia and ischemic brain was newly investigated. The contribution of PPARγ to adiposome formation was also evaluated for the first time in ischemic microglia. Focal cerebral ischemia/reperfusion (I/R) animal model and the in vitro glucose-oxygen-serum deprivation (GOSD) cell model were both applied in the study. GOSD- or I/R-induced adiposome formation, inflammatory activity, cell death of microglia, and brain infarction were, respectively, determined, in the absence or presence of NS-398 (adiposome inhibitor) or GW9662 (PPARγ antagonist). GOSD-increased adiposome formation played a critical role in stimulating the inflammatory activity (production of TNF-α and IL-1β) and cell death of microglia. Similar results were also found in ischemic brain tissues. GOSD-induced PPARγ partially contributed to the increase of adiposomes and adiposome-mediated inflammatory responses of microglia. Blockade of adiposome formation with NS-398 or GW9662 significantly reduced not only the inflammatory activity and death rate of GOSD-treated microglia but also the brain infarct volume and motor function deficit of ischemic rats. The pathological role of microglia-derived adiposome in cerebral ischemia has been confirmed and attributed to its pro-inflammatory and/or pro-death effect upon ischemic brain cells and tissues. Adiposome and its upstream regulator PPARγ were therefore as potential targets for the treatment of ischemic stroke. Therapeutic values of NS-398 and GW9662 have been suggested.
Collapse
Affiliation(s)
- Chi-Hsin Lin
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City, Taiwan
| | - Li-Ya Liao
- Department of Life Sciences, National Chung Hsing University, 145, Xingda Road, Taichung, 402, Taiwan, ROC
| | - Tsung-Ying Yang
- Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Jyun Chang
- Department of Life Sciences, National Chung Hsing University, 145, Xingda Road, Taichung, 402, Taiwan, ROC
| | - Chia-Wen Tung
- Department of Life Sciences, National Chung Hsing University, 145, Xingda Road, Taichung, 402, Taiwan, ROC
| | - Shih-Lan Hsu
- Department of Life Sciences, National Chung Hsing University, 145, Xingda Road, Taichung, 402, Taiwan, ROC
| | - Chi-Mei Hsueh
- Department of Life Sciences, National Chung Hsing University, 145, Xingda Road, Taichung, 402, Taiwan, ROC.
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, ROC.
| |
Collapse
|
14
|
Targeting Cyclooxygenase-2 in Pheochromocytoma and Paraganglioma: Focus on Genetic Background. Cancers (Basel) 2019; 11:cancers11060743. [PMID: 31142060 PMCID: PMC6627450 DOI: 10.3390/cancers11060743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 02/08/2023] Open
Abstract
Cyclooxygenase 2 (COX-2) is a key enzyme of the tumorigenesis-inflammation interface and can be induced by hypoxia. A pseudohypoxic transcriptional signature characterizes pheochromocytomas and paragangliomas (PPGLs) of the cluster I, mainly represented by tumors with mutations in von Hippel–Lindau (VHL), endothelial PAS domain-containing protein 1 (EPAS1), or succinate dehydrogenase (SDH) subunit genes. The aim of this study was to investigate a possible association between underlying tumor driver mutations and COX-2 in PPGLs. COX-2 gene expression and immunoreactivity were examined in clinical specimens with documented mutations, as well as in spheroids and allografts derived from mouse pheochromocytoma (MPC) cells. COX-2 in vivo imaging was performed in allograft mice. We observed significantly higher COX-2 expression in cluster I, especially in VHL-mutant PPGLs, however, no specific association between COX-2 mRNA levels and a hypoxia-related transcriptional signature was found. COX-2 immunoreactivity was present in about 60% of clinical specimens as well as in MPC spheroids and allografts. A selective COX-2 tracer specifically accumulated in MPC allografts. This study demonstrates that, although pseudohypoxia is not the major determinant for high COX-2 levels in PPGLs, COX-2 is a relevant molecular target. This potentially allows for employing selective COX-2 inhibitors as targeted chemotherapeutic agents and radiosensitizers. Moreover, available models are suitable for preclinical testing of these treatments.
Collapse
|
15
|
Paardekooper LM, Vos W, van den Bogaart G. Oxygen in the tumor microenvironment: effects on dendritic cell function. Oncotarget 2019; 10:883-896. [PMID: 30783517 PMCID: PMC6368231 DOI: 10.18632/oncotarget.26608] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Solid tumors grow at a high speed leading to insufficient blood supply to tumor cells. This makes the tumor hypoxic, resulting in the Warburg effect and an increased generation of reactive oxygen species (ROS). Hypoxia and ROS affect immune cells in the tumor micro-environment, thereby affecting their immune function. Here, we review the known effects of hypoxia and ROS on the function and physiology of dendritic cells (DCs). DCs can (cross-)present tumor antigen to activate naive T cells, which play a pivotal role in anti-tumor immunity. ROS might enter DCs via aquaporins in the plasma membrane, diffusion across the plasma membrane or via extracellular vesicles (EVs) released by tumor cells. Hypoxia and ROS exert complex effects on DCs, and can both inhibit and activate maturation of immature DCs. Furthermore, ROS transferred by EVs and/or produced by the DC can both promote antigen (cross-)presentation through phagosomal alkalinization, which preserves antigens by inhibiting proteases, and by direct oxidative modification of proteases. Hypoxia leads to a more migratory and inflammatory DC phenotype. Lastly, hypoxia alters DCs to shift the T- cell response towards a tumor suppressive Th17 phenotype. From numerous studies, the concept is emerging that hypoxia and ROS are mutually dependent effectors on DC function in the tumor micro-environment. Understanding their precise roles and interplay is important given that an adaptive immune response is required to clear tumor cells.
Collapse
Affiliation(s)
- Laurent M Paardekooper
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willemijn Vos
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
16
|
Hypoxia potentiates monocyte-derived dendritic cells for release of tumor necrosis factor α via MAP3K8. Biosci Rep 2018; 38:BSR20182019. [PMID: 30463908 PMCID: PMC6294625 DOI: 10.1042/bsr20182019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) constantly sample peripheral tissues for antigens, which are subsequently ingested to derive peptides for presentation to T cells in lymph nodes. To do so, DCs have to traverse many different tissues with varying oxygen tensions. Additionally, DCs are often exposed to low oxygen tensions in tumors, where vascularization is lacking, as well as in inflammatory foci, where oxygen is rapidly consumed by inflammatory cells during the respiratory burst. DCs respond to oxygen levels to tailor immune responses to such low-oxygen environments. In the present study, we identified a mechanism of hypoxia-mediated potentiation of release of tumor necrosis factor α (TNF-α), a pro-inflammatory cytokine with important roles in both anti-cancer immunity and autoimmune disease. We show in human monocyte-derived DCs (moDCs) that this potentiation is controlled exclusively via the p38/mitogen-activated protein kinase (MAPK) pathway. We identified MAPK kinase kinase 8 (MAP3K8) as a target gene of hypoxia-induced factor (HIF), a transcription factor controlled by oxygen tension, upstream of the p38/MAPK pathway. Hypoxia increased expression of MAP3K8 concomitant with the potentiation of TNF-α secretion. This potentiation was no longer observed upon siRNA silencing of MAP3K8 or with a small molecule inhibitor of this kinase, and this also decreased p38/MAPK phosphorylation. However, expression of DC maturation markers CD83, CD86, and HLA-DR were not changed by hypoxia. Since DCs play an important role in controlling T-cell activation and differentiation, our results provide novel insight in understanding T-cell responses in inflammation, cancer, autoimmune disease and other diseases where hypoxia is involved.
Collapse
|
17
|
Shupp AB, Kolb AD, Mukhopadhyay D, Bussard KM. Cancer Metastases to Bone: Concepts, Mechanisms, and Interactions with Bone Osteoblasts. Cancers (Basel) 2018; 10:E182. [PMID: 29867053 PMCID: PMC6025347 DOI: 10.3390/cancers10060182] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
The skeleton is a unique structure capable of providing support for the body. Bone resorption and deposition are controlled in a tightly regulated balance between osteoblasts and osteoclasts with no net bone gain or loss. However, under conditions of disease, the balance between bone resorption and deposition is upset. Osteoblasts play an important role in bone homeostasis by depositing new bone osteoid into resorption pits. It is becoming increasingly evident that osteoblasts additionally play key roles in cancer cell dissemination to bone and subsequent metastasis. Our laboratory has evidence that when osteoblasts come into contact with disseminated breast cancer cells, the osteoblasts produce factors that initially reduce breast cancer cell proliferation, yet promote cancer cell survival in bone. Other laboratories have demonstrated that osteoblasts both directly and indirectly contribute to dormant cancer cell reactivation in bone. Moreover, we have demonstrated that osteoblasts undergo an inflammatory stress response in late stages of breast cancer, and produce inflammatory cytokines that are maintenance and survival factors for breast cancer cells and osteoclasts. Advances in understanding interactions between osteoblasts, osteoclasts, and bone metastatic cancer cells will aid in controlling and ultimately preventing cancer cell metastasis to bone.
Collapse
Affiliation(s)
- Alison B Shupp
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Alexus D Kolb
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Dimpi Mukhopadhyay
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Karen M Bussard
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
18
|
Liu C, Liu B, Liu L, Zhang EL, Sun BD, Xu G, Chen J, Gao YQ. Arachidonic Acid Metabolism Pathway Is Not Only Dominant in Metabolic Modulation but Associated With Phenotypic Variation After Acute Hypoxia Exposure. Front Physiol 2018; 9:236. [PMID: 29615930 PMCID: PMC5864929 DOI: 10.3389/fphys.2018.00236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
Background: The modulation of arachidonic acid (AA) metabolism pathway is identified in metabolic alterations after hypoxia exposure, but its biological function is controversial. We aimed at integrating plasma metabolomic and transcriptomic approaches to systematically explore the roles of the AA metabolism pathway in response to acute hypoxia using an acute mountain sickness (AMS) model. Methods: Blood samples were obtained from 53 enrolled subjects before and after exposure to high altitude. Ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry and RNA sequencing were separately performed for metabolomic and transcriptomic profiling, respectively. Influential modules comprising essential metabolites and genes were identified by weighted gene co-expression network analysis (WGCNA) after integrating metabolic information with phenotypic and transcriptomic datasets, respectively. Results: Enrolled subjects exhibited diverse response manners to hypoxia. Combined with obviously altered heart rate, oxygen saturation, hemoglobin, and Lake Louise Score (LLS), metabolomic profiling detected that 36 metabolites were highly related to clinical features in hypoxia responses, out of which 27 were upregulated and nine were downregulated, and could be mapped to AA metabolism pathway significantly. Integrated analysis of metabolomic and transcriptomic data revealed that these dominant molecules showed remarkable association with genes in gas transport incapacitation and disorders of hemoglobin metabolism pathways, such as ALAS2, HEMGN. After detailed description of AA metabolism pathway, we found that the molecules of 15-d-PGJ2, PGA2, PGE2, 12-O-3-OH-LTB4, LTD4, LTE4 were significantly up-regulated after hypoxia stimuli, and increased in those with poor response manner to hypoxia particularly. Further analysis in another cohort showed that genes in AA metabolism pathway such as PTGES, PTGS1, GGT1, TBAS1 et al. were excessively elevated in subjects in maladaptation to hypoxia. Conclusion: This is the first study to construct the map of AA metabolism pathway in response to hypoxia and reveal the crosstalk between phenotypic variation under hypoxia and the AA metabolism pathway. These findings may improve our understanding of the advanced pathophysiological mechanisms in acute hypoxic diseases and provide new insights into critical roles of the AA metabolism pathway in the development and prevention of these diseases.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Bao Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China.,The 12th Hospital of Chinese People's Liberation Army, Kashi, China
| | - Lu Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Er-Long Zhang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Bind-da Sun
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Jian Chen
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| |
Collapse
|
19
|
Kim YE, Lee M, Gu H, Kim J, Jeong S, Yeo S, Lee YJ, Im SH, Sung YC, Kim HJ, Weissman IL, Ahn GO. Hypoxia-inducible factor-1 (HIF-1) activation in myeloid cells accelerates DSS-induced colitis progression in mice. Dis Model Mech 2018; 11:dmm.033241. [PMID: 29967068 PMCID: PMC6078398 DOI: 10.1242/dmm.033241] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 06/25/2018] [Indexed: 12/26/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease, in which the intestinal epithelium loses its barrier function. Given the existence of the oxygen gradient in the intestinal epithelium and that inflammation further contributes to the tissue hypoxia, we investigated the role of hypoxia-inducible factor (HIF), a transcription factor activated under hypoxic conditions in myeloid cells, in the progression of IBD. To do this, we utilized myeloid-specific knockout (KO) mice targeting HIF pathways, created by a Cre-loxP system with human MRP8 (hMRP8), an intracellular calcium-binding protein, as the myeloid promoter. By feeding 5% dextran sodium sulfate (DSS) to hMRP8 von Hippel Lindau (Vhl) KO mice, in which HIF-1α and HIF-2α are constitutively activated in myeloid cells, we found that these mice were highly susceptible to DSS-induced colitis, demonstrating greater body weight loss, increased mortality, faster onset of rectal bleeding, shortened colon length, and increased CD11b- or Gr-1-positive myeloid cells in the colon compared with wild-type (WT) mice. These parameters were restored to, if not better than, the WT levels when we examined hMRP8 Hif-1a KO mice upon 5% DSS feeding. hMRP8 Hif-2a KO mice, on the other hand, exhibited a similar degree of DSS-induced colitis to that of WT mice. Lastly, when DSS was given together with azoxymethane to induce tumorigenesis in the colon, we found that hMRP8 Hif-1a KO mice exhibited comparable levels of colorectal tumors to those of WT mice, indicating that HIF-1α in myeloid cells is dispensable for tumorigenesis. Collectively, our results suggest that HIF-1α activation in myeloid cells critically regulates IBD progression. Summary: We challenged myeloid-specific knockout mice targeting the hypoxia-inducible factor (HIF) pathway to dextran sodium sulfate-induced colitis, demonstrating that HIF-1α, but not HIF-2α, activation in myeloid cells regulates colitis severity in mice.
Collapse
Affiliation(s)
- Young-Eun Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Minji Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Gyeongbuk 37673, Korea
| | - Hyejung Gu
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Jeongwoo Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Seongju Jeong
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Sujin Yeo
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - You Jeong Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Gyeongbuk 37673, Korea
| | - Sin-Hyeog Im
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Gyeongbuk 37673, Korea
| | - Young-Chul Sung
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Irving L. Weissman
- Stem Cell Institute and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - G-One Ahn
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| |
Collapse
|
20
|
Lin CL, Hsiao CJ, Hsu CH, Wang SE, Jen PHS, Wu CH. Hypothermic neuroprotections in the brain of an echolocation bat, Hipposideros terasensis. Neuroreport 2017; 28:956-962. [PMID: 28914739 DOI: 10.1097/wnr.0000000000000856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The present study aimed to investigate how bats protect their brain in a hypothermic situation. Formosan leaf-nosed bats (Hipposideros terasensis) were used in this study and treated under three conditions: room temperature (25±1°C), low temperature (4±1°C), and hibernation. The reactive oxygen species (ROS) levels in the blood and apoptosis-related proteins in the brain tissue were assessed and then compared among those bats under three conditions. Our results showed that the blood ROS levels of bats treated under conditions of low temperature and hibernation were significantly reduced compared with bats treated under the condition of room temperature. Both immunohistochemistry and immunoblotting expressions of hypoxia, inflammation, and apoptosis-related proteins in the brain tissue of bats treated under the condition of hibernation were significantly reduced compared with those bats treated under conditions of room temperature and low temperature. Thus, we suggested that bats can protect the brain in cold environment by reducing blood ROS levels and decreasing expressions of hypoxia, inflammation, and apoptosis-related proteins in the brain. Possible protection mechanisms involved in hypothermic adaptations need to be further clarified.
Collapse
Affiliation(s)
- Ching-Lung Lin
- aDepartment of Life Sciences, National Taiwan Normal University, Taipei, Taiwan bDepartment of Neuroscience, Division of Biological Sciences, University of Missouri, Columbia, Missouri, USA
| | | | | | | | | | | |
Collapse
|
21
|
Wang Y, Guan PP, Yu X, Guo YS, Zhang YJ, Wang ZY, Wang P. COX-2 metabolic products, the prostaglandin I 2 and F 2α, mediate the effects of TNF-α and Zn 2+ in stimulating the phosphorylation of Tau. Oncotarget 2017; 8:99296-99311. [PMID: 29245902 PMCID: PMC5725093 DOI: 10.18632/oncotarget.21853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022] Open
Abstract
Although the roles of cyclooxygenase-2 (COX-2) and prostaglandins (PGs) in regulating amyloid precursor protein (APP) cleavage and β-amyloid protein (Aβ) production have been the subjects of numerous investigations, their effects on tau phosphorylation have been largely overlooked. Using human TauP301S transgenic (Tg) mice as in vivo model, our results demonstrated that PGI2 and PGF2α mediated the effects of tumor necrosis factor α (TNF-α) and Zinc ions (Zn2+) on upregulating the phosphorylation of tau via the PI3-K/AKT, ERK1/2 and JNK/c-Jun signaling pathways. Specifically, we initially found that high level of Zn2+ upregulates the expression of COX-2 via stimulating the activity of TNF-α in a zinc transporter 3 (ZnT3)-dependent mechanism. COX-2 upregulation then stimulates the phosphorylation of tau at both Ser 202 and Ser 400/Thr 403/Ser 404 via PGI2 and F2α treatment either in i.c.v.-injected mice or in n2a cells. Using n2a cells as in vitro model, we further revealed critical roles for the PI3-K/AKT, ERK1/2 and JNK/c-Jun pathways in mediating the effects of PGI2 and F2α in the phosphorylation of tau. Finally, NS398 treatment delayed the onset of cognitive decline in TauP301S Tg mice according to the nest construction or limb clasping test.
Collapse
Affiliation(s)
- Yue Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, P.R. China.,Department of Tissue Culture, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, Shenyang, P.R. China
| | - Xin Yu
- College of Life and Health Sciences, Northeastern University, Shenyang, P.R. China
| | - Yan-Su Guo
- Key laboratory of Hebei Neurology, Hebei Medical University, Shijiazhuang, P.R. China.,Institute of Cardiocerebrovascular Disease, Hebei Medical University, Shijiazhuang, P.R. China
| | - Ying-Jie Zhang
- College of Biology, Hunan University, Changsha, P.R. China.,Shenzhen Institute, Hunan University, Shenzhen, P.R. China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, P.R. China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, P.R. China
| |
Collapse
|
22
|
HIF-1α induction during reperfusion avoids maladaptive repair after renal ischemia/reperfusion involving miR127-3p. Sci Rep 2017; 7:41099. [PMID: 28106131 PMCID: PMC5247697 DOI: 10.1038/srep41099] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/15/2016] [Indexed: 12/11/2022] Open
Abstract
Ischemia/reperfusion (I/R) leads to Acute Kidney Injury. HIF-1α is a key factor during organ response to I/R. We previously demonstrated that HIF-1α is induced during renal reperfusion, after ischemia. Here we investigate the role of HIF-1α and the HIF-1α dependent mechanisms in renal repair after ischemia. By interference of HIF-1α in a rat model of renal I/R, we observed loss of expression and mis-localization of e-cadherin and induction of α-SMA, MMP-13, TGFβ, and collagen I. Moreover, we demonstrate that HIF-1α inhibition promotes renal cell infiltrates by inducing IL-1β, TNF-α, MCP-1 and VCAM-1, through NFkB activity. In addition, HIF-1α inhibition induced proximal tubule cells proliferation but it did not induce compensatory apoptosis, both in vivo. In vitro, HIF-1α knockdown in HK2 cells subjected to hypoxia/reoxygenation (H/R) promote cell entry into S phase, correlating with in vivo data. HIF-1α interference leads to downregulation of miR-127-3p and induction of its target gene Bcl6 in vivo. Moreover, modulation of miR-127-3p in HK2 cells subjected to H/R results in EMT regulation: miR127-3p inhibition promote loss of e-cadherin and induction of α-SMA and collagen I. In conclusion, HIF-1α induction during reperfusion is a protector mechanism implicated in a normal renal tissue repair after I/R.
Collapse
|
23
|
Davydov DA, Avdalyan AM, Agadzhanyan VV, Lushnikova EL, Ustyantseva IM. [Morphometric and molecular biological features of femoral head tissue in different nosological entities of coxarthrosis]. Arkh Patol 2016; 78:20-26. [PMID: 27804942 DOI: 10.17116/patol201678520-26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
AIM to comparatively analyze the morphometric and molecular biological characteristics of femoral head tissue in different nosological entities of coxarthrosis. MATERIAL AND METHODS A total of 95 samples of femoral head tissue extirpated during hip endoprosthesis in patients with coxarthrosis were investigated. Clinical findings were used to identify the following nosological entities of coxarthrosis: dysplastic, postischemic and posttraumatic. Histological, immunohistochemical and morphometric studies were used. Osteoclast resorptive activity was assessed by determining the cytoplasmic expression of TRAcP (9C5, «Cell Marque»). Vasculogenesis was evaluated by estimating the mean area of vessels with CD34 (QBEnd/10, «Ventana») from the positive stained endothelium and by determining the cytoplasmic expression level of VEGF (SP28, «Spring Bio») in osteoblasts and osteoclasts. RESULTS Specific histopathological signs were described for each nosological entity of coxarthrosis. Morphometric analysis could reveal a number of additional characteristics of the magnitude of fibrous changes and the thickness of the articular surface and bone rods. Immunohistochemical assessment of molecular biological parameters, such as the expression level of VEGF and TRAcP, also pointed to the characteristic features of bony tissue in the above-mentioned nosological entities of coxarthrosis. In dysplastic coxarthrosis, the maximal expression level of VEGF was recorded in osteoblasts and the expression of VEGF and TRAcP in osteoclasts remained at the minimum level. The lowest expression of VEGF in osteoblasts was found in posttraumatic coxarthrosis. In postischemic coxarthrosis, the highest expression of VEGF and TRAcP was recorded in osteoclasts. CONCLUSION The comparative analysis of the morphometric and molecular biological characteristics of femoral head tissue in different nosological entities of coxarthrosis indicated a number of peculiar features. The most specific manifestations of certain morphological and molecular biological signs were identified for each nosological entity of coxarthrosis.
Collapse
Affiliation(s)
- D A Davydov
- Regional Clinical Center for Miners' Health Prote ction, Leninsk-Kuznetsky, Russian Federation
| | - A M Avdalyan
- Laboratory of Tumor Molecular Genetic Characteristics, Altai Branch, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Barnaul, Russian Federation
| | - V V Agadzhanyan
- Regional Clinical Center for Miners' Health Prote ction, Leninsk-Kuznetsky, Russian Federation
| | - E L Lushnikova
- Institute of Molecular Pathology and Pathomorphology, Siberian Branch, Russian Academy of Medical Sciences, Novosibirsk, Russia
| | - I M Ustyantseva
- Regional Clinical Center for Miners' Health Prote ction, Leninsk-Kuznetsky, Russian Federation
| |
Collapse
|