1
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 PMCID: PMC12028135 DOI: 10.3390/ijms26083852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Hasmiq L. Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Birandra K. Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
2
|
Kundi M, Nersesyan A, Schmid G, Hutter HP, Eibensteiner F, Mišík M, Knasmüller S. Mobile phone specific radiation disturbs cytokinesis and causes cell death but not acute chromosomal damage in buccal cells: Results of a controlled human intervention study. ENVIRONMENTAL RESEARCH 2024; 251:118634. [PMID: 38452915 DOI: 10.1016/j.envres.2024.118634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Several human studies indicate that mobile phone specific electromagnetic fields may cause cancer in humans but the underlying molecular mechanisms are currently not known. Studies concerning chromosomal damage (which is causally related to cancer induction) are controversial and those addressing this issue in mobile phone users are based on the use of questionnaires to assess the exposure. We realized the first human intervention trial in which chromosomal damage and acute toxic effects were studied under controlled conditions. The participants were exposed via headsets at one randomly assigned side of the head to low and high doses of a UMTS signal (n = 20, to 0.1 W/kg and n = 21 to 1.6 W/kg Specific Absorption Rate) for 2 h on 5 consecutive days. Before and three weeks after the exposure, buccal cells were collected from both cheeks and micronuclei (MN, which are formed as a consequence of structural and numerical chromosomal aberrations) and other nuclear anomalies reflecting mitotic disturbance and acute cytotoxic effects were scored. We found no evidence for induction of MN and of nuclear buds which are caused by gene amplifications, but a significant increase of binucleated cells which are formed as a consequence of disturbed cell divisions, and of karyolitic cells, which are indicative for cell death. No such effects were seen in cells from the less exposed side. Our findings indicate that mobile phone specific high frequency electromagnetic fields do not cause acute chromosomal damage in oral mucosa cells under the present experimental conditions. However, we found clear evidence for disturbance of the cell cycle and cytotoxicity. These effects may play a causal role in the induction of adverse long term health effects in humans.
Collapse
Affiliation(s)
- Michael Kundi
- Center for Public Health, Department of Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Armen Nersesyan
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Gernot Schmid
- EMC & Optics, Seibersdorf Labor GmbH, 2444 Seibersdorf, Austria
| | - Hans-Peter Hutter
- Center for Public Health, Department of Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Florian Eibensteiner
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Miroslav Mišík
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Siegfried Knasmüller
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria.
| |
Collapse
|
3
|
Lin CY, Tam Ly M, Yang SH, Lai SC, Chang TW, Lin IH, Tzeng YJ. Tanshinone IIA Shows Higher Antiproliferative Activities than Sinapic Acid in 4 Cancer Cell Lines and Simultaneously Induces Apoptosis and Necroptosis in Human Lung Cancer A549 Cells. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211050521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tanshinone IIA (Tan IIA) and sinapic acid (SA) are 2 components separately isolated from 2 Asian medicinal plants, Hydnophytum formicarum Jack and Salvia miltiorrhiza Bunge. The antitumor activities of them were worth exploring, therefore, we examined their antitumor activities in A549, HCT116, HeLa, and Colo320 cancer cell lines by means of WST-1 assay. The results show that Tan IIA exerted far higher (IC50 from 1.0 ± 0.0 to 166.3 ± 24.0 µg/mL) antiproliferative activities than SA (IC50 from 2236.3 ± 484.1 to >10 000.0 µg/mL). Of the 4 cell lines, A549 cells were the most sensitive to Tan IIA; thus, we used Western blotting to explore the cytotoxic mechanisms of Tan IIA in A549 cells and found that they rely on simultaneous induction of apoptosis and necroptosis in the cells. Apoptosis was hallmarked by the induction of cleaved caspase-3 by Tan IIA and necroptosis by the necroptotic marker proteins cyclophilin A and high mobility group box 1 (HMGB1), as well as increased lactate dehydrogenase (LDH) activities. The necroptotic effect was confirmed by the necroptosis inhibitor necrostatin-1 (Nec-1), which eliminated these effects and restored cell survival rates. The levels of cyclophilin A decreased in response to the pan-caspase inhibitor z-VAD-fmk, and those of cleaved caspase-3 decreased in response to Nec-1. Conclusively, Tan IIA has the potential to prevent lung cancer and the mechanism seems to be apoptosis and necroptosis, of which the relationship is mutually interdependent. This is the first report of Tan IIA eliciting necroptosis in A549 cells. Tan IIA may be used for necroptosis-based cancer therapy, especially to overcome apoptosis resistance.
Collapse
Affiliation(s)
- Chueh-Yu Lin
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien City, Taiwan
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien City, Taiwan
| | - Minh Tam Ly
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
| | - Shih Hsien Yang
- Department of Physical Medicine and Rehabilitation, School of Medicine, Tzu Chi University, Hualien City, Taiwan
- Department of Physical Medicine and Rehabilitation, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien City, Taiwan
| | - Shang-Chih Lai
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien City, Taiwan
- Department of Pharmacy of Buddhist Hualien Tzu Chi Hospital, Hualien City, Taiwan
| | - Tung-Wu Chang
- Division of Crop Improvement, Hualien District Agricultural Research and Extension Station, Council of Agriculture, Hualien County, Taiwan
| | - I-Hsin Lin
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien City, Taiwan
- Department of Chinese Medicine of Buddhist Hualien Tzu Chi Hospital, Hualien City, Taiwan
| | - Yin-Jeh Tzeng
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien City, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Life Science, Tzu Chi University, Hualien City, Taiwan
| |
Collapse
|
4
|
Palmblad K, Schierbeck H, Sundberg E, Horne AC, Erlandsson Harris H, Henter JI, Andersson U. Therapeutic administration of etoposide coincides with reduced systemic HMGB1 levels in macrophage activation syndrome. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2021; 27:48. [PMID: 33975537 PMCID: PMC8111379 DOI: 10.1186/s10020-021-00308-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Macrophage activation syndrome (MAS) is a potentially fatal complication of systemic inflammation. HMGB1 is a nuclear protein released extracellularly during proinflammatory lytic cell death or secreted by activated macrophages, NK cells, and additional cell types during infection or sterile injury. Extracellular HMGB1 orchestrates central events in inflammation as a prototype alarmin. TLR4 and the receptor for advanced glycation end products operate as key HMGB1 receptors to mediate inflammation. METHODS Standard ELISA and cytometric bead array-based methods were used to examine the kinetic pattern for systemic release of HMGB1, ferritin, IL-18, IFN-γ, and MCP-1 before and during treatment of four children with critical MAS. Three of the patients with severe underlying systemic rheumatic diseases were treated with biologics including tocilizumab or anakinra when MAS developed. All patients required intensive care therapy due to life-threatening illness. Add-on etoposide therapy was administered due to insufficient clinical response with standard treatment. Etoposide promotes apoptotic rather than proinflammatory lytic cell death, conceivably ameliorating subsequent systemic inflammation. RESULTS This therapeutic intervention brought disease control coinciding with a decline of the increased systemic HMGB1, IFN-γ, IL-18, and ferritin levels whereas MCP-1 levels evolved independently. CONCLUSION Systemic HMGB1 levels in MAS have not been reported before. Our results suggest that the molecule is not merely a biomarker of inflammation, but most likely also contributes to the pathogenesis of MAS. These observations encourage further studies of HMGB1 antagonists. They also advocate therapeutic etoposide administration in severe MAS and provide a possible biological explanation for its mode of action.
Collapse
Affiliation(s)
- Karin Palmblad
- Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Hanna Schierbeck
- Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Erik Sundberg
- Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Anna-Carin Horne
- Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Helena Erlandsson Harris
- Rheumatology Unit, Department of Medicine, Karolinska Institute at Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Jan-Inge Henter
- Childhood Cancer Research Unit, Department of Women׳s and Children׳s Health, Karolinska Institute, 17177, Stockholm, Sweden.,Theme of Children, Karolinska University Hospital, 17176, Solna, Stockholm, Sweden
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
5
|
Zemskova M, McClain N, Niihori M, Varghese MV, James J, Rafikov R, Rafikova O. Necrosis-Released HMGB1 (High Mobility Group Box 1) in the Progressive Pulmonary Arterial Hypertension Associated With Male Sex. Hypertension 2020; 76:1787-1799. [PMID: 33012199 DOI: 10.1161/hypertensionaha.120.16118] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Damage-associated molecular patterns, such as HMGB1 (high mobility group box 1), play a well-recognized role in the development of pulmonary arterial hypertension (PAH), a progressive fatal disease of the pulmonary vasculature. However, the contribution of the particular type of vascular cells, type of cell death, or the form of released HMGB1 in PAH remains unclear. Moreover, although male patients with PAH show a higher level of circulating HMGB1, its involvement in the severe PAH phenotype reported in males is unknown. In this study, we aimed to investigate the sources and active forms of HMGB1 released from damaged vascular cells and their contribution to the progressive type of PAH in males. Our results showed that HMGB1 is released by either pulmonary artery human endothelial cells or human pulmonary artery smooth muscle cells that underwent necrotic cell death, although only human pulmonary artery smooth muscle cells produce HMGB1 during apoptosis. Moreover, only human pulmonary artery smooth muscle cell death induced a release of dimeric HMGB1, found to be mitochondrial reactive oxygen species dependent, and TLR4 (toll-like receptor 4) activation. The modified Sugen/Hypoxia rat model replicates the human sexual dimorphism in PAH severity (right ventricle systolic pressure in males versus females 54.7±2.3 versus 44.6±2 mm Hg). By using this model, we confirmed that necroptosis and necrosis are the primary sources of circulating HMGB1 in the male rats, although only necrosis increased circulation of HMGB1 dimers. Attenuation of necrosis but not apoptosis or necroptosis prevented TLR4 activation in males and blunted the sex differences in PAH severity. We conclude that necrosis, through the release of HMGB1 dimers, predisposes males to a progressive form of PAH.
Collapse
Affiliation(s)
- Marina Zemskova
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Nolan McClain
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Maki Niihori
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Mathews V Varghese
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Joel James
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Ruslan Rafikov
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Olga Rafikova
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| |
Collapse
|
6
|
Pan H, Dai H, Wang L, Lin S, Tao Y, Zheng Y, Jiang R, Fang F, Wu Y. MicroRNA-410-3p modulates chondrocyte apoptosis and inflammation by targeting high mobility group box 1 (HMGB1) in an osteoarthritis mouse model. BMC Musculoskelet Disord 2020; 21:486. [PMID: 32709223 PMCID: PMC7379779 DOI: 10.1186/s12891-020-03489-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background Osteoarthritis (OA) is the most prevalent type of arthritis, which commonly involves inflammation in the articular cartilage in OA pathogenesis. MicroRNAs (miRNAs) play essential roles in the regulation and pathophysiology of various diseases including OA. MiR-410-3p has been demonstrated to mediate inflammatory pathways, however, the regulatory functions of miR-410-3p in OA remain largely unknown. Methods The regulations of miR-410-3p were investigated in OA. Mouse primary chondrocytes and mouse in vivo models were used. The expression levels of miR-410-3p and HMGB1 were measured by qPCR. The transcription activity of NF-κB was assessed by luciferase reporter assay. MTT assay was performed to assess cellular proliferation. Cell apoptosis was evaluated with the Fluorescein Isothiocyanate (FITC) Annexin V assay. Expression levels of proteins were determined by Western blot. Results The results demonstrated that miR-410-3p was markedly downregulated in articular cartilage tissues as well as in lipopolysaccharide (LPS)-treated chondrocytes in OA mice. In addition, upregulation of miR-410-3p markedly inhibited LPS-induced apoptosis of chondrocytes. The results also demonstrated that the high mobility group box 1 (HMGB1) was a target of miR-410-3p. LPS-induced upregulated expression of HMGB1 significantly suppressed expression of miR-410-3p. Furthermore, upregulation of miR-410-3p markedly inhibited HMGB1 expression, the nuclear factor (NF)-kB activity and pro-inflammatory cytokines production. Taken together, the results suggested that miR-410-3p targeted HMGB1 and modulated chondrocytes apoptosis and inflammation through the NF-κB signaling pathway. Conclusions These findings provide insights into the potential of miR-410-3p/ HMGB1 as therapeutic targets for OA treatment.
Collapse
Affiliation(s)
- Hong Pan
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China.
| | - Huming Dai
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Linzhi Wang
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Silong Lin
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Yuefeng Tao
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Yi Zheng
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Renyi Jiang
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Fan Fang
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Yifan Wu
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| |
Collapse
|
7
|
Sex-specific stress response and HMGB1 release in pulmonary endothelial cells. PLoS One 2020; 15:e0231267. [PMID: 32271800 PMCID: PMC7145198 DOI: 10.1371/journal.pone.0231267] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 03/05/2020] [Indexed: 01/03/2023] Open
Abstract
Women are known to be associated with a higher susceptibility to pulmonary arterial hypertension (PAH). In contrast, male PAH patients have a worse survival prognosis. In this study, we investigated whether the contribution of sex goes beyond the effects of sex hormones by comparing the ability of isolated male and female pulmonary endothelial cells to respire, proliferate and tolerate the stress. Mouse lung endothelial cells (MLEC) were isolated from the lungs of male and female 3-week old mice. Male MLEC showed an increased basal mitochondrial respiration rate, elevated maximal respiration, a significantly greater level of mitochondrial polarization, and a higher rate of proliferation. Exposure of cells to hypoxia (2% of O2 for 24 hours) induced a strong apoptotic response in female but not male MLEC. In contrast, treatment with mitochondrial respiratory Complex III inhibitor Antimycin A (AA, 50μM) mediated severe necrosis specifically in male MLEC, while female cells again responded primarily by apoptosis. The same effect with female cells responding to the stress by apoptosis and male cells responding by necrosis was confirmed in starved pulmonary endothelial cells isolated from human donors. Elevated necrosis seen in male cells was associated with a significant release of damage-associated alarmin, HMGB1. No stimuli induced a significant elevation of HMGB1 secretion in females. We conclude that male cells appear to be protected against mild stress conditions, such as hypoxia, possibly due to increased mitochondrial respiration. In contrast, they are more sensitive to impaired mitochondrial function, to which they respond by necrotic death. Necrosis in male vascular cells releases a significant amount of HMGB1 that could contribute to the pro-inflammatory phenotype known to be associated with the male gender.
Collapse
|
8
|
Rafikova O, Al Ghouleh I, Rafikov R. Focus on Early Events: Pathogenesis of Pulmonary Arterial Hypertension Development. Antioxid Redox Signal 2019; 31:933-953. [PMID: 31169021 PMCID: PMC6765063 DOI: 10.1089/ars.2018.7673] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022]
Abstract
Significance: Pulmonary arterial hypertension (PAH) is a progressive disease of the lung vasculature characterized by the proliferation of all vascular wall cell types, including endothelial, smooth muscle, and fibroblasts. The disease rapidly advances into a form with extensive pulmonary vascular remodeling, leading to a rapid increase in pulmonary vascular resistance, which results in right heart failure. Recent Advances: Most current research in the PAH field has been focused on the late stage of the disease, largely due to an urgent need for patient treatment options in clinics. Further, the pathobiology of PAH is multifaceted in the advanced disease, and there has been promising recent progress in identifying various pathological pathways related to the late clinical picture. Critical Issues: Early stage PAH still requires additional attention from the scientific community, and although the survival of patients with early diagnosis is comparatively higher, the disease develops in patients asymptomatically, making it difficult to identify and treat early. Future Directions: There are several reasons to focus on the early stage of PAH. First, the complexity of late stage disease, owing to multiple pathways being activated in a complex system with intra- and intercellular signaling, leads to an unclear picture of the key contributors to the pathobiology. Second, an understanding of early pathophysiological events can increase the ability to identify PAH patients earlier than what is currently possible. Third, the prompt diagnosis of PAH would allow for the therapy to start earlier, which has proved to be a more successful strategy, and it ensures better survival in PAH patients.
Collapse
Affiliation(s)
- Olga Rafikova
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| | - Imad Al Ghouleh
- Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruslan Rafikov
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
9
|
Rafikov R, Nair V, Sinari S, Babu H, Sullivan JC, Yuan JXJ, Desai AA, Rafikova O. Gender Difference in Damage-Mediated Signaling Contributes to Pulmonary Arterial Hypertension. Antioxid Redox Signal 2019; 31:917-932. [PMID: 30652485 PMCID: PMC6765065 DOI: 10.1089/ars.2018.7664] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aims: Pulmonary arterial hypertension (PAH) is a progressive lethal disease with a known gender dimorphism. Female patients are more susceptible to PAH, whereas male patients have a lower survival rate. Initial pulmonary vascular damage plays an important role in PAH pathogenesis. Therefore, this study aimed at investigating the role of gender in activation of apoptosis/necrosis-mediated signaling pathways in PAH. Results: The media collected from pulmonary artery endothelial cells (PAECs) that died by necrosis or apoptosis were used to treat naive PAECs. Necrotic cell death stimulated phosphorylation of toll-like receptor 4, accumulation of interleukin 1 beta, and expression of E-selectin in a redox-dependent manner; apoptosis did not induce any of these effects. In the animal model of severe PAH, the necrotic marker, high mobility group box 1 (HMGB1), was visualized in the pulmonary vascular wall of male but not female rats. This vascular necrosis was associated with male-specific redox changes in plasma, activation of the same inflammatory signaling pathway seen in response to necrosis in vitro, and an increased endothelial-leukocyte adhesion in small pulmonary arteries. In PAH patients, gender-specific changes in redox homeostasis correlated with the prognostic marker, B-type natriuretic peptide. Males had also shown elevated circulating levels of HMGB1 and pro-inflammatory changes. Innovation: This study discovered the role of gender in the initiation of damage-associated signaling in PAH and highlights the importance of the gender-specific approach in PAH therapy. Conclusion: In PAH, the necrotic cell death is augmented in male patients compared with female patients. Factors released from necrotic cells could alter redox homeostasis and stimulate inflammatory signaling pathways.
Collapse
Affiliation(s)
- Ruslan Rafikov
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| | - Vineet Nair
- Division of Cardiology, Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Shripad Sinari
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, Arizona
| | | | | | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, University of Arizona, Tucson, Arizona
| | - Ankit A Desai
- Division of Cardiology, Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Olga Rafikova
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
10
|
Zhao J, Sun T, Wu S, Liu Y. High Mobility Group Box 1: An Immune-regulatory Protein. Curr Gene Ther 2019; 19:100-109. [PMID: 31223085 DOI: 10.2174/1566523219666190621111604] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 12/26/2022]
Abstract
High mobility group box 1 (HMGB1) presents in almost all somatic cells as a component of the cell nucleus. It is necessary for transcription regulation during cell development. Recent studies indicate that extracellular HMGB1, coming from necrotic cells or activated immune cells, triggers inflammatory response whereas intracellular HMGB1 controls the balance between autophagy and apoptosis. In addition, reduced HMGB1 can effectively mediate tissue regeneration. HMGB1, therefore, is regarded as a therapeutic target for inflammatory diseases. In this review, we summarized and discussed the immunomodulatory effect of HMGB1.
Collapse
Affiliation(s)
- Jingjing Zhao
- Department of Intensive Care Medicine, Hefei No. 2 People Hospital, Hefei 230000, China
| | - Tianle Sun
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215000, China
| | - Shengdi Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yufeng Liu
- Department of Nursing, General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
11
|
RGMb protects against acute kidney injury by inhibiting tubular cell necroptosis via an MLKL-dependent mechanism. Proc Natl Acad Sci U S A 2018; 115:E1475-E1484. [PMID: 29382757 DOI: 10.1073/pnas.1716959115] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tubular cell necrosis is a key histological feature of acute kidney injury (AKI). Necroptosis is a type of programed necrosis, which is executed by mixed lineage kinase domain-like protein (MLKL) upon its binding to the plasma membrane. Emerging evidence indicates that necroptosis plays a critical role in the development of AKI. However, it is unclear whether renal tubular cells undergo necroptosis in vivo and how the necroptotic pathway is regulated during AKI. Repulsive guidance molecule (RGM)-b is a member of the RGM family. Our previous study demonstrated that RGMb is highly expressed in kidney tubular epithelial cells, but its biological role in the kidney has not been well characterized. In the present study, we found that RGMb reduced membrane-associated MLKL levels and inhibited necroptosis in cultured cells. During ischemia/reperfusion injury (IRI) or oxalate nephropathy, MLKL was induced to express on the apical membrane of proximal tubular (PT) cells. Specific knockout of Rgmb in tubular cells (Rgmb cKO) increased MLKL expression at the apical membrane of PT cells and induced more tubular cell death and more severe renal dysfunction compared with wild-type mice. Treatment with the necroptosis inhibitor Necrostatin-1 or GSK'963 reduced MLKL expression on the apical membrane of PT cells and ameliorated renal function impairment after IRI in both wild-type and Rgmb cKO mice. Taken together, our results suggest that proximal tubular cell necroptosis plays an important role in AKI, and that RGMb protects against AKI by inhibiting MLKL membrane association and necroptosis in proximal tubular cells.
Collapse
|
12
|
Luhrs KA, Pink D, Schulte W, Zijlstra A, Lewis JD, Parseghian MH. In vivo histone H1 migration from necrotic to viable tissue. Oncotarget 2017; 8:16275-16292. [PMID: 28187445 PMCID: PMC5369962 DOI: 10.18632/oncotarget.15181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/25/2017] [Indexed: 11/29/2022] Open
Abstract
Necrosis is induced by ischemic conditions within the core of many solid tumors. Using fluorescent fusion proteins, we provide in vivo evidence of histone trafficking among cancer cells in implanted tumors. In particular, the most abundant H1 isoform (H1.2) was found to be transported from necrotic tumor cells into surrounding viable cells where histones are selectively taken up by energy-dependent endocytosis. We propose that intercellular histone trafficking could function as a target for drug delivery. This concept was validated using an anti-histone antibody that was co-internalized with histones from dead cells into viable ones surrounding the necrotic regions of a tumor, where some of the most chemoresistant cells reside. These findings demonstrate that cellular translocation of conjugated drugs using anti-histone antibodies is a promising strategy for targeted drug delivery to chemoresistant tumors.
Collapse
Affiliation(s)
- Keith A. Luhrs
- Allergan Inc., Irvine, CA, USA
- Peregrine Pharmaceuticals Inc., Tustin, CA, USA
| | - Desmond Pink
- Innovascreen Inc., Halifax, NS, Canada
- University of Alberta, Edmonton, AB, Canada
| | | | - Andries Zijlstra
- Innovascreen Inc., Halifax, NS, Canada
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - John D. Lewis
- Innovascreen Inc., Halifax, NS, Canada
- University of Alberta, Edmonton, AB, Canada
| | - Missag H. Parseghian
- Rubicon Biotechnology, Lake Forest, CA, USA
- Peregrine Pharmaceuticals Inc., Tustin, CA, USA
| |
Collapse
|
13
|
Su Z, Ni P, She P, Liu Y, Richard SA, Xu W, Zhu H, Wang J. Bio-HMGB1 from breast cancer contributes to M-MDSC differentiation from bone marrow progenitor cells and facilitates conversion of monocytes into MDSC-like cells. Cancer Immunol Immunother 2017; 66:391-401. [PMID: 27987020 PMCID: PMC11028758 DOI: 10.1007/s00262-016-1942-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 12/04/2016] [Indexed: 01/08/2023]
Abstract
Myeloid-derived suppressor cells (MDSC) constitute the major cell population that regulates immune responses. They are known to accumulate in tumors, chronic inflammatory and autoimmune diseases. Previous data indicate that high mobility group box 1(HMGB1) facilitates MDSC differentiation from bone marrow, suppresses NK cells, CD4+ and CD8+ T cells and is involved in cancer development. However, it remains unclear what potential mechanisms of HMGB1 facilitate MDSC differentiation. In the present work, we clearly demonstrate that HMGB1 secreted by cancer cells is N-glycosylated at Asn37, which facilitates monocytic (M)-MDSC differentiation from bone marrow via the p38/NFκB/Erk1/2 pathway and also contributes to conversion of monocytes into MDSC-like cells; HMGB1 blockade by a monoclonal antibody against the HMGB1 B box obviously reduced the accumulation of M-MDSC in tumor-bearing mice, delaying tumor growth and development; additionally, MDSC expansion and HMGB1 up-regulation were also found in breast cancer patients. All these data indicate that HMGB1 might be a potential tumor immunotherapy target.
Collapse
Affiliation(s)
- Zhaoliang Su
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Department of Immunology, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Ping Ni
- Department of Immunology, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Peng She
- Department of Immunology, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yueqin Liu
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Seidu A Richard
- Department of Immunology, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wenlin Xu
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Haitao Zhu
- The Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Jia Wang
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Department of Immunology, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
14
|
Luo F, Herrup K, Qi X, Yang Y. Inhibition of Drp1 hyper-activation is protective in animal models of experimental multiple sclerosis. Exp Neurol 2017; 292:21-34. [PMID: 28238799 DOI: 10.1016/j.expneurol.2017.02.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 12/20/2022]
Abstract
Multiple Sclerosis (MS), a leading neurological disorder of young adults, is characterized by the loss of oligodendrocytes (OLs), demyelination, inflammation and neuronal degeneration. Here we show that dynamin-related protein 1 (Drp1), a mitochondrial fission protein, is activated in primary OL cells exposed to TNF-α induced inflammation or oxidative stress, as well as in EAE-immunized and cuprizone toxicity-induced demyelinating mouse models. Inhibition of Drp1 hyper-activation by the selective inhibitor P110 abolishes Drp1 translocation to the mitochondria, reduces mitochondrial fragmentation and stems necrosis in primary OLs exposed to TNF-α and H2O2. Notably, in both types of mouse models, treatment with P110 significantly reduces the loss of mature OLs and demyelination, attenuates the number of active microglial cells and astrocytes, yet has no effect on the differentiation of oligodendrocyte precursor cells. Drp1 activation appears to be mediated through the RIPK1/RIPK3/MLKL/PGAM5 pathway during TNF-α-induced oligodendroglia necroptosis. Our results demonstrate a critical role of Drp1 hyper-activation in OL cell death and suggest that an inhibitor of Drp1 hyper-activation such as P110 is worth exploring for its ability to halt or slow the progression of MS.
Collapse
Affiliation(s)
- Fucheng Luo
- Department of Neurology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Karl Herrup
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA; Center for Mitochondria Diseases, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| | - Yan Yang
- Department of Neurology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA; Center for Translational Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| |
Collapse
|
15
|
Kim J, Kim CS, Sohn E, Kim JS. Cytoplasmic translocation of high-mobility group box-1 protein is induced by diabetes and high glucose in retinal pericytes. Mol Med Rep 2016; 14:3655-61. [PMID: 27599553 PMCID: PMC5042730 DOI: 10.3892/mmr.2016.5702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 07/18/2016] [Indexed: 01/13/2023] Open
Abstract
The aim of the present study was to assess the involvement of the high-mobility group box-1 (HMGB1) protein, receptor for advanced glycation end products (RAGE) and nuclear factor (NF)-κB signaling pathway in the development of diabetic retinopathy. Rat primary retinal pericytes were exposed to 25 mmol/l D-glucose for 48 h. Diabetic retinal vessels were prepared from streptozotocin-induced diabetic rats 12 weeks following the induction of diabetes. The expression of HMGB1 was detected using immunofluorescence staining. The expression of RAGE and the activity of NF-κB were analyzed using western blot and electrophoretic mobility shift assays, respectively. The results showed that HMGB1 was translocated to the cytoplasm of the high glucose-treated pericytes and diabetic retinal pericytes, whereas, in the control cells and the normal retinas, HMGB1 was expressed in the cell nuclei only. The expression of RAGE, a potential receptor for HMGB1, and the activity of NF-κB were also increased in the high glucose-treated pericytes, compared with the normal control cells. In addition, high glucose increased the binding of NF-κB to the RAGE promoter. These findings suggested that the cytoplasmic translocation of HMGB1 may be caused by diabetes and high glucose in retinal pericytes, and that the pathogenic role of HMGB1 may be dependent on the expression of RAGE and activation of NF-κB.
Collapse
Affiliation(s)
- Junghyun Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Chan-Sik Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Eunjin Sohn
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jin Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| |
Collapse
|
16
|
King CR, Tessier TM, Mymryk JS. Color Me Infected: Painting Cellular Chromatin with a Viral Histone Mimic. Trends Microbiol 2016; 24:774-776. [PMID: 27592243 DOI: 10.1016/j.tim.2016.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022]
Abstract
Viruses manipulate cellular chromatin to create a favourable milieu for infection. In several cases, virally-encoded proteins structurally mimic cellular histones to molecularly rewire the host cell. A recent study identified a novel mechanism whereby adenovirus protein VII, a viral histone, binds and manipulates host cell chromatin to suppress inflammatory signalling.
Collapse
Affiliation(s)
- Cason R King
- Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tanner M Tessier
- Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | - Joe S Mymryk
- Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada; Departments of Oncology and Otolaryngology, University of Western Ontario, London, Ontario, Canada; London Regional Cancer Program and Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
17
|
Abstract
The primary function of the genome is to store, propagate, and express the genetic information that gives rise to a cell's architectural and functional machinery. However, the genome is also a major structural component of the cell. Besides its genetic roles, the genome affects cellular functions by nongenetic means through its physical and structural properties, particularly by exerting mechanical forces and by serving as a scaffold for binding of cellular components. Major cellular processes affected by nongenetic functions of the genome include establishment of nuclear structure, signal transduction, mechanoresponses, cell migration, and vision in nocturnal animals. We discuss the concept, mechanisms, and implications of nongenetic functions of the genome.
Collapse
Affiliation(s)
| | - Tom Misteli
- National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Lee W, Ku SK, Park EJ, Na DH, Kim KM, Bae JS. Exendin-4 inhibits HMGB1-induced inflammatory responses in HUVECs and in murine polymicrobial sepsis. Inflammation 2015; 37:1876-88. [PMID: 24826914 DOI: 10.1007/s10753-014-9919-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exendin-4 (EX4) has been reported to attenuate myocardial ischemia and reperfusion (I/R) injury and inflammatory and oxidative responses. Nuclear DNA-binding protein high-mobility group box 1 (HMGB1) protein acts as a late mediator of severe vascular inflammatory conditions. However, the effect of EX4 on HMGB1-induced inflammatory response has not been studied. First, we accessed this question by monitoring the effects of posttreatment EX4 on lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of proinflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. Posttreatment EX4 was found to suppress LPS-mediated release of HMGB1 and inhibited HMGB1-mediated hyperpermeability and leukocyte migration in septic mice. EX4 also induced downregulation of CLP-induced release of HMGB1, production of IL-6, and mortality. Collectively, these results suggest that EX4 may be regarded as a candidate therapeutic agent for treatment of vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
19
|
Palmblad K, Schierbeck H, Sundberg E, Horne AC, Harris HE, Henter JI, Antoine DJ, Andersson U. High systemic levels of the cytokine-inducing HMGB1 isoform secreted in severe macrophage activation syndrome. Mol Med 2015; 20:538-47. [PMID: 25247290 DOI: 10.2119/molmed.2014.00183] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 09/15/2014] [Indexed: 01/31/2023] Open
Abstract
Macrophage activation syndrome (MAS) is a potentially fatal complication of systemic inflammation. High mobility group box 1 (HMGB1) is a nuclear protein extensively leaked extracellularly during necrotic cell death or actively secreted by natural killer (NK) cells, macrophages and additional cells during infection or sterile injury. Extracellular HMGB1 orchestrates key events in inflammation as a prototypic alarmin. The redox states of its three cysteines render the molecule mutually exclusive functions: fully reduced "all-thiol HMGB1" exerts chemotactic activity; "disulfide HMGB1" has cytokine-inducing, toll-like receptor 4 (TLR4)-mediated effects—while terminally oxidized "sulfonyl HMGB1" lacks inflammatory activity. This study examines the kinetic pattern of systemic HMGB1 isoform expression during therapy in four children with severe MAS. Three of the four patients with underlying systemic rheumatic diseases were treated with biologics and two suffered from triggering herpes virus infections at the onset of MAS. All patients required intensive care unit therapy due to life-threatening illness. Tandem mass-spectrometric analysis revealed dramatically increased systemic levels of the cytokine-inducing HMGB1 isoform during early MAS. Disease control coincided with supplementary etoposide therapy initiated to boost apoptotic cell death, when systemic HMGB1 levels drastically declined and the molecule emerged mainly in its oxidized, noninflammatory isoform. Systemic interferon (IFN)-γ and ferritin peaked concomitantly with HMGB1, whereas interleukin (IL)-18 and monocyte chemotactic protein (MCP)-1 levels developed differently. In conclusion, this work provides new insights in HMGB1 biology, suggesting that the molecule is not merely a biomarker of inflammation, but most likely also contributes to the pathogenesis of MAS. These observations encourage further studies of disulfide HMGB1 antagonists to improve outcome of MAS.
Collapse
Affiliation(s)
- Karin Palmblad
- Unit of Pediatric Rheumatology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Schierbeck
- Unit of Pediatric Rheumatology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundberg
- Unit of Pediatric Rheumatology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anna-Carin Horne
- Unit of Pediatric Rheumatology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Helena Erlandsson Harris
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jan-Inge Henter
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Daniel J Antoine
- Medical Research Council Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, United Kingdom
| | - Ulf Andersson
- Unit of Pediatric Rheumatology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Pei JX, Xiong GL, Xie Y. Relationship between high mobility group box 1 protein and inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2014; 22:5092-5099. [DOI: 10.11569/wcjd.v22.i33.5092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
High mobility group box 1 protein (HMGB1) is a DNA binding protein that can promote the maintenance of nucleosomal structures and regulate gene transcription in mammalian cells. HMGB1 is a ubiquitous nuclear protein that is widely distributed among mammalian cells, passively released from necrotic cells and actively released from stimulated inflammatory cells. HMGB1 might function as an endogenous immune adjuvant and play a crucial role in the development of various inflammatory diseases, and blockade of HMGB1 expression attenuates the intestinal inflammation and damage in animal models. This article reviews recent progress in understanding the relationship between HMGB1 and inflammatory bowel disease.
Collapse
|
21
|
Drp1 stabilizes p53 on the mitochondria to trigger necrosis under oxidative stress conditions in vitro and in vivo. Biochem J 2014; 461:137-46. [PMID: 24758576 DOI: 10.1042/bj20131438] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxidative-stress-induced necrosis is considered to be one of the main pathological mediators in various neurological disorders, such as brain ischaemia. However, little is known about the mechanism by which cells modulate necrosis in response to oxidative stress. In the present study, we showed that Drp1 (dynamin-related protein 1), a primary mitochondrial fission protein, stabilizes the well-known stress gene p53 and is required for p53 translocation to the mitochondria under conditions of oxidative stress. We found that Drp1 binding to p53 induced mitochondria-related necrosis. In contrast, inhibition of Drp1 hyperactivation by Drp1 siRNA reduced necrotic cell death in cell cultures exposed to oxidative stress. Most significantly, we demonstrated that inhibition of Drp1 by the Drp1 peptide inhibitor P110, which was developed recently by our group, abolished p53 association with the mitochondria and reduced brain infarction in rats subjected to brain ischaemia/reperfusion injury. Taken together, these findings reveal a novel mechanism of Drp1 hyperactivation in the induction of mitochondrial damage and subsequent cell death. We propose that a Drp1 inhibitor such as P110 is a possible therapeutic agent for diseases in which hyperactivated Drp1 contributes to the pathology.
Collapse
|
22
|
Lee W, Ku SK, Bae JS. Factor Xa inhibits HMGB1-induced septic responses in human umbilical vein endothelial cells and in mice. Thromb Haemost 2014; 112:757-69. [PMID: 25007770 DOI: 10.1160/th14-03-0233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/12/2014] [Indexed: 12/24/2022]
Abstract
Nuclear DNA-binding protein high mobility group box 1 (HMGB1) acts as a late mediator of severe vascular inflammatory conditions, such as sepsis. Activated factor X (FXa) is an important player in the coagulation cascade responsible for thrombin generation, and it influences cell signalling in various cell types by activating protease-activated receptors (PARs). However, the effect of FXa on HMGB1-induced inflammatory response has not been studied. First, we addressed this issue by monitoring the effects of post-treatment with FXa on lipopolysaccharide (LPS)- and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. Post-treatment with FXa was found to suppress LPS-mediated release of HMGB1 and HMGB1-mediated cytoskeletal rearrangements. FXa also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in septic mice. In addition, FXa inhibited the production of tumour necrosis factor-α and interleukin (IL)-1β. FXa also facilitated the downregulation of CLP-induced release of HMGB1, production of IL-6, and mortality. Collectively, these results suggest that FXa may be regarded as a candidate therapeutic agent for treating vascular inflammatory diseases by inhibiting the HMGB1 signalling pathway.
Collapse
Affiliation(s)
| | | | - Jong-Sup Bae
- Jong-Sup Bae, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 702-701, Republic of Korea, Tel.: +82 53 950 8570, Fax: +82 53 950 8557, E-mail:
| |
Collapse
|
23
|
Lee W, Ku S, Yoo H, Song K, Bae J. Andrographolide inhibits HMGB1-induced inflammatory responses in human umbilical vein endothelial cells and in murine polymicrobial sepsis. Acta Physiol (Oxf) 2014; 211:176-87. [PMID: 24581270 DOI: 10.1111/apha.12264] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/06/2013] [Accepted: 02/24/2014] [Indexed: 01/18/2023]
Abstract
AIM Nuclear DNA-binding protein high-mobility group box 1 (HMGB1) protein acts as a late mediator of severe vascular inflammatory conditions, such as septic shock, upregulating pro-inflammatory cytokines. Andrographolide (AG) is isolated from the plant of Andrographis paniculata and used as a folk medicine for treatment of viral infection, diarrhoea, dysentery and fever. However, the effect of AG on HMGB1-induced inflammatory response has not been studied. METHODS Firstly, we accessed this question by monitoring the effects of post-treatment AG on lipopolysaccharide (LPS) and caecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. RESULTS Post-treatment AG was found to suppress LPS-mediated release of HMGB1 and HMGB1-mediated cytoskeletal rearrangements. AG also inhibited HMGB1-mediated hyperpermeability and leucocyte migration in septic mice. In addition, AG inhibited production of tumour necrosis factor-α (TNF-α) and activation of AKT, nuclear factor-κB (NF-κB) and extracellular-regulated kinases (ERK) 1/2 by HMGB1 in HUVECs. AG also induced downregulation of CLP-induced release of HMGB1, production of interleukin (IL) 1β/6/8 and mortality. CONCLUSION Collectively, these results suggest that AG may be regarded as a candidate therapeutic agent for the treatment of vascular inflammatory diseases via inhibition of the HMGB1 signalling pathway.
Collapse
Affiliation(s)
- W. Lee
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
- Department of Biochemistry and Cell Biology; School of Medicine; Kyungpook National University; Daegu Korea
| | - S. Ku
- Department of Anatomy and Histology; College of Korean Medicine; Daegu Haany University; Gyeongsan Korea
| | - H. Yoo
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| | - K. Song
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| | - J. Bae
- College of Pharmacy; CMRI; Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu Korea
| |
Collapse
|
24
|
Brauchle E, Thude S, Brucker SY, Schenke-Layland K. Cell death stages in single apoptotic and necrotic cells monitored by Raman microspectroscopy. Sci Rep 2014; 4:4698. [PMID: 24732136 PMCID: PMC3986703 DOI: 10.1038/srep04698] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 04/01/2014] [Indexed: 12/12/2022] Open
Abstract
Although apoptosis and necrosis have distinct features, the identification and discrimination of apoptotic and necrotic cell death in vitro is challenging. Immunocytological and biochemical assays represent the current gold standard for monitoring cell death pathways; however, these standard assays are invasive, render large numbers of cells and impede continuous monitoring experiments. In this study, both room temperature (RT)-induced apoptosis and heat-triggered necrosis were analyzed in individual Saos-2 and SW-1353 cells by utilizing Raman microspectroscopy. A targeted analysis of defined cell death modalities, including early and late apoptosis as well as necrosis, was facilitated based on the combination of Raman spectroscopy with fluorescence microscopy. Spectral shifts were identified in the two cell lines that reflect biochemical changes specific for either RT-induced apoptosis or heat-mediated necrosis. A supervised classification model specified apoptotic and necrotic cell death based on single cell Raman spectra. To conclude, Raman spectroscopy allows a non-invasive, continuous monitoring of cell death, which may help shedding new light on complex pathophysiological or drug-induced cell death processes.
Collapse
Affiliation(s)
- Eva Brauchle
- Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), Department of Cell and Tissue Engineering, Nobelstr. 12, 70569 Stuttgart, Germany
- University of Stuttgart, Institute of Interfacial Process Engineering and Plasma Technology (IGVP), Nobelstr. 12, 70569 Stuttgart, Germany
| | - Sibylle Thude
- Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), Department of Cell and Tissue Engineering, Nobelstr. 12, 70569 Stuttgart, Germany
| | - Sara Y. Brucker
- University Women's Hospital, Eberhard-Karls-University Tübingen, Silcherstr. 7/1, 72076 Tübingen, Germany
| | - Katja Schenke-Layland
- Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), Department of Cell and Tissue Engineering, Nobelstr. 12, 70569 Stuttgart, Germany
- University Women's Hospital, Eberhard-Karls-University Tübingen, Silcherstr. 7/1, 72076 Tübingen, Germany
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive South, MRL 3645, Los Angeles, CA, USA
| |
Collapse
|
25
|
The spectrum of anti-chromatin/nucleosome autoantibodies: independent and interdependent biomarkers of disease. J Immunol Res 2014; 2014:368274. [PMID: 24804269 PMCID: PMC3996305 DOI: 10.1155/2014/368274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/03/2014] [Indexed: 01/08/2023] Open
Abstract
Autoantibodies directed to chromatin components date back to the discovery of the LE cell and the LE cell phenomenon circa 1950, and subsequent evidence that major components of that reaction were chromatin components and histones in particular. Over time, immunoassays ranging from ELISA and line immunoassays to more modern bead-based assays incorporated histone and DNA mixtures, purified histones, and purified nucleosomes leading to a more thorough understanding of the genesis and pathogenetic relationships of antibodies to chromatin components in systemic lupus erythematosus and other autoimmune conditions. More recently, interest has focussed on other components of chromatin such as high mobility group (HMG) proteins both as targets of B cell responses and pro-inflammatory mediators. This review will focus on immunoassays that utilize chromatin components, their clinical relationships, and newer evidence implicating HMG proteins and DNA neutrophil extracellular traps (NETs) as important players in systemic autoimmune rheumatic diseases.
Collapse
|
26
|
Chitanuwat A, Laosrisin N, Dhanesuan N. Role of HMGB1 in proliferation and migration of human gingival and periodontal ligament fibroblasts. J Oral Sci 2014; 55:45-50. [PMID: 23485600 DOI: 10.2334/josnusd.55.45] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
High mobility group box 1 (HMGB1) was originally defined as a nuclear protein. However, later studies showed that HMGB1 was released from damaged cells into the extracellular milieu and functioned as a danger signaling molecule. HMGB1 has also been shown to exert proliferative and chemoattractant effects on many cell types. In this study, we investigated the in vitro effect of human recombinant HMGB1 on the proliferation and migration of human gingival fibroblasts (HGF) and human periodontal ligament fibroblasts (HPDLF). For the proliferation assay, HGF and HPDLF were cultured in the presence of 5, 10, and 50 ng/mL HMGB1. After a period of 6 days, cell proliferation was determined by MTT assay. The migration assay was performed by culturing the two cell types in Transwells with HMGB1 in the lower chamber as a chemoattractant. Cell migration during 16 h was determined by crystal violet staining of the cells that migrated across the membrane. The results showed that HMGB1, at 50 ng/mL, was able to significantly induce proliferation of HGF by up to 171.4 ± 17.1%. No such proliferation induction was seen for HPDLF. In the migration assay, however, 100 ng/mL HMGB1 induced migration of both cell types. The counts of cells that migrated across the membrane, as compared with the control, were increased to 273 ± 24.1% and 410.3 ± 158% for HGF and HPDLF, respectively. Since proliferation and migration are basic abilities of cells required for proper tissue repair, these data suggest that HMGB1 plays an important role in these functions of periodontal cells.
Collapse
Affiliation(s)
- Atithan Chitanuwat
- Department of Conservative Dentistry and Prosthodontics, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | | | | |
Collapse
|
27
|
Osteoarthritic cartilage lesions in the bovine patellar groove: a macroscopic, histological and immunohistological analysis. Vet J 2014; 200:88-95. [PMID: 24581814 DOI: 10.1016/j.tvjl.2014.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 01/30/2014] [Accepted: 01/31/2014] [Indexed: 02/02/2023]
Abstract
A high percentage of osteoarthritis (OA)-like patellar groove lesions in the stifle joint in calcium-deficient bulls has been recently reported. The prevalence of these lesions in bulls deficient in or supplemented with calcium was compared to findings in culled and healthy bulls to determine whether they represent normal anatomical variations, developmental anomalies or OA. It was hypothesized that the patellar groove lesions may represent OA. Distal cartilage samples from 160 femurs were analysed using a macroscopic Société Française d'Arthroscopie (SFA) OA grading system. Samples representing different SFA grades were subjected to Osteoarthritis Research Society International (OARSI) histological and high-mobility group box 1 (HMGB1) immunohistological OA grading. For a qualitative analysis three OA samples were immunostained for interleukin (IL)-1β, matrix metalloproteinase (MMP)-13 and collagenase-produced COL2-3/4M neoepitopes. Patellar groove lesions were found in 48% of the femurs and were highest in calcium-deficient animals (71%, P<0.001). All three different grading systems disclosed OA in culled bulls, but no focal areas of cartilage necrosis. OARSI and HMGB1 grades were fairly concordant (Spearman's ρ=0.95, P<0.001; Cohen's κ=0.23, P<0.005), both with a slight disparity with the SFA grade (ρ=0.80 and 0.87, P<0.01; κ=0.36 and 0.46, P<0.001). IL-1β, MMP-13 and COL2-3/4M staining patterns were compatible with OA. The study showed that patellar groove lesions are common in bulls. In all SFA, OARSI and HMGB1 graded samples the lesions clearly demonstrated OA and showed OA-typical pathophysiology. Arthroscopic SFA grading showed similar changes in calcium-deficient and calcium-supplemented bulls, but in the absence of a time course study and histological data the primary nature of these lesions could not be established with certainty.
Collapse
|
28
|
Lee W, Ku SK, Lee YM, Bae JS. Anti-septic effects of glyceollins in HMGB1-induced inflammatory responses in vitro and in vivo. Food Chem Toxicol 2014; 63:1-8. [DOI: 10.1016/j.fct.2013.10.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/26/2013] [Accepted: 10/20/2013] [Indexed: 12/11/2022]
|
29
|
Rui X, Shengli M, Zhankui J, Guofu P, Jinjian Y. HMGB1: a potential target for treatment of benign prostatic hyperplasia. Med Hypotheses 2013; 81:892-5. [PMID: 23948596 DOI: 10.1016/j.mehy.2013.07.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/22/2013] [Accepted: 07/23/2013] [Indexed: 10/26/2022]
Abstract
Previous studies have demonstrated an increased incidence of benign prostatic hyperplasia (BPH) in men with prostatitis. In addition to androgens and age, recent studies also pointed to an important role for inflammation in causing and promoting the progression of BPH. Inflammatory infiltrates are frequently observed in prostate tissue specimens, and the degree of inflammation has been correlated with prostate volume and weight. Furthermore, a pro-inflammatory microenvironment is closely related to BPH stromal hyperproliferation and tissue remodeling, although its role in BPH remains unclear. Accumulating evidence indicates that HMGB1 acts as a potent proinflammatory cytokine that contributes to the pathogenesis of many inflammatory and infectious disorders. Experimental studies also reported that HMGB1 promotes cell chemotaxis and proliferation. These observations led us to propose that HMGB1 contributes to the progress of BPH, and that targeting the HMGB1 signaling pathway might be a new strategy to treat prostatic enlargement.
Collapse
Affiliation(s)
- Xue Rui
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, NO.1 Jian She Dong Avenue, Zhengzhou 450002, People's Republic of China; Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, NO.1 Jian She Dong Avenue, Zhengzhou 450002, People's Republic of China
| | | | | | | | | |
Collapse
|
30
|
Furlani D, Donndorf P, Westien I, Ugurlucan M, Pittermann E, Wang W, Li W, Vollmar B, Steinhoff G, Kaminski A, Ma N. HMGB-1 induces c-kit+ cell microvascular rolling and adhesion via both toll-like receptor-2 and toll-like receptor-4 of endothelial cells. J Cell Mol Med 2012; 16:1094-105. [PMID: 21762373 PMCID: PMC4365888 DOI: 10.1111/j.1582-4934.2011.01381.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
High-mobility group box 1 (HMGB-1) is a strong chemo-attractive signal for both inflammatory and stem cells. The aim of this study is to evaluate the mechanisms regulating HMGB-1-mediated adhesion and rolling of c-kit(+) cells and assess whether toll-like receptor-2 (TLR-2) and toll-like receptor-4 (TLR-4) of endothelial cells or c-kit(+) cells are implicated in the activation of downstream migration signals to peripheral c-kit(+) cells. Effects of HMGB-1 on the c-kit(+) cells/endothelial interaction were evaluated by a cremaster muscle model in wild-type (WT), TLR-2 (-/-) and Tlr4 (LPS-del) mice. The mRNA and protein expression levels of endothelial nitric oxide synthase were determined by quantitative real-time PCR and immunofluorescence staining. Induction of crucial adhesion molecules for rolling and adhesion of stem cells and leukocytes were monitored in vivo and in vitro. Following local HMGB-1 administration, a significant increase in cell rolling was detected (32.4 ± 7.1% in 'WT' versus 9.9 ± 3.2% in 'control', P < 0.05). The number of firmly adherent c-kit(+) cells was more than 13-fold higher than that of the control group (14.6 ± 5.1 cells/mm(2) in 'WT' versus 1.1 ± 1.0 cells/mm(2) in 'control', P < 0.05). In knockout animals, the fraction of rolling cells did not differ significantly from control levels. Firm endothelial adhesion was significantly reduced in TLR-2 (-/-) and Tlr4 (LPS-del) mice compared to WT mice (1.5 ± 1.4 cells/mm(2) in 'TLR-2 (-/-)' and 2.4 ± 1.4 cells/mm(2) in 'Tlr4 (LPS-del)' versus 14.6 ± 5.1 cells/mm(2) in 'WT', P < 0.05). TLR-2 (-/-) and Tlr4 (LPS-del) stem cells in WT mice did not show significant reduction in rolling and adhesion compared to WT cells. HMGB-1 mediates c-kit(+) cell recruitment via endothelial TLR-2 and TLR-4.
Collapse
Affiliation(s)
- Dario Furlani
- Reference and Translation Centre for Cardiac Stem Cell Therapy, Department of Cardiac Surgery, University Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Vaseva AV, Marchenko ND, Ji K, Tsirka SE, Holzmann S, Moll UM. p53 opens the mitochondrial permeability transition pore to trigger necrosis. Cell 2012; 149:1536-48. [PMID: 22726440 PMCID: PMC3383624 DOI: 10.1016/j.cell.2012.05.014] [Citation(s) in RCA: 594] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 02/01/2012] [Accepted: 05/02/2012] [Indexed: 11/30/2022]
Abstract
Ischemia-associated oxidative damage leading to necrosis is a major cause of catastrophic tissue loss, and elucidating its signaling mechanism is therefore of paramount importance. p53 is a central stress sensor responding to multiple insults, including oxidative stress to orchestrate apoptotic and autophagic cell death. Whether p53 can also activate oxidative stress-induced necrosis is, however, unknown. Here, we uncover a role for p53 in activating necrosis. In response to oxidative stress, p53 accumulates in the mitochondrial matrix and triggers mitochondrial permeability transition pore (PTP) opening and necrosis by physical interaction with the PTP regulator cyclophilin D (CypD). Intriguingly, a robust p53-CypD complex forms during brain ischemia/reperfusion injury. In contrast, reduction of p53 levels or cyclosporine A pretreatment of mice prevents this complex and is associated with effective stroke protection. Our study identifies the mitochondrial p53-CypD axis as an important contributor to oxidative stress-induced necrosis and implicates this axis in stroke pathology.
Collapse
Affiliation(s)
| | | | - Kyungmin Ji
- Dept. of Pharmacology, Stony Brook University, Stony Brook NY 11794, USA
| | - Stella E. Tsirka
- Dept. of Pharmacology, Stony Brook University, Stony Brook NY 11794, USA
| | - Sonja Holzmann
- Dept. of Molecular Oncology, University of Göttingen, 37077 Göttingen, Germany
| | - Ute M. Moll
- Dept. of Pathology, Stony Brook University, Stony Brook NY 11794, USA
- Dept. of Molecular Oncology, University of Göttingen, 37077 Göttingen, Germany
| |
Collapse
|
32
|
Kim J, Sohn E, Kim CS, Jo K, Kim JS. The role of high-mobility group box-1 protein in the development of diabetic nephropathy. Am J Nephrol 2011; 33:524-9. [PMID: 21606643 DOI: 10.1159/000327992] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 03/29/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND/AIMS The purpose of the experiment reported here was to assess the involvement of high-mobility group box-1 (HMGB1), receptor for advanced glycation end products (RAGE) and nuclear factor (NF)-κB signaling pathway in the development of rat diabetic nephropathy. METHODS Diabetes was induced by intraperitoneal streptozotocin injection in 7-week-old male rats. At 20 weeks of age, renal expression of HMGB1 was detected by immunohistochemistry. The expression of RAGE and NF-κB activity was studied by Western blot and electrophoretic mobility shift assay in renal tissues of normoglycemic and diabetic rats, respectively. RESULTS HMGB1 was highly expressed in both the cytoplasmic and nuclear patterns in diabetic renal glomerular cells and tubular epithelial cells, although in normal rats, HMGB1 was expressed only in the cell nuclei. The expression of RAGE, a potential receptor for HMGB1, and NF-κB activity were also greater in diabetic than in normal rats. Moreover, diabetes increased the binding of NF-κB to the RAGE promoter. CONCLUSION These findings suggest that hyperglycemia-induced HMGB1 release may induce the renal injury in diabetic rats, and that the pathogenic role of HMGB1 might be dependent on RAGE and through activation of NF-κB.
Collapse
Affiliation(s)
- Junghyun Kim
- Diabetic Complications Research Center, Division of Traditional Korean Medicine Integrated Research, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | | | | | | | | |
Collapse
|
33
|
Polyanichko AM, Rodionova TJ, Vorob’ev VI, Chikhirzhina EV. Conformational properties of nuclear protein HMGB1 and specificity of its interaction with DNA. ACTA ACUST UNITED AC 2011. [DOI: 10.1134/s1990519x1102012x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
Immunosuppressant cytoprotection correlates with HMGB1 suppression in primary astrocyte cultures exposed to combined oxygen-glucose deprivation. Pharmacol Rep 2011; 63:392-402. [DOI: 10.1016/s1734-1140(11)70505-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 09/23/2010] [Indexed: 01/09/2023]
|
35
|
Affiliation(s)
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston, MA 02115
| |
Collapse
|
36
|
Someya Y, Yui H. Fluorescence lifetime probe for solvent microviscosity utilizing anilinonaphthalene sulfonate. Anal Chem 2010; 82:5470-6. [PMID: 20524653 DOI: 10.1021/ac100116j] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The correlation between the fluorescent dynamics of excited anilinonaphthalene sulfonate (ANS) and the microviscosity of solvent molecules surrounding ANS is investigated by time-resolved fluorescence spectroscopy. ANS has been widely used to probe the local hydrophobicity due to the drastic change in its intensity. It is revealed that the fluorescence lifetime from the charge transfer (CT) state of ANS sensitively reflects the microviscosity. The higher sensitivity of 2,6-ANS than of 1,8-ANS demonstrates that the spatial freedom of the rotating phenylamino group in the photoexcited ANS is an important factor that determines the sensitivity. As an application, the measurements of the microviscosity of water in biologically important systems, such as hyaluronan, gellan gum, and gelatin aqueous solutions are also presented. The present results suggest that the fluorescence lifetime of ANS enables the estimation of the solvent microviscosity and provide a useful probe molecule for fluorescence lifetime imaging microscopy.
Collapse
Affiliation(s)
- Yuu Someya
- Department of Chemistry, Faculty of Science, Tokyo University of Science, 12 Ichigaya-Funagawaramachi, Shinjyuku-ku, Tokyo 162-0826, Japan
| | | |
Collapse
|
37
|
High-mobility group box 1 (HMGB1) as a master regulator of innate immunity. Cell Tissue Res 2010; 343:189-99. [PMID: 20835834 DOI: 10.1007/s00441-010-1033-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 08/03/2010] [Indexed: 02/08/2023]
Abstract
Damage-associated molecular patterns (DAMPs) comprise intracellular molecules characterized by the ability to reach the extracellular environment, where they prompt inflammation and tissue repair. The high-mobility box group 1 (HMGB1) protein is a prototypic DAMP and is highly conserved in evolution. HMGB1 is released upon cell and tissue necrosis and is actively produced by immune cells. Evidence suggests that HMGB1 acts as a key molecule of innate immunity, downstream of persistent tissue injury, orchestrating inflammation, stem cell recruitment/activation, and eventual tissue remodeling.
Collapse
|
38
|
Gasparri J, Speroni L, Chiaramoni NS, Valle Alonso SD. Relationship between the adjuvant and cytotoxic effects of the positive charges and polymerization in liposomes. J Liposome Res 2010; 21:124-33. [DOI: 10.3109/08982104.2010.491073] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Oyama Y, Hashiguchi T, Taniguchi N, Tancharoen S, Uchimura T, Biswas KK, Kawahara KI, Nitanda T, Umekita Y, Lotz M, Maruyama I. High-mobility group box-1 protein promotes granulomatous nephritis in adenine-induced nephropathy. J Transl Med 2010; 90:853-66. [PMID: 20231821 PMCID: PMC3096665 DOI: 10.1038/labinvest.2010.64] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Granulomatous nephritis can be triggered by diverse factors and results in kidney failure. However, despite accumulating data about granulomatous inflammation, pathogenetic mechanisms in nephritis remain unclear. The DNA-binding high-mobility group box-1 protein (HMGB1) initiates and propagates inflammation when released by activated macrophages, and functions as an 'alarm cytokine' signaling tissue damage. In this study, we showed elevated HMGB1 expression in renal granulomas in rats with crystal-induced granulomatous nephritis caused by feeding an adenine-rich diet. HMGB1 levels were also raised in urine and serum, as well as in monocyte chemoattractant protein-1 (MCP-1), a mediator of granulomatous inflammation. Injection of HMGB1 worsened renal function and upregulated MCP-1 in rats with crystal-induced granulomatous nephritis. HMGB1 also induced MCP-1 secretion through mitogen-activated protein kinase (MAPK) and phosphoinositide-3-kinase (PI3K) pathways in rat renal tubular epithelial cells in vitro. Hmgb1(+/-) mice with crystal-induced nephritis displayed reduced MCP-1 expression in the kidneys and in urine and the number of macrophages in the kidneys was significantly decreased. We conclude that HMGB1 is a new mediator involved in crystal-induced nephritis that amplifies granulomatous inflammation in a cycle where MCP-1 attracts activated macrophages, resulting in excessive and sustained HMGB1 release. HMGB1 could be a novel target for inhibiting chronic granulomatous diseases.
Collapse
Affiliation(s)
- Yoko Oyama
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Noboru Taniguchi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California
| | - Salunya Tancharoen
- Pharmacology Department, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Tomonori Uchimura
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kamal K. Biswas
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ko-ichi Kawahara
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takao Nitanda
- Division of Surgical Pathology, Kagoshima University Hospital, Kagoshima, Japan
| | - Yoshihisa Umekita
- Department of Tumor Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Martin Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California
| | - Ikuro Maruyama
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
40
|
Avalos AM, Kiefer K, Tian J, Christensen S, Shlomchik M, Coyle AJ, Marshak-Rothstein A. RAGE-independent autoreactive B cell activation in response to chromatin and HMGB1/DNA immune complexes. Autoimmunity 2010; 43:103-10. [PMID: 20014975 DOI: 10.3109/08916930903384591] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Increasing evidence suggests that the excessive accumulation of apoptotic or necrotic cellular debris may contribute to the pathology of systemic autoimmune disease. HMGB1 is a nuclear DNA-associated protein, which can be released from dying cells thereby triggering inflammatory processes. We have previously shown that IgG2a-reactive B cell receptor (BCR) transgenic AM14 B cells proliferate in response to endogenous chromatin immune complexes (ICs), in the form of the anti-nucleosome antibody PL2-3 and cell debris, in a TLR9-dependent manner, and that these ICs contain HMGB1. Activation of AM14 B cells by these chromatin ICs was inhibited by a soluble form of the HMGB1 receptor, RAGE-Fc, suggesting HMGB1-RAGE interaction was important for this response. To further explore the role of HMGB1 in autoreactive B cell activation, we assessed the capacity of purified calf thymus HMGB1 to bind dsDNA fragments and found that HMGB1 bound both CG-rich and CG-poor DNA. However, HMGB1-DNA complexes could not activate AM14 B cells unless HMGB1 was bound by IgG2a and thereby able to engage the BCR. To ascertain the role of RAGE in autoreactive B cell responses to chromatin ICs, we intercrossed AM14 and RAGE-deficient mice. We found that spontaneous and defined DNA ICs activated RAGE+ and RAGE(- ) AM14 B cells to a comparable extent. These results suggest that HMGB1 promotes B cell responses to endogenous TLR9 ligands through a RAGE-independent mechanism.
Collapse
Affiliation(s)
- Ana M Avalos
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Luan ZG, Zhang H, Yang PT, Ma XC, Zhang C, Guo RX. HMGB1 activates nuclear factor-κB signaling by RAGE and increases the production of TNF-α in human umbilical vein endothelial cells. Immunobiology 2010; 215:956-62. [PMID: 20163887 DOI: 10.1016/j.imbio.2009.11.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 09/27/2009] [Accepted: 11/02/2009] [Indexed: 12/19/2022]
Abstract
OBJECTIVE High mobility group box chromosomal protein 1 (HMGB1) is a lately discovered candidate molecule identified as an important extracellular mediator in systemic inflammation. Systemic inflammation results in endothelial cell activation and microvascular injury. In the present study, we investigated the effects of HMGB1 on the activation of human umbilical vein endothelial cells (HUVECs) and defined pathways activated by HMGB1. METHODS HUVECs obtained by collagenase treatment of umbilical cord veins were stimulated in vitro with HMGB1. The activation of HUVECs was studied regarding (i) the kinetics of tumor necrosis factor-α (TNF-α) production in HUVECs, (ii) HMGB1-induced up-regulation of receptor for advanced glycation end products (RAGE), (iii) HMGB1-induced nuclear translocation of nuclear factor kappa B (NF-κB) in HUVECs, (iv) the activation of signalling transduction pathways. RESULTS HUVECs activation was stimulated by HMGB1 partially in a RAGE-dependent manner. Additionally, the HMGB1-induced activation of HUVECs was significantly inhibited by anti-RAGE monoclonal antibody and Ethyl pyruvate (EP) that had been shown to be an effective anti-inflammatory agent. Short-term prestimulation of HUVECs with HMGB1 caused a time-dependent increase in the secretion of TNF-α and expression of RAGE. Furthermore, HMGB1 stimulation resulted in nuclear translocation of transcription factor NF-κB. Most importantly, pretreatment with anti-RAGE monoclonal antibody significantly decreased the amounts of TNF-α and inhibited the nuclear translocation of NF-κB. Additionally in HUVECs cultures, EP specifically inhibited activation of NF-κB signaling pathway that are critical for TNF-α release. CONCLUSIONS In conclusion, Our data present a link between HMGB1and RAGE function of endothelial cells and demonstrate the pathway activated by HMGB1. These findings may provide a novel therapeutic strategy to improve the endothelial cells function.
Collapse
Affiliation(s)
- Zheng-Gang Luan
- Department of Intensive Care Unit, The First Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, China
| | | | | | | | | | | |
Collapse
|
42
|
Frey B, Schildkopf P, Rödel F, Weiss EM, Munoz LE, Herrmann M, Fietkau R, Gaipl US. AnnexinA5 renders dead tumor cells immunogenic—implications for multimodal cancer therapies. J Immunotoxicol 2009; 6:209-16. [DOI: 10.3109/15476910903204058] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
43
|
Zhu XM, Yao YM, Liang HP, Xu S, Dong N, Yu Y, Sheng ZY. The Effect of High Mobility Group Box-1 Protein on Splenic Dendritic Cell Maturation in Rats. J Interferon Cytokine Res 2009; 29:677-86. [PMID: 19642897 DOI: 10.1089/jir.2008.0104] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Xiao-mei Zhu
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yong-ming Yao
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Hua-ping Liang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Department 1, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Shan Xu
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Ning Dong
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yan Yu
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Zhi-yong Sheng
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
44
|
Reeves R. Nuclear functions of the HMG proteins. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2009; 1799:3-14. [PMID: 19748605 DOI: 10.1016/j.bbagrm.2009.09.001] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 09/04/2009] [Indexed: 12/12/2022]
Abstract
Although the three families of mammalian HMG proteins (HMGA, HMGB and HMGN) participate in many of the same nuclear processes, each family plays its own unique role in modulating chromatin structure and regulating genomic function. This review focuses on the similarities and differences in the mechanisms by which the different HMG families impact chromatin structure and influence cellular phenotype. The biological implications of having three architectural transcription factor families with complementary, but partially overlapping, nuclear functions are discussed.
Collapse
Affiliation(s)
- Raymond Reeves
- School of Molecular Biosciences, Washington State University, Biotechnology/Life Sciences Bldg., Rm. 143, Pullman, WA 99164-7520, USA.
| |
Collapse
|
45
|
Cardinal J, Pan P, Dhupar R, Ross M, Nakao A, Lotze M, Billiar T, Geller D, Tsung A. Cisplatin prevents high mobility group box 1 release and is protective in a murine model of hepatic ischemia/reperfusion injury. Hepatology 2009; 50:565-74. [PMID: 19492424 DOI: 10.1002/hep.23021] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED The nuclear protein high mobility group box 1 (HMGB1) is an important inflammatory mediator involved in the pathogenesis of liver ischemia/reperfusion (I/R) injury. Strategies aimed at preventing its release from stressed or damaged cells may be beneficial in preventing inflammation after I/R. Cisplatin is a member of the platinating chemotherapeutic agents and can induce DNA lesions that are capable of retaining high mobility group proteins inside the nucleus of cells. In vitro studies in primary cultured rat hepatocytes show that nontoxic concentrations of cisplatin can sequester HMGB1 inside the nucleus of hypoxic cells. Similarly, the in vivo administration of nontoxic doses of cisplatin prevents liver damage associated with a well-established murine model of hepatic I/R as measured by lower circulating serum aminotransferase levels, lower hepatic inflammatory cytokine levels including tumor necrosis factor alpha and interleukin-6, lower inducible NO synthase expression, and fewer I/R-associated histopathologic changes. The mechanism of action in vivo appears to involve the capacity of cisplatin to prevent the I/R-induced release of HMGB1 as well as to alter cell survival and stress signaling in the form of autophagy and mitogen-activated protein kinase activation, respectively. CONCLUSION Low, nontoxic doses of cisplatin can sequester HMGB1 inside the nucleus of redox-stressed hepatocytes in vitro and prevent its release in vivo in a murine model of hepatic I/R. Furthermore, cell survival and stress signaling pathways are altered by low-dose cisplatin. Therefore, platinating agents may provide a novel approach to mitigating the deleterious effects of I/R-mediated disease processes.
Collapse
Affiliation(s)
- Jon Cardinal
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Several nuclear events during apoptosis depend on caspase-3 activation but do not constitute a common pathway. PLoS One 2009; 4:e6234. [PMID: 19641621 PMCID: PMC2713420 DOI: 10.1371/journal.pone.0006234] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 05/31/2009] [Indexed: 12/21/2022] Open
Abstract
A number of nuclear events occur during apoptosis, including DNA laddering, nuclear lamina breakdown, phosphorylation of histones H2B and histone H2AX, and the tight binding to chromatin of HMGB1 and CAD, the nuclease responsible for DNA laddering. We have performed an epistasis analysis to investigate whether these events cluster together in pathways. We find that all depend directly or indirectly on caspase-3 activation. CAD activation, H2AX phosphorylation and DNA laddering cluster together into a pathway, but all other events appear to be independent of each other downstream of caspase-3, and likely evolved subject to different functional pressures.
Collapse
|
47
|
Gangelhoff TA, Mungalachetty PS, Nix JC, Churchill MEA. Structural analysis and DNA binding of the HMG domains of the human mitochondrial transcription factor A. Nucleic Acids Res 2009; 37:3153-64. [PMID: 19304746 PMCID: PMC2691818 DOI: 10.1093/nar/gkp157] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2008] [Revised: 02/23/2009] [Accepted: 02/24/2009] [Indexed: 11/20/2022] Open
Abstract
The mitochondrial transcription factor A (mtTFA) is central to assembly and initiation of the mitochondrial transcription complex. Human mtTFA (h-mtTFA) is a dual high mobility group box (HMGB) protein that binds site-specifically to the mitochondrial genome and demarcates the promoters for recruitment of h-mtTFB1, h-mtTFB2 and the mitochondrial RNA polymerase. The stoichiometry of h-mtTFA was found to be a monomer in the absence of DNA, whereas it formed a dimer in the complex with the light strand promoter (LSP) DNA. Each of the HMG boxes and the C-terminal tail were evaluated for their ability to bind to the LSP DNA. Removal of the C-terminal tail only slightly decreased nonsequence specific DNA binding, and box A, but not box B, was capable of binding to the LSP DNA. The X-ray crystal structure of h-mtTFA box B, at 1.35 A resolution, revealed the features of a noncanonical HMG box. Interactions of box B with other regions of h-mtTFA were observed. Together, these results provide an explanation for the unusual DNA-binding properties of box B and suggest possible roles for this domain in transcription complex assembly.
Collapse
Affiliation(s)
- Todd A. Gangelhoff
- Department of Pharmacology, Molecular Biology Program, University of Colorado Denver, School of Medicine, 12801 East 17th Avenue, Aurora, CO 80045-0511 and Molecular Biology Consortium, Beamline 4.2.2, Advanced Light Source, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Purnima S. Mungalachetty
- Department of Pharmacology, Molecular Biology Program, University of Colorado Denver, School of Medicine, 12801 East 17th Avenue, Aurora, CO 80045-0511 and Molecular Biology Consortium, Beamline 4.2.2, Advanced Light Source, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Jay C. Nix
- Department of Pharmacology, Molecular Biology Program, University of Colorado Denver, School of Medicine, 12801 East 17th Avenue, Aurora, CO 80045-0511 and Molecular Biology Consortium, Beamline 4.2.2, Advanced Light Source, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Mair E. A. Churchill
- Department of Pharmacology, Molecular Biology Program, University of Colorado Denver, School of Medicine, 12801 East 17th Avenue, Aurora, CO 80045-0511 and Molecular Biology Consortium, Beamline 4.2.2, Advanced Light Source, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| |
Collapse
|
48
|
The p53-cathepsin axis cooperates with ROS to activate programmed necrotic death upon DNA damage. Proc Natl Acad Sci U S A 2009; 106:1093-8. [PMID: 19144918 DOI: 10.1073/pnas.0808173106] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Three forms of cell death have been described: apoptosis, autophagic cell death, and necrosis. Although genetic and biochemical studies have formulated a detailed blueprint concerning the apoptotic network, necrosis is generally perceived as a passive cellular demise resulted from unmanageable physical damages. Here, we conclude an active de novo genetic program underlying DNA damage-induced necrosis, thus assigning necrotic cell death as a form of "programmed cell death." Cells deficient of the essential mitochondrial apoptotic effectors, BAX and BAK, ultimately succumbed to DNA damage, exhibiting signature necrotic characteristics. Importantly, this genotoxic stress-triggered necrosis was abrogated when either transcription or translation was inhibited. We pinpointed the p53-cathepsin axis as the quintessential framework underlying necrotic cell death. p53 induces cathepsin Q that cooperates with reactive oxygen species (ROS) to execute necrosis. Moreover, we presented the in vivo evidence of p53-activated necrosis in tumor allografts. Current study lays the foundation for future experimental and therapeutic discoveries aimed at "programmed necrotic death."
Collapse
|
49
|
Chou MY, Hsiao CD, Chen SC, Chen IW, Liu ST, Hwang PP. Effects of hypothermia on gene expression in zebrafish gills: upregulation in differentiation and function of ionocytes as compensatory responses. ACTA ACUST UNITED AC 2008; 211:3077-84. [PMID: 18805806 DOI: 10.1242/jeb.019950] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ectothermic vertebrates are different from mammals that are sensitive to hypothermia and have to maintain core temperature for survival. Why and how ectothermic animals survive, grow and reproduce in low temperature have been for a long time a scientifically challenging and important inquiry to biologists. We used a microarray to profile the gill transcriptome in zebrafish (Danio rerio) after exposure to low temperature. Adult zebrafish were acclimated to a low temperature of 12 degrees C for 1 day and up to 30 days, and the gill transcriptome was compared with that of control fish in 28 degrees C by oligonucleotide microarray hybridization. Results showed 11 and 22 transcripts were found to be upregulated, whereas 56 and 70 transcripts were downregulated by low-temperature treatment for 1 day and 30 days, respectively. The gill transcriptome profiles revealed that ionoregulation-related genes were highly upregulated in cold-acclimated zebrafish. This paved the way to investigate the role of ionoregulatory genes in zebrafish gills during cold acclimation. Cold acclimation caused upregulation of genes that are essential for ionocyte specification, differentiation, ionoregulation, acid-base balance and the number of cells expressing these genes increased. For instance, epithelial Ca2+ channel (EcaC; an ionoregulatory protein) mRNA increased in parallel with the level of Ca2+ influx, revealing a functional compensation after long-term acclimation to cold. Phosphohistone H3 and TUNEL staining showed that the cell turnover rate was retarded in cold-acclimated gills. Altogether, these results suggest that gills may sustain their functions by producing mature ionocytes from pre-existing undifferentiated progenitors in low-temperature environments.
Collapse
Affiliation(s)
- Ming-Yi Chou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | | | | | | | | | | |
Collapse
|
50
|
Polyanichko AM, Leonenko ZV, Cramb D, Wieser H, Vorob’ev VI, Chikhirzhina EV. Visualization of DNA complexes with HMGB1 and its C-truncated form HMGB1(A+B). Biophysics (Nagoya-shi) 2008. [DOI: 10.1134/s0006350908030044] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|