1
|
Xu H, Huang M, Zhou M, Guo R, Qin K, Dong Z. Construction of Silk Fibroin 3D Microfiber Scaffolds and Their Applications in Anti-Osteoporosis Drug Prediction. Molecules 2024; 29:5681. [PMID: 39683840 DOI: 10.3390/molecules29235681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Silk microfiber scaffolds have garnered increasing interest due to their outstanding properties, with degumming being the process used to extract the sericin from the cocoon. In the present study, an attempt to tune the biodegradation period of silk through degumming with various sodium borohydride (NaBH4) concentrations and degumming times was studied. We considered the process, the number of baths used, and the salt concentration. Herein, we report a novel method of expanding microfibers from two-dimensional (2D) to three-dimensional (3D) using a modified gas-foaming technique. Porous three-dimensional (3D) silk fibroin (SF) scaffolds were fabricated by the SF fibers, which were extracted by the NaBH4 degumming method and NaBH4 gas-foaming approach. This study showed that higher salt concentrations, reaching 1.5% in a double bath, effectively removed sericin from silk fibroin, resulting in clean, smooth 3D scaffolds. These scaffolds were then fabricated using a freeze-drying method. The scaffolds were then submerged in solutions containing semen cuscutae (SC) and their surfaces were coated with various percentages of total flavonoids. The scaffolds had no toxicity to the cells in vitro. This work provides a new route for achieving a TFSC-loaded scaffold; it is proved that the coated silk fibroin fiber scaffold has excellent compatibility. Compared with non-drug-loaded silk scaffolds, drug-loaded silk scaffolds promote cell growth.
Collapse
Affiliation(s)
- Hua Xu
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Mengfan Huang
- Southern Medical University, Guangzhou 511453, China
| | - Mengyuan Zhou
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Rong Guo
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Kunming Qin
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zibo Dong
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
2
|
Hwang N, Ghanta S, Li Q, Lamattina AM, Murzin E, Lederer JA, El-Chemaly S, Chung SW, Liu X, Perrella MA. Carbon monoxide-induced autophagy enhances human mesenchymal stromal cell function via paracrine actions in murine polymicrobial sepsis. Mol Ther 2024; 32:2232-2247. [PMID: 38734903 PMCID: PMC11286814 DOI: 10.1016/j.ymthe.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis is a life-threatening process due to organ dysfunction resulting from severe infections. Mesenchymal stromal cells (MSCs) are being investigated as therapy for sepsis, along with conditioning regimens to improve their function. Carbon monoxide (CO) gas, which is cytoprotective at low doses, induces autophagy and is a mediator of inflammation. We evaluated CO-induced autophagy in human MSCs (hMSCs), and its impact on cell function in murine cecal ligation and puncture. Conditioning of hMSCs with CO ex vivo resulted in enhanced survival and bacterial clearance in vivo, and neutrophil phagocytosis of bacteria in vitro. Decreased neutrophil infiltration and less parenchymal cell death in organs were associated with increased macrophage efferocytosis of apoptotic neutrophils, promoting resolution of inflammation. These CO effects were lost when the cells were exposed to autophagy inhibition prior to gas exposure. When assessing paracrine actions of CO-induced autophagy, extracellular vesicles (EVs) were predominantly responsible. CO had no effect on EV production, but altered their miRNA cargo. Increased expression of miR-145-3p and miR-193a-3p by CO was blunted with disruption of autophagy, and inhibitors of these miRNAs led to a loss of neutrophil phagocytosis and macrophage efferocytosis. Collectively, CO-induced autophagy enhanced hMSC function during sepsis via paracrine actions of MSC-derived EVs.
Collapse
Affiliation(s)
- Narae Hwang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sailaja Ghanta
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qifei Li
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Jackson Health System, Miami, FL, USA
| | - Anthony M Lamattina
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ekaterina Murzin
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Su Wol Chung
- School of Biological Sciences, University of Ulsan, Ulsan, South Korea
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Florczyk-Soluch U, Polak K, Jelinkova S, Bronisz-Budzyńska I, Sabo R, Bolisetty S, Agarwal A, Werner E, Józkowicz A, Stępniewski J, Szade K, Dulak J. Targeted expression of heme oxygenase-1 in satellite cells improves skeletal muscle pathology in dystrophic mice. Skelet Muscle 2024; 14:13. [PMID: 38867250 PMCID: PMC11167827 DOI: 10.1186/s13395-024-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.
Collapse
MESH Headings
- Animals
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Mice, Inbred mdx
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mice
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Male
- Mice, Inbred C57BL
- Physical Conditioning, Animal
- Membrane Proteins
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Sarka Jelinkova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Reece Sabo
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Laboratory of Stem Cells Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
4
|
Hong G, Li S, Zheng G, Zheng X, Zhan Q, Zhou L, Wei Q, He W, Chen Z. Therapeutic potential of a prominent dihydroxyflavanone pinocembrin for osteolytic bone disease: In vitro and in vivo evidence. J Orthop Translat 2024; 45:197-210. [PMID: 38685969 PMCID: PMC11056316 DOI: 10.1016/j.jot.2023.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 05/02/2024] Open
Abstract
Background/objective As the pivotal cellular mediators of bone resorption and pathological bone remodeling, osteoclasts have emerged as a prominent target for anti-resorptive interventions. Pinocembrin (PIN), a predominant flavonoid found in damiana, honey, fingerroot, and propolis, has been recognized for its potential therapeutic effects in osteolysis. The purpose of our project is to investigate the potential of PIN to prevent bone resorption in ovariectomized (OVX) mice by suppressing osteoclast production through its underlying mechanisms. Methods The study commenced by employing protein-ligand molecular docking to ascertain the specific interaction between PIN and nuclear factor-κB (NF-κB) ligand (RANKL). Subsequently, PIN was introduced to bone marrow macrophages (BMMs) under the stimulation of RANKL. The impact of PIN on osteoclastic activity was assessed through the utilization of a positive TRAcP staining kit and a hydroxyapatite resorption assay. Furthermore, the study investigated the generation of reactive oxygen species (ROS) in osteoclasts induced by RANKL using H2DCFDA. To delve deeper into the underlying mechanisms, molecular cascades triggered by RANKL, including NF-κB, ROS, calcium oscillations, and NFATc1-mediated signaling pathways, were explored using Luciferase gene report, western blot analysis, and quantitative real-time polymerase chain reaction. Moreover, an estrogen-deficient osteoporosis murine model was established to evaluate the therapeutic effects of PIN in vivo. Results In this study, we elucidated the profound inhibitory effects of PIN on osteoclastogenesis and bone resorption, achieved through repression of NF-κB and NFATc1-mediated signaling pathways. Notably, PIN also exhibited potent anti-oxidative properties by mitigating RANKL-induced ROS generation and augmenting activities of ROS-scavenging enzymes, ultimately leading to a reduction in intracellular ROS levels. Moreover, PIN effectively abrogated the expression of osteoclast-specific marker genes (Acp5, Cathepsin K, Atp6v0d2, Nfatc1, c-fos, and Mmp9), further underscoring its inhibitory impact on osteoclast differentiation and function. Additionally, employing an in vivo mouse model, we demonstrated that PIN effectively prevented osteoclast-induced bone loss resultant from estrogen deficiency. Conclusion Our findings highlight the potent inhibitory effects of PIN on osteoclastogenesis, bone resorption, and RANKL-induced signaling pathways, thereby establishing PIN as a promising therapeutic candidate for the prevention and management of osteolytic bone diseases. The translational potential of this article PIN serves as a promising therapeutic agent for the prevention and management of osteolytic bone diseases and holds promise for future clinical applications in addressing conditions characterized by excessive bone resorption. PIN is a natural compound found in various sources, including damiana, honey, fingerroot, and propolis. Its widespread availability and potential for therapeutic use make it an attractive candidate for further investigation and development as a clinical intervention.
Collapse
Affiliation(s)
- Guoju Hong
- Traumatology & Orthopaedics Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
| | - Shuqiang Li
- Department of Oncology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
| | - Guanqiang Zheng
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
| | - Xiaoxia Zheng
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, PR China
| | - Qunzhang Zhan
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, PR China
| | - Lin Zhou
- Department of Endocrinology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Qiushi Wei
- Traumatology & Orthopaedics Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
| | - Wei He
- Traumatology & Orthopaedics Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510378, PR China
| | - Zhenqiu Chen
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, PR China
| |
Collapse
|
5
|
Hong G, Zhou L, Zheng G, Zheng X, Chen Z, He W, Wei Q. A novel Glycyrrhiza glabra extract liquiritin targeting NFATc1 activity and ROS levels to counteract ovariectomy-induced osteoporosis and bone loss in murine model. Front Pharmacol 2023; 14:1287827. [PMID: 38026985 PMCID: PMC10663366 DOI: 10.3389/fphar.2023.1287827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Osteoporosis, a prevalent osteolytic condition worldwide, necessitates effective strategies to inhibit excessive bone resorption by curbing osteoclast hyperactivation. Liquiritin (LIQ), an flavanone derivative employed in acute lung injury and rheumatoid arthritis treatment, possesses an unclear role in addressing excessive bone resorption. In this investigation, we found that LIQ demonstrates the ability to inhibit osteoclast formation and the bone-resorbing activity induced by RANKL. At a specific concentration, LIQ significantly attenuated NF-κB-Luc activity induced by RANKL and curtailed NF-κB activation in RANKL-stimulated RAW264.7 cells, resulting in reduced IκB-α breakdown and diminished nuclear NF-κB levels. Furthermore, LIQ markedly inhibited RANKL-induced NFATc1 activation, as evidenced by diminished NFATc1 luciferase activity, reduced NFATc1 mRNA levels, and decreased nuclear NFATc1 protein levels. Subsequent experiments demonstrated that LIQ effectively restrained the RANKL-induced elevation of intracellular calcium as well as reactive oxygen species. Additionally, LIQ exhibited a downregulating effect on the expression of osteoclast-specific genes, which include Acp5, Cathepsin K, Atp6v0d2, Nfatc1, c-Fos, and Mmp9. Notably, our findings revealed the potential of LIQ to counteract decreased bone density in mice that underwent ovariectomy. Collectively, the data indicate that LIQ impedes osteoclast formation triggered by RANKL and the subsequent reduction in bone mass by mitigating ROS levels and suppressing the Ca2+/MAPK-NFATc1 signaling pathway, suggesting its promising candidacy as a therapeutic agent for RANKL-mediated osteoporosis.
Collapse
Affiliation(s)
- Guoju Hong
- Traumatology and Orthopedics Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Zhou
- Key Laboratory of Biological Targeting Diagnosis, Department of Endocrinology, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Guanqiang Zheng
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoxia Zheng
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhenqiu Chen
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei He
- Traumatology and Orthopedics Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qiushi Wei
- Traumatology and Orthopedics Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Horii S, Mori S, Ogata R, Nukaga S, Nishida R, Kishi S, Sasaki R, Ikemoto A, Owari T, Maesaka F, Honoki K, Miyake M, Tanaka Y, Fujimoto K, Fujiwara-Tani R, Kuniyasu H. 5-Aminolevrinic Acid Exhibits Dual Effects on Stemness in Human Sarcoma Cell Lines under Dark Conditions. Int J Mol Sci 2023; 24:ijms24076189. [PMID: 37047157 PMCID: PMC10094087 DOI: 10.3390/ijms24076189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
5-aminolevulinic acid (ALA) is used for tumor-targeting phototherapy because it is converted to protoporphyrin IX (PPIX) upon excitation and induces phototoxicity. However, the effect of ALA on malignant cells under unexcited conditions is unclear. This information is essential when administering ALA systemically. We used sarcoma cell lines that usually arise deep in the body and are rarely exposed to light to examine the effects of ALA treatment under light (daylight lamp irradiation) and dark (dark room) conditions. ALA-treated human SW872 liposarcoma cells and human MG63 osteosarcoma cells cultured under light exhibited growth suppression and increased oxidative stress, while cells cultured in the dark showed no change. However, sphere-forming ability increased in the dark, and the expression of stem-cell-related genes was induced in dark, but not light, conditions. ALA administration increased heme oxygenase 1 (HO-1) expression in both cell types; when carbon monoxide (CO), a metabolite of HO-1, was administered to sarcoma cells via carbon-monoxide-releasing molecule 2 (CORM2), it enhanced sphere-forming ability. We also compared the concentration of biliverdin (BVD) (a co-product of HO-1 activity alongside CO) with sphere-forming ability when HO-1 activity was inhibited using ZnPPIX in the dark. Both cell types showed a peak in sphere-forming ability at 60–80 μM BVD. Furthermore, a cell death inhibitor assay revealed that the HO-1-induced suppression of sphere formation was rescued by apoptosis or ferroptosis inhibitors. These findings suggest that in the absence of excitation, ALA promotes HO-1 expression and enhances the stemness of sarcoma cells, although excessive HO-1 upregulation induces apoptosis and ferroptosis. Our data indicate that systemic ALA administration induces both enhanced stemness and cell death in malignant cells located in dark environments deep in the body and highlight the need to pay attention to drug delivery and ALA concentrations during phototherapy.
Collapse
|
7
|
GSK 650394 Inhibits Osteoclasts Differentiation and Prevents Bone Loss via Promoting the Activities of Antioxidant Enzymes In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3458560. [PMID: 36164394 PMCID: PMC9509242 DOI: 10.1155/2022/3458560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Osteoporosis (OP) is one of the most common bone disorders among the elderly, characterized by abnormally elevated bone resorption caused by formation and activation of osteoblast (OC). Excessive reactive oxygen species (ROS) accumulation might contribute to the formation process of OC as an essential role. Although accumulated advanced treatment target on OP have been proposed in recent years, clinical outcomes remain unexcellence attributed to severe side effects. The purpose of present study was to explore the underlying mechanisms of GSK 650394 (GSK) on inhibiting formation and activation of OC and bone resorption in vitro and in vivo. GSK could inhibit receptor activator of nuclear-κB ligand (RANKL-)-mediated Oc formation via suppressing the activation of NF-κB and MAPK signaling pathways, regulating intracellular redox status, and downregulate the expression of nuclear factor of activated T cells c1 (NFATc1). In addition, quantitative RT-PCR results show that GSK could suppress the expression of OC marker gene and antioxidant enzyme genes. Consistent with in vitro cellular results, GSK treatment improved bone density in the mouse with ovariectomized-induced bone loss according to the results of CT parameters, HE staining, and Trap staining. Furthermore, GSK treatment could enhance the capacity of antioxidant enzymes in vivo. In conclusion, this study suggested that GSK could suppress the activation of osteoclasts and therefore maybe a potential therapeutic reagent for osteoclast activation-related osteoporosis.
Collapse
|
8
|
Zhou X, Yuan W, Xiong X, Zhang Z, Liu J, Zheng Y, Wang J, Liu J. HO-1 in Bone Biology: Potential Therapeutic Strategies for Osteoporosis. Front Cell Dev Biol 2021; 9:791585. [PMID: 34917622 PMCID: PMC8669958 DOI: 10.3389/fcell.2021.791585] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis is a prevalent bone disorder characterized by bone mass reduction and deterioration of bone microarchitecture leading to bone fragility and fracture risk. In recent decades, knowledge regarding the etiological mechanisms emphasizes that inflammation, oxidative stress and senescence of bone cells contribute to the development of osteoporosis. Studies have demonstrated that heme oxygenase 1 (HO-1), an inducible enzyme catalyzing heme degradation, exhibits anti-inflammatory, anti-oxidative stress and anti-apoptosis properties. Emerging evidence has revealed that HO-1 is critical in the maintenance of bone homeostasis, making HO-1 a potential target for osteoporosis treatment. In this Review, we aim to provide an introduction to current knowledge of HO-1 biology and its regulation, focusing specifically on its roles in bone homeostasis and osteoporosis. We also examine the potential of HO-1-based pharmacological therapeutics for osteoporosis and issues faced during clinical translation.
Collapse
Affiliation(s)
- Xueman Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxiu Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yingcheng Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jin Liu
- Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Kang IS, Kim RI, Kim C. Carbon Monoxide Regulates Macrophage Differentiation and Polarization toward the M2 Phenotype through Upregulation of Heme Oxygenase 1. Cells 2021; 10:3444. [PMID: 34943953 PMCID: PMC8700076 DOI: 10.3390/cells10123444] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Carbon monoxide (CO) is generated by heme oxygenase (HO), and HO-1 is highly induced in monocytes and macrophages upon stimulation. Monocytes differentiate into macrophages, including pro-inflammatory (M1) and anti-inflammatory (M2) cells, in response to environmental signals. The present study investigated whether CO modulates macrophage differentiation and polarization, by applying the CO-releasing molecule-3 (CORM-3). Results showed that murine bone marrow cells are differentiated into macrophages by CORM-3 in the presence of macrophage colony-stimulating factor. CORM-3 increases expressions of macrophage markers, including F4/80 and CD11b, and alters the cell morphology into elongated spindle-shaped cells, which is a typical morphology of M2 cells. CORM-3 upregulates the expressions of genes and molecules involved in M2 polarization and M2 phenotype markers, such as STAT6, PPARγ, Ym1, Fizz1, arginase-1, and IL-10. However, exposure to CORM-3 inhibits the iNOS expression, suggesting that CO enhances macrophage differentiation and polarization toward M2. Increased HO-1 expression is observed in differentiated macrophages, and CORM-3 further increases this expression. Hemin, an HO-1 inducer, results in increased macrophage differentiation, whereas the HO-1 inhibitor zinc protoporphyrin IX inhibits differentiation. In addition, CORM-3 increases the proportion of macrophages in peritoneal exudate cells and enhances the expression of HO-1 and arginase-1 but inhibits iNOS. Taken together, these results suggest that the abundantly produced CO in activated macrophages enhances proliferation, differentiation, and polarization toward M2. It will probably help clear apoptotic cells, resolve inflammation, and promote wound healing and tissue remodeling.
Collapse
Affiliation(s)
- In-Soon Kang
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea; (I.-S.K.); (R.-I.K.)
| | - Rang-Ie Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea; (I.-S.K.); (R.-I.K.)
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea; (I.-S.K.); (R.-I.K.)
- BK21 Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Korea
- Convergent Research Center for Metabolism and Immunoregulation, Inha University, Incheon 22212, Korea
| |
Collapse
|
10
|
Mei L, Zheng Y, Ma T, Xia B, Gao X, Hao Y, Luo Z, Huang J. The Novel Antioxidant Compound JSH-23 Prevents Osteolysis by Scavenging ROS During Both Osteoclastogenesis and Osteoblastogenesis. Front Pharmacol 2021; 12:734774. [PMID: 34566656 PMCID: PMC8458573 DOI: 10.3389/fphar.2021.734774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory osteolysis is a pathological skeletal disease associated with not only the production of inflammatory cytokines but also local oxidative status. Excessive reactive oxygen species (ROS) promote bone resorption by osteoclasts and induce the apoptosis of osteoblasts. In consideration of the lack of effective preventive or treatments options against osteolysis, the exploitation of novel pharmacological compounds/agents is critically required. In our study, we found that a novel antioxidant compound, JSH-23, plays a role in restoring bone homeostasis by scavenging intracellular ROS during both osteoclastogenesis and osteoblastogenesis. Mechanically, JSH-23 suppressed RANKL-induced osteoclastogenesis, bone resorption and the expression of specific genes (including NFATc1, c-Fos, TRAP, CTSK and DC-STAMP) via inhibition of the NF-κB signaling pathway. Meanwhile, JSH-23 suppressed RANKL-induced ROS generation via the TRAF6/Rac1/NOX1 pathway and the enhanced expression of Nrf2/HO-1. In addition, JSH-23 attenuated H2O2-induced apoptosis and mineralization reduction in osteoblasts by reducing ROS production and enhancing Nrf2/HO-1 expression. Our in vivo results further revealed that JSH-23 exerts its protective effects on bone mass through its antioxidant activity. In conclusion, our results show that the application of JSH-23 might be a novel and plausible strategy for the treatment of osteolysis-related disease.
Collapse
Affiliation(s)
- Liangwei Mei
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yi Zheng
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xue Gao
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Petrone MV, Toro A, Vazquez Echegaray C, Francia MG, Solari C, Cosentino MS, Vazquez E, Guberman A. The pluripotency transcription factor OCT4 represses heme oxygenase-1 gene expression. FEBS Lett 2021; 595:1949-1961. [PMID: 34056710 DOI: 10.1002/1873-3468.14138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/17/2021] [Accepted: 05/23/2021] [Indexed: 01/03/2023]
Abstract
In embryonic stem (ES) cells, oxidative stress control is crucial for genomic stability, self-renewal, and cell differentiation. Heme oxygenase-1 (HO-1) is a key player of the antioxidant system and is also involved in stem cell differentiation and pluripotency acquisition. We found that the HO-1 gene is expressed in ES cells and induced after promoting differentiation. Moreover, downregulation of the pluripotency transcription factor (TF) OCT4 increased HO-1 mRNA levels in ES cells, and analysis of ChIP-seq public data revealed that this TF binds to the HO-1 gene locus in pluripotent cells. Finally, ectopic expression of OCT4 in heterologous systems repressed a reporter carrying the HO-1 gene promoter and the endogenous gene. Hence, this work highlights the connection between pluripotency and redox homeostasis.
Collapse
Affiliation(s)
- María Victoria Petrone
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Ayelén Toro
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Camila Vazquez Echegaray
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Marcos Gabriel Francia
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Claudia Solari
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Maria Soledad Cosentino
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Elba Vazquez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales Universidad de Buenos Aires, Buenos Aires, Argentina.,IQUIBICEN, CONICET-UBA, Buenos Aires, Argentina
| | - Alejandra Guberman
- IQUIBICEN, CONICET-UBA, Buenos Aires, Argentina.,Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales Universidad de Buenos Aires, Argentina
| |
Collapse
|
12
|
Abstract
Nitric oxide, studied to evaluate its role in cardiovascular physiology, has cardioprotective and therapeutic effects in cellular signaling, mitochondrial function, and in regulating inflammatory processes. Heme oxygenase (major role in catabolism of heme into biliverdin, carbon monoxide (CO), and iron) has similar effects as well. CO has been suggested as the molecule that is responsible for many of the above mentioned cytoprotective and therapeutic pathways as CO is a signaling molecule in the control of physiological functions. This is counterintuitive as toxic effects are related to its binding to hemoglobin. However, CO is normally produced in the body. Experimental evidence indicates that this toxic gas, CO, exerts cytoprotective properties related to cellular stress including the heart and is being assessed for its cytoprotective and cytotherapeutic properties. While survival of adult cardiomyocytes depends on oxidative phosphorylation (survival and resulting cardiac function is impaired by mitochondrial damage), mitochondrial biogenesis is modified by the heme oxygenase-1/CO system and can result in promotion of mitochondrial biogenesis by associating mitochondrial redox status to the redox-active transcription factors. It has been suggested that the heme oxygenase-1/CO system is important in differentiation of embryonic stem cells and maturation of cardiomyocytes which is thought to mitigate progression of degenerative cardiovascular diseases. Effects on other cardiac cells are being studied. Acute exposure to air pollution (and, therefore, CO) is associated with cardiovascular mortality, myocardial infarction, and heart failure, but changes in the endogenous heme oxygenase-1 system (and, thereby, CO) positively affect cardiovascular health. We will review the effect of CO on heart health and function in this article.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Si Z, Wang X, Kang Y, Wang X, Sun C, Li Y, Xu J, Wu J, Zhang Z, Li L, Peng Y, Li J, Sun C, Hui Y, Gao X. Heme Oxygenase 1 Inhibits Adult Neural Stem Cells Proliferation and Survival via Modulation of Wnt/β-Catenin Signaling. J Alzheimers Dis 2021; 76:623-641. [PMID: 32568195 DOI: 10.3233/jad-200114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Adult hippocampal neurogenesis is critical for renewing hippocampal neural circuits and maintaining hippocampal cognitive function and is closely associated with age-related neurodegenerative diseases. Heme oxygenase 1 (HO-1) is a stress protein that catalyzes the degradation of heme into free iron, biliverdin, and carbon monoxide. Elevated HO-1 level constitutes a pathological feature of Alzheimer's disease, Parkinson's disease, and many other age-related neurodegenerative diseases. OBJECTIVE Here we research the precise role of HO-1 in adult hippocampal neurogenesis. METHODS To explore the effect of HO-1 overexpression on adult neural stem cells (aNSCs) and elucidate its mechanisms, Tg(HO-1) was constructed. The transgenic mice and aNSCs were subjected to neurosphereing assay, clonal analysis, and BrdU labelling to detect the proliferation and self-renewal ability. LiCl, MG132, CHX, and IGF-1 treatment were used to research the signaling pathways which regulated by HO-1. RESULTS HO-1 overexpression decreased proliferation ability and induced apoptosis of aNSCs in subgranular zoon (SGZ) in vivo and in vitro. Furthermore, HO-1 overexpression inactivated canonical WNT/β-catenin pathway. Re-activate canonical WNT/β-catenin pathway rescued aNSCs proliferation and survival upon HO-1 overexpression. More importantly, phosphorylation of AKTS473 and GSK3βS9 was found to be significantly decreased in HO-1 overexpressed aNSCs. Re-activation of AKT signaling proved that HO-1 inhibited Wnt/β-catenin signaling pathway via AKT/GSK3β signaling pathway. CONCLUSION These results demonstrated a critical role of HO-1 in regulating aNSCs survival and proliferation by inhibiting Wnt/β-catenin pathway through repression of AKT/GSK3β, which provide a novel insight into the role of HO-1 in Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Zizhen Si
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Xue Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Yuchun Kang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Xidi Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China.,State-Province Key Laboratories of Biomedicine-Pharmaceutics of China
| | - Changhui Sun
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Yuanxin Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Jiakun Xu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Jiajia Wu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Zhujun Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Ling Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Yahui Peng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China.,State-Province Key Laboratories of Biomedicine-Pharmaceutics of China
| | - Jihong Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China.,State-Province Key Laboratories of Biomedicine-Pharmaceutics of China
| | - Chongran Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University Medical School, Zhejiang, China
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China.,State-Province Key Laboratories of Biomedicine-Pharmaceutics of China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China.,State-Province Key Laboratories of Biomedicine-Pharmaceutics of China
| |
Collapse
|
14
|
Hong G, Chen Z, Han X, Zhou L, Pang F, Wu R, Shen Y, He X, Hong Z, Li Z, He W, Wei Q. A novel RANKL-targeted flavonoid glycoside prevents osteoporosis through inhibiting NFATc1 and reactive oxygen species. Clin Transl Med 2021; 11:e392. [PMID: 34047464 PMCID: PMC8140192 DOI: 10.1002/ctm2.392] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 03/31/2021] [Accepted: 04/07/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Osteoporosis is characterized by excessive bone resorption due to enhanced osteoclast activation. Stimulation of nuclear factor of activated T cells 1 (NFATc1) and accumulation of reactive oxygen species (ROS) are important mechanisms underlying osteoclastogenesis. Robinin (Rob) is a flavonoid glycoside that has shown anti-inflammatory and antioxidative effects in previous studies, but little is known about its effects on bone homeostasis. The purpose of our research was to investigate whether Rob could prevent bone resorption in ovariectomized (OVX) mice by suppressing osteoclast production through its underlying mechanisms. METHODS The docking pose of Rob and RANKL was identified by protein-ligand molecular docking. Rob was added to bone marrow macrophages (BMMs) stimulated by nuclear factor-κB (NF-κB) ligand (RANKL). The effects of Rob on osteoclastic activity were evaluated by positive tartrate resistant acid phosphatase (TRAcP) staining kit and hydroxyapatite resorption assay. RANKL-induced ROS generation in osteoclasts was detected by H2 DCFDA and MitoSox Red staining. The classic molecular cascades triggered by RANKL, such as NF-κB, ROS, calcium oscillations, and NFATc1-mediated signaling pathways, were investigated using Fluo4 staining, western blot, and quantitative real-time polymerase chain reaction. In addition, an OVX mouse model mimicking estrogen-deficient osteoporosis was created to evaluate the therapeutic effects of Rob in vivo. RESULTS Computational docking results showed that Rob could bind specifically to RANKL's predicted binding sites. In vitro, Rob inhibited RANKL-mediated osteoclastogenesis dose-dependently without obvious cytotoxicity at low concentrations. We also found that Rob attenuated RANKL-induced mitochondrial ROS production or enhanced activities of ROS-scavenging enzymes, and ultimately reduced intracellular ROS levels. Rob abrogated the RANKL-induced mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways, and subsequently blocked NFATc1 signaling and TRAcP expression. In addition, Rob inhibited osteoclast proliferation by downregulating the expression of osteoclast target genes (Acp5, Cathepsin K, Atp6v0d2, Nfact1, c-Fos, and Mmp9) and reducing Ca2+ oscillations. Our in vivo results showed that Rob reduced bone resorption in OVX animal model by repressing osteoclast activity and function. CONCLUSIONS Rob inhibits the activation of osteoclasts by targeting RANKL and is therefore a potential osteoporosis drug.
Collapse
Affiliation(s)
- Guoju Hong
- Division of Orthopaedic SurgeryThe University of AlbertaEdmontonAlbertaCanada
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Zhenqiu Chen
- Department of OrthopaedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Xiaorui Han
- Division of Bioengineering, School of MedicineSouth China University of TechnologyGuangzhouGuangdongP.R. China
| | - Lin Zhou
- Department of Endocrinologythe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongP.R. China
| | - Fengxiang Pang
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Rishana Wu
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Yingshan Shen
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Xiaoming He
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Zhinan Hong
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- Department of OrthopaedicsThe Third Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Ziqi Li
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- Department of OrthopaedicsThe Third Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Wei He
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- Department of OrthopaedicsThe Third Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Qiushi Wei
- Traumatology and Orthopedics InstituteGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
- Department of OrthopaedicsThe Third Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| |
Collapse
|
15
|
Role of Heme-Oxygenase-1 in Biology of Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells. Cells 2021; 10:cells10030522. [PMID: 33804563 PMCID: PMC8000937 DOI: 10.3390/cells10030522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. HO-1 was demonstrated to affect cardiac differentiation of murine pluripotent stem cells (PSCs), regulate the metabolism of murine adult cardiomyocytes, and influence regeneration of infarcted myocardium in mice. However, the enzyme’s effect on human cardiogenesis and human cardiomyocytes’ electromechanical properties has not been described so far. Thus, this study aimed to investigate the role of HO-1 in the differentiation of human induced pluripotent stem cells (hiPSCs) into hiPSC-derived cardiomyocytes (hiPSC-CMs). hiPSCs were generated from human fibroblasts and peripheral blood mononuclear cells using Sendai vectors and subjected to CRISPR/Cas9-mediated HMOX1 knock-out. After confirming lack of HO-1 expression on the protein level, isogenic control and HO-1-deficient hiPSCs were differentiated into hiPSC-CMs. No differences in differentiation efficiency and hiPSC-CMs metabolism were observed in both cell types. The global transcriptomic analysis revealed, on the other hand, alterations in electrophysiological pathways in hiPSC-CMs devoid of HO-1, which also demonstrated increased size. Functional consequences in changes in expression of ion channels genes were then confirmed by patch-clamp analysis. To the best of our knowledge, this is the first report demonstrating the link between HO-1 and electrophysiology in human cardiomyocytes.
Collapse
|
16
|
Heme Oxgenase-1, a Cardinal Modulator of Regulated Cell Death and Inflammation. Cells 2021; 10:cells10030515. [PMID: 33671004 PMCID: PMC7997353 DOI: 10.3390/cells10030515] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Heme oxygenase catalyzes the rate-limiting step in heme degradation in order to generate biliverdin, carbon monoxide (CO), and iron. The inducible form of the enzyme, heme oxygenase-1 (HO-1), exerts a central role in cellular protection. The substrate, heme, is a potent pro-oxidant that can accelerate inflammatory injury and promote cell death. HO-1 has been implicated as a key mediator of inflammatory cell and tissue injury, as validated in preclinical models of acute lung injury and sepsis. A large body of work has also implicated HO-1 as a cytoprotective molecule against various forms of cell death, including necrosis, apoptosis and newly recognized regulated cell death (RCD) programs such as necroptosis, pyroptosis, and ferroptosis. While the antiapoptotic potential of HO-1 and its reaction product CO in apoptosis regulation has been extensively characterized, relatively fewer studies have explored the regulatory role of HO-1 in other forms of necrotic and inflammatory RCD (i.e., pyroptosis, necroptosis and ferroptosis). HO-1 may provide anti-inflammatory protection in necroptosis or pyroptosis. In contrast, in ferroptosis, HO-1 may play a pro-death role via enhancing iron release. HO-1 has also been implicated in co-regulation of autophagy, a cellular homeostatic program for catabolic recycling of proteins and organelles. While autophagy is primarily associated with cell survival, its occurrence can coincide with RCD programs. This review will summarize the roles of HO-1 and its reaction products in co-regulating RCD and autophagy programs, with its implication for both protective and detrimental tissue responses, with emphasis on how these impact HO-1 as a candidate therapeutic target in disease.
Collapse
|
17
|
Bukowska-Strakova K, Włodek J, Pitera E, Kozakowska M, Konturek-Cieśla A, Cieśla M, Gońka M, Nowak W, Wieczorek A, Pawińska-Wąsikowska K, Józkowicz A, Siedlar M. Role of HMOX1 Promoter Genetic Variants in Chemoresistance and Chemotherapy Induced Neutropenia in Children with Acute Lymphoblastic Leukemia. Int J Mol Sci 2021; 22:ijms22030988. [PMID: 33498175 PMCID: PMC7863945 DOI: 10.3390/ijms22030988] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/15/2021] [Indexed: 12/17/2022] Open
Abstract
Whilst the survival rates of childhood acute lymphoblastic leukemia (ALL) have increased remarkably over the last decades, the therapy resistance and toxicity are still the major causes of treatment failure. It was shown that overexpression of heme oxygenase-1 (HO-1) promotes proliferation and chemoresistance of cancer cells. In humans, the HO-1 gene (HMOX1) expression is modulated by two polymorphisms in the promoter region: (GT)n-length polymorphism and single-nucleotide polymorphism (SNP) A(−413)T, with short GT repeat sequences and 413-A variants linked to an increased HO-1 inducibility. We found that the short alleles are significantly more frequent in ALL patients in comparison to the control group, and that their presence may be associated with a higher risk of treatment failure, reflecting the role of HO-1 in chemoresistance. We also observed that the presence of short alleles may predispose to develop chemotherapy-induced neutropenia. In case of SNP, the 413-T variant co-segregated with short or long alleles, while 413-A almost selectively co-segregated with long alleles, hence it is not possible to determine if SNPs are actually of phenotypic significance. Our results suggest that HO-1 can be a potential target to overcome the treatment failure in ALL patients.
Collapse
Affiliation(s)
- Karolina Bukowska-Strakova
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 31-663 Kraków, Poland; (J.W.); (E.P.)
- Correspondence: (K.B.-S.); (A.J.); (M.S.); Tel.: +48-(12)-664-6411 (A.J.); +48-(12)-658-2486 (M.S.); Fax: +48-(12)-658-1756 (M.S.)
| | - Joanna Włodek
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 31-663 Kraków, Poland; (J.W.); (E.P.)
| | - Ewelina Pitera
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 31-663 Kraków, Poland; (J.W.); (E.P.)
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Kraków, Poland; (M.K.); (A.K.-C.); (M.C.); (M.G.); (W.N.)
| | - Anna Konturek-Cieśla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Kraków, Poland; (M.K.); (A.K.-C.); (M.C.); (M.G.); (W.N.)
| | - Maciej Cieśla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Kraków, Poland; (M.K.); (A.K.-C.); (M.C.); (M.G.); (W.N.)
| | - Monika Gońka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Kraków, Poland; (M.K.); (A.K.-C.); (M.C.); (M.G.); (W.N.)
| | - Witold Nowak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Kraków, Poland; (M.K.); (A.K.-C.); (M.C.); (M.G.); (W.N.)
| | - Aleksandra Wieczorek
- Pediatric, Oncology and Hematology Department, Institute of Pediatrics, Jagiellonian University Medical College, 30-387 Krakow, Poland; (A.W.); (K.P.-W.)
| | - Katarzyna Pawińska-Wąsikowska
- Pediatric, Oncology and Hematology Department, Institute of Pediatrics, Jagiellonian University Medical College, 30-387 Krakow, Poland; (A.W.); (K.P.-W.)
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Kraków, Poland; (M.K.); (A.K.-C.); (M.C.); (M.G.); (W.N.)
- Correspondence: (K.B.-S.); (A.J.); (M.S.); Tel.: +48-(12)-664-6411 (A.J.); +48-(12)-658-2486 (M.S.); Fax: +48-(12)-658-1756 (M.S.)
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 31-663 Kraków, Poland; (J.W.); (E.P.)
- Correspondence: (K.B.-S.); (A.J.); (M.S.); Tel.: +48-(12)-664-6411 (A.J.); +48-(12)-658-2486 (M.S.); Fax: +48-(12)-658-1756 (M.S.)
| |
Collapse
|
18
|
Toro A, Anselmino N, Solari C, Francia M, Oses C, Sanchis P, Bizzotto J, Vazquez Echegaray C, Petrone MV, Levi V, Vazquez E, Guberman A. Novel Interplay between p53 and HO-1 in Embryonic Stem Cells. Cells 2020; 10:cells10010035. [PMID: 33383653 PMCID: PMC7823265 DOI: 10.3390/cells10010035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
Stem cells genome safeguarding requires strict oxidative stress control. Heme oxygenase-1 (HO-1) and p53 are relevant components of the cellular defense system. p53 controls cellular response to multiple types of harmful stimulus, including oxidative stress. Otherwise, besides having a protective role, HO-1 is also involved in embryo development and in embryonic stem (ES) cells differentiation. Although both proteins have been extensively studied, little is known about their relationship in stem cells. The aim of this work is to explore HO-1-p53 interplay in ES cells. We studied HO-1 expression in p53 knockout (KO) ES cells and we found that they have higher HO-1 protein levels but similar HO-1 mRNA levels than the wild type (WT) ES cell line. Furthermore, cycloheximide treatment increased HO-1 abundance in p53 KO cells suggesting that p53 modulates HO-1 protein stability. Notably, H2O2 treatment did not induce HO-1 expression in p53 KO ES cells. Finally, SOD2 protein levels are also increased while Sod2 transcripts are not in KO cells, further suggesting that the p53 null phenotype is associated with a reinforcement of the antioxidant machinery. Our results demonstrate the existence of a connection between p53 and HO-1 in ES cells, highlighting the relationship between these stress defense pathways.
Collapse
Affiliation(s)
- Ayelén Toro
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Nicolás Anselmino
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Claudia Solari
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Marcos Francia
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Camila Oses
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Pablo Sanchis
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Juan Bizzotto
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Camila Vazquez Echegaray
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - María Victoria Petrone
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Valeria Levi
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
| | - Elba Vazquez
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Correspondence: (E.V.); (A.G.); Tel.: +54-91144087796 (E.V.); +54-115-285-8683 (A.G.)
| | - Alejandra Guberman
- CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (A.T.); (N.A.); (C.S.); (M.F.); (C.O.); (P.S.); (J.B.); (C.V.E.); (M.V.P.); (V.L.)
- Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Correspondence: (E.V.); (A.G.); Tel.: +54-91144087796 (E.V.); +54-115-285-8683 (A.G.)
| |
Collapse
|
19
|
Nuclear heme oxygenase-1 improved the hypoxia-mediated dysfunction of blood-spinal cord barrier via the miR-181c-5p/SOX5 signaling pathway. Neuroreport 2020; 32:112-120. [PMID: 33290309 DOI: 10.1097/wnr.0000000000001568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Our previous study demonstrated that adenovirus-delivered GFP nuclear heme oxygenase-1 (nuclear HO-1, NHO-1) fragments lacking 23 amino acids at the C-terminus (Ad-GFP-HO-1C[INCREMENT]23) showed the potential therapeutic effects mediated by its improvement of the blood-spinal cord barrier (BSCB) integrity. However, the NHO-1-mediated molecular mechanism in regulating the BSCB function remains unclear. The BSCB model in vitro was established via a coculture of primary rat brain microvascular endothelial cells (RBMECs) and spinal cord astrocytes on transwell system. NHO-1 markedly reduced the disruption of the BSCB integrity induced by hypoxia. And NHO-1 significantly attenuated the expression of miR-181c-5p, but increased the expression level of SOX5 protein. miR-181c-5p was shown as an essential miRNA for increasing the BSCB permeability under hypoxia condition. Furthermore, we identified that miR-181c-5p could regulate the expression of SOX5 through binding to the 3'-UTR of its mRNA. And the decreased BSCB permeability and upregulation of tight junction (TJ) protein expression induced by NHO-1 could be partly reversed by the inhibition of SOX5 or miR-181c-5p (+). The present study results provide a better understanding of the molecular mechanisms induced by NHO-1 in improving the BSCB integrity, which is associated with the regulation of miR-181c-5p/SOX5/TJ signaling pathway.
Collapse
|
20
|
Suzuki K, Matsumoto M, Katoh Y, Liu L, Ochiai K, Aizawa Y, Nagatomi R, Okuno H, Itoi E, Igarashi K. Bach1 promotes muscle regeneration through repressing Smad-mediated inhibition of myoblast differentiation. PLoS One 2020; 15:e0236781. [PMID: 32776961 PMCID: PMC7416950 DOI: 10.1371/journal.pone.0236781] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
It has been reported that Bach1-deficient mice show reduced tissue injuries in diverse disease models due to increased expression of heme oxygenase-1 (HO-1)that possesses an antioxidant function. In contrast, we found that Bach1 deficiency in mice exacerbated skeletal muscle injury induced by cardiotoxin. Inhibition of Bach1 expression in C2C12 myoblast cells using RNA interference resulted in reduced proliferation, myotube formation, and myogenin expression compared with control cells. While the expression of HO-1 was increased by Bach1 silencing in C2C12 cells, the reduced myotube formation was not rescued by HO-1 inhibition. Up-regulations of Smad2, Smad3 and FoxO1, known inhibitors of muscle cell differentiation, were observed in Bach1-deficient mice and Bach1-silenced C2C12 cells. Therefore, Bach1 may promote regeneration of muscle by increasing proliferation and differentiation of myoblasts.
Collapse
Affiliation(s)
- Katsushi Suzuki
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yasutake Katoh
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Japan Agency for Medical Research and Development, Chiyoda, Tokyo, Japan
| | - Liang Liu
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yuta Aizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ryoichi Nagatomi
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroshi Okuno
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Orthopaedic Surgery, Tohoku Rosai Hospital, Sendai, Miyagi, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- * E-mail:
| |
Collapse
|
21
|
Jin W, Zhu X, Yao F, Xu X, Chen X, Luo Z, Zhao D, Li X, Leng X, Sun L. Cytoprotective effect of Fufang Lurong Jiangu capsule against hydrogen peroxide-induced oxidative stress in bone marrow stromal cell-derived osteoblasts through the Nrf2/HO-1 signaling pathway. Biomed Pharmacother 2019; 121:109676. [PMID: 31810119 DOI: 10.1016/j.biopha.2019.109676] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Oxidative stress is increasingly recognized as a risk factor associated with the development and progression of osteoporosis. Fufang Lurong Jiangu Capsule (FLJC) has a known anti-osteoporotic effect, but its pharmacological effect on osteoblasts is not clearly understood. This study was designed to investigate FLJC effects/mechanisms on in vitro hydrogen peroxide (H2O2)-induced oxidative damage of osteoblasts and on in vivo lipopolysaccharide (LPS)-induced mice bone loss. FLJC alleviates osteoporosis via unknown pharmacological mechanisms. METHODS Chemical compositions of FLJC preparations were analyzed using high-performance liquid chromatographic fingerprinting. After rat bone marrow mesenchymal stem cell differentiation induction, resulting osteoblasts received various 48 h FLJC pretreatments before H2O2-based (200 μM) oxidative stress exposure. FLJC effects were measured on osteoblast cell viability, morphological changes, levels of intracellular reactive oxygen species (ROS), localization of mitochondria, activity of antioxidant enzymes, alkaline phosphatase (ALP) and mineralization, the secretion of Col I and expression of osteogenic markers. The percentages of apoptosis were determined by flow cytometric analysis; apoptosis-related protein levels, including nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1 (HO-1) with or without Nrf2 inhibitor were analyzed via western blot. Hematoxylin and eosin (H&E) and ALP staining revealed in vivo FLJC effect on mice LPS-induced bone loss. RESULTS Five chemical components in FLJC were identified, and fingerprint analysis showed good reproducibility. FLJC pretreatment significantly reduced H2O2-induced ROS levels in osteoblasts and increased antioxidant enzyme activities to reduce oxidative damage. With regard to osteoblast differentiation, FLJC pretreatment increased ALP expression, as well as levels of mineralization and osteoblast markers. Additionally, FLJC protected against H2O2-induced apoptosis by inhibiting changes in expression of major Bcl-2 family effector proteins of the mitochondrial apoptosis pathway. Furthermore, FLJC protected cells from H2O2-induced oxidative damage by up-regulating Nrf2 and HO-1 protein levels. Finally, we confirmed that FLJC administration could reverse the bone loss in LPS-induced mice. CONCLUSION These results indicate that FLJC may significantly attenuate oxidative damage of osteoblasts induced by H2O2 via the Nrf2/HO-1 signaling pathway, providing new insights to guide development of treatments for osteoporosis induced by oxidative injury.
Collapse
Affiliation(s)
- Wenqi Jin
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaoqian Zhu
- Technology Innovation Center for Chinese Medicine Biotechnology, College of Science, Beihua University, Jilin, Jilin, China
| | - Fan Yao
- Center of Preventive Treatment of Diseases, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaohao Xu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xuenan Chen
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zongjian Luo
- Department of Orthopedics, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Daqing Zhao
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Ginseng Academy, Changchun University of Chinese Medicine, Jilin, China
| | - Xiangyan Li
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Ginseng Academy, Changchun University of Chinese Medicine, Jilin, China
| | - Xiangyang Leng
- Department of Orthopedics, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China.
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China.
| |
Collapse
|
22
|
Bottino-Rojas V, Pereira LOR, Silva G, Talyuli OAC, Dunkov BC, Oliveira PL, Paiva-Silva GO. Non-canonical transcriptional regulation of heme oxygenase in Aedes aegypti. Sci Rep 2019; 9:13726. [PMID: 31551499 PMCID: PMC6760526 DOI: 10.1038/s41598-019-49396-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenase (HO) is a ubiquitous enzyme responsible for heme breakdown, which yields carbon monoxide (CO), biliverdin (BV) and ferrous ion. Here we show that the Aedes aegypti heme oxygenase gene (AeHO - AAEL008136) is expressed in different developmental stages and tissues. AeHO expression increases after a blood meal in the midgut, and its maximal transcription levels overlaps with the maximal rate of the further modified A. aegypti biglutaminyl-biliverdin (AeBV) pigment production. HO is a classical component of stress response in eukaryotic cells, being activated under oxidative stress or increased heme levels. Indeed, the final product of HO activity in the mosquito midgut, AeBV, exerts a protective antioxidant activity. AeHO, however, does not seem to be under a classical redox-sensitive transcriptional regulation, being unresponsive to heme itself, and even down regulated when insects face a pro-oxidant insult. In contrast, AeHO gene expression responds to nutrient sensing mechanisms, through the target of rapamycin (TOR) pathway. This unusual transcriptional control of AeHO, together with the antioxidant properties of AeBV, suggests that heme degradation by HO, in addition to its important role in protection of Aedes aegypti against heme exposure, also acts as a digestive feature, being an essential adaptation to blood feeding.
Collapse
Affiliation(s)
- Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Luiza O R Pereira
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, 21040-360, Brazil
| | - Gabriela Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Boris C Dunkov
- Center for Insect Science, The University of Arizona, Tucson, AZ, 85721-0106, USA
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
23
|
Zhou Z, Ma D, Liu P, Wang P, Wei D, Yu K, Li P, Fang Q, Wang J. Deletion of HO-1 blocks development of B lymphocytes in mice. Cell Signal 2019; 63:109378. [PMID: 31369826 DOI: 10.1016/j.cellsig.2019.109378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 12/29/2022]
Abstract
B lymphocytes, a key cluster of cells composing the immune system, can protect against abnormal biological factors. Heme oxygenase-1 (HO-1) plays important roles in cell proliferation and immune regulation, but its effects on the development and growth of B lymphocytes are still unknown. Herein, the count of B lymphocytes in HO-1 gene knockout (HO-1+/-) mice was significantly lower than that of the HO-1 gene wild-type (HO-1WT) mice. Meanwhile, the cell count of HO-1+/- mice did not recover after irradiation for one week, due to the G0/G1 phase arrest of Pro-B cells and the augmented apoptosis of Pre-B cells. Up-regulation of HO-1 by lentivirus attenuated the Pro-B cell cycle arrest and Pre-B cell apoptosis. To understand the molecular mechanism by which HO-1 knockout blocked B lymphocyte development, protein-to-protein interaction network and Western blot were used. The PI3K/AKT signaling pathway mediated the regulatory effects of HO-1 on B lymphocytes. In conclusion, HO-1 is a crucial transcriptional repressor for B cell development.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Ping Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Danna Wei
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Kunling Yu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Peifan Li
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China.
| |
Collapse
|
24
|
Hörbelt T, Tacke C, Markova M, Herzfeld de Wiza D, Van de Velde F, Bekaert M, Van Nieuwenhove Y, Hornemann S, Rödiger M, Seebeck N, Friedl E, Jonas W, Thoresen GH, Kuss O, Rosenthal A, Lange V, Pfeiffer AFH, Schürmann A, Lapauw B, Rudovich N, Pivovarova O, Ouwens DM. The novel adipokine WISP1 associates with insulin resistance and impairs insulin action in human myotubes and mouse hepatocytes. Diabetologia 2018; 61:2054-2065. [PMID: 29754289 DOI: 10.1007/s00125-018-4636-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/12/2018] [Indexed: 01/19/2023]
Abstract
AIMS/HYPOTHESIS Wingless-type (Wnt) inducible signalling pathway protein-1 (WISP1) has been recently identified as a proinflammatory adipokine. We examined whether WISP1 expression and circulating levels are altered in type 2 diabetes and whether WISP1 affects insulin signalling in muscle cells and hepatocytes. METHODS Serum and visceral adipose tissue (VAT) biopsies, for analysis of circulating WISP1 levels by ELISA and WISP1 mRNA expression by real-time quantitative RT-PCR, were collected from normal-weight men (control group, n = 33) and obese men with (n = 46) and without type 2 diabetes (n = 56) undergoing surgery. Following incubation of primary human skeletal muscle cells (hSkMCs) and murine AML12 hepatocytes with WISP1 and insulin, insulin signalling was analysed by western blotting. The effect of WISP1 on insulin-stimulated glycogen synthesis and gluconeogenesis was investigated in hSkMCs and murine hepatocytes, respectively. RESULTS Circulating WISP1 levels were higher in obese men (independent of diabetes status) than in normal-weight men (mean [95% CI]: 70.8 [55.2, 86.4] ng/l vs 42.6 [28.5, 56.6] ng/l, respectively; p < 0.05). VAT WISP1 expression was 1.9-fold higher in obese men vs normal-weight men (p < 0.05). Circulating WISP1 levels were positively associated with blood glucose in the OGTT and circulating haem oxygenase-1 and negatively associated with adiponectin levels. In hSkMCs and AML12 hepatocytes, recombinant WISP1 impaired insulin action by inhibiting phosphorylation of insulin receptor, Akt and its substrates glycogen synthase kinase 3β, FOXO1 and p70S6 kinase, and inhibiting insulin-stimulated glycogen synthesis and suppression of gluconeogenic genes. CONCLUSIONS/INTERPRETATION Circulating WISP1 levels and WISP1 expression in VAT are increased in obesity independent of glycaemic status. Furthermore, WISP1 impaired insulin signalling in muscle and liver cells.
Collapse
Affiliation(s)
- Tina Hörbelt
- Institute for Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | - Christopher Tacke
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité University Medicine, Berlin, Germany
| | - Mariya Markova
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Daniella Herzfeld de Wiza
- Institute for Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | | | - Marlies Bekaert
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | | | - Silke Hornemann
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Maria Rödiger
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam, Germany
| | - Nicole Seebeck
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Elisabeth Friedl
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Wenke Jonas
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam, Germany
| | - G Hege Thoresen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oliver Kuss
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Duesseldorf, Germany
| | | | - Volker Lange
- Center for Obesity and Metabolic Surgery, Vivantes Hospital, Berlin, Germany
- Helios Hospital Berlin-Buch, Berlin, Germany
| | - Andreas F H Pfeiffer
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité University Medicine, Berlin, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam, Germany
| | - Bruno Lapauw
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Natalia Rudovich
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité University Medicine, Berlin, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Spital Bülach, Bülach, Switzerland
| | - Olga Pivovarova
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany.
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
- Department of Endocrinology, Diabetes and Nutrition, Charité University Medicine, Berlin, Germany.
| | - D Margriet Ouwens
- Institute for Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
25
|
Ferreira AF, Calin GA, Picanço-Castro V, Kashima S, Covas DT, de Castro FA. Hematopoietic stem cells from induced pluripotent stem cells - considering the role of microRNA as a cell differentiation regulator. J Cell Sci 2018; 131:131/4/jcs203018. [PMID: 29467236 DOI: 10.1242/jcs.203018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although hematopoietic stem cell (HSC) therapy for hematological diseases can lead to a good outcome from the clinical point of view, the limited number of ideal donors, the comorbidity of patients and the increasing number of elderly patients may limit the application of this therapy. HSCs can be generated from induced pluripotent stem cells (iPSCs), which requires the understanding of the bone marrow and liver niches components and function in vivo iPSCs have been extensively applied in several studies involving disease models, drug screening and cellular replacement therapies. However, the somatic reprogramming by transcription factors is a low-efficiency process. Moreover, the reprogramming process is also regulated by microRNAs (miRNAs), which modulate the expression of the transcription factors OCT-4 (also known as POU5F1), SOX-2, KLF-4 and MYC, leading somatic cells to a pluripotent state. In this Review, we present an overview of the challenges of cell reprogramming protocols with regard to HSC generation from iPSCs, and highlight the potential role of miRNAs in cell reprogramming and in the differentiation of induced pluripotent stem cells.
Collapse
Affiliation(s)
- Aline F Ferreira
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - George A Calin
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Virgínia Picanço-Castro
- Center of Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Simone Kashima
- Center of Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Dimas T Covas
- Center of Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, São Paulo 14051-140, Brazil.,Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Fabiola A de Castro
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| |
Collapse
|
26
|
Stepniewski J, Pacholczak T, Skrzypczyk A, Ciesla M, Szade A, Szade K, Bidanel R, Langrzyk A, Grochowski R, Vandermeeren F, Kachamakova-Trojanowska N, Jez M, Drabik G, Nakanishi M, Jozkowicz A, Dulak J. Heme oxygenase-1 affects generation and spontaneous cardiac differentiation of induced pluripotent stem cells. IUBMB Life 2018; 70:129-142. [DOI: 10.1002/iub.1711] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/20/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Jacek Stepniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Tomasz Pacholczak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Aniela Skrzypczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Maciej Ciesla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Romain Bidanel
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | | | - Radoslaw Grochowski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Felix Vandermeeren
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Mateusz Jez
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Grazyna Drabik
- Pathology Department; University Children's Hospital of Cracow; Krakow Poland
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST); Tsukuba Ibaraki Japan
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
- Kardio-Med Silesia; Zabrze Poland
| |
Collapse
|
27
|
Lai YL, Lin CY, Jiang WC, Ho YC, Chen CH, Yet SF. Loss of heme oxygenase-1 accelerates mesodermal gene expressions during embryoid body development from mouse embryonic stem cells. Redox Biol 2017; 15:51-61. [PMID: 29216542 PMCID: PMC5722471 DOI: 10.1016/j.redox.2017.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/06/2017] [Accepted: 11/20/2017] [Indexed: 02/04/2023] Open
Abstract
Heme oxygenase (HO)-1 is an inducible stress response protein and well known to protect cells and tissues against injury. Despite its important function in cytoprotection against physiological stress, the role of HO-1 in embryonic stem cell (ESC) differentiation remains largely unknown. We showed previously that induced pluripotent stem (iPS) cells that lack HO-1 are more sensitive to oxidant stress-induced cell death and more prone to lose pluripotent markers upon LIF withdrawal. To elucidate the role of HO-1 in ESC differentiation and to rule out the controversy of potential gene flaws in iPS cells, we derived and established mouse HO-1 knockout ESC lines from HO-1 knockout blastocysts. Using wild type D3 and HO-1 knockout ESCs in the 3-dimensional embryoid body (EB) differentiation model, we showed that at an early time point during EB development, an absence of HO-1 led to enhanced ROS level, concomitant with increased expressions of master mesodermal regulator brachyury and endodermal marker GATA6. In addition, critical smooth muscle cell (SMC) transcription factor serum response factor and its coactivator myocardin were enhanced. Furthermore, HO-1 deficiency increased Smad2 in ESCs and EBs, revealing a role of HO-1 in controlling Smad2 level. Smad2 not only mediates mesendoderm differentiation of mouse ESCs but also SMC development. Collectively, loss of HO-1 resulted in higher level of mesodermal and SMC regulators, leading to accelerated and enhanced SMC marker SM α-actin expression. Our results reveal a previously unrecognized function of HO-1 in regulating SMC gene expressions during ESC-EB development. More importantly, our findings may provide a novel strategy in enhancing ESC differentiation toward SMC lineage. Loss of HO-1 in ESCs promotes adipogenesis but reduces osteogenesis. During early EB development, loss of HO-1 results in robust induction of brachyury. During early EB development, lack of HO-1 leads to enhanced ROS level. Loss of HO-1 increases SMC transcription factor SRF and cofactor myocardin. HO-1 deficiency promotes mesodermal SMC differentiation during EB development.
Collapse
Affiliation(s)
- Yan-Liang Lai
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Chen-Yu Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Wei-Cheng Jiang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yen-Chun Ho
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chung-Huang Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University , Taichung, Taiwan.
| |
Collapse
|
28
|
Lack of Heme Oxygenase-1 Induces Inflammatory Reaction and Proliferation of Muscle Satellite Cells after Cardiotoxin-Induced Skeletal Muscle Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:491-506. [PMID: 29169990 DOI: 10.1016/j.ajpath.2017.10.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 12/29/2022]
Abstract
Heme oxygenase-1 (HO-1, Hmox1) regulates viability, proliferation, and differentiation of many cell types; hence, it may affect regeneration of injured skeletal muscle. Here, we injected cardiotoxin into gastrocnemius muscle of Hmox1+/+ and Hmox1-/- animals and analyzed cellular response after muscle injury, focusing on muscle satellite cells (SCs), inflammatory reaction, fibrosis, and formation of new blood vessels. HO-1 is strongly induced after muscle injury, being expressed mostly in the infiltrating leukocytes (CD45+ cells), including macrophages (F4/80+ cells). Lack of HO-1 augments skeletal muscle injury, evidenced by increased creatinine kinase and lactate dehydrogenase, as well as expression of monocyte chemoattractant protein-1, IL-6, IL-1β, and insulin-like growth factor-1. This, together with disturbed proportion of M1/M2 macrophages, accompanied by enhanced formation of arterioles, may be responsible for shift of Hmox1-/- myofiber size distribution toward larger one. Importantly, HO-1-deficient SCs are prone to activation and have higher proliferation on injury. This effect can be partially mimicked by stimulation of Hmox1+/+ SCs with monocyte chemoattractant protein-1, IL-6, IL-1β, and is associated with increased MyoD expression, suggesting that Hmox1-/- SCs are shifted toward more differentiated myogenic population. However, multiple rounds of degeneration/regeneration in conditions of HO-1 deficiency may lead to exhaustion of SC pool, and the number of SCs is decreased in old Hmox1-/- mice. In summary, HO-1 modulates muscle repair mechanisms preventing its uncontrolled acceleration.
Collapse
|
29
|
Chang LC, Hsieh MT, Yang JS, Lu CC, Tsai FJ, Tsao JW, Chiu YJ, Kuo SC, Lee KH. Effect of bis(hydroxymethyl) alkanoate curcuminoid derivative MTH-3 on cell cycle arrest, apoptotic and autophagic pathway in triple-negative breast adenocarcinoma MDA-MB-231 cells: An in vitro study. Int J Oncol 2017; 52:67-76. [PMID: 29138806 PMCID: PMC5743386 DOI: 10.3892/ijo.2017.4204] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/29/2017] [Indexed: 12/27/2022] Open
Abstract
Curcumin has been shown to exert potential antitumor activity in vitro and in vivo involved in multiple signaling pathways. However, the application of curcumin is still limited because of its poor hydrophilicity and low bio-availability. In the present study, we investigated the therapeutic effects of a novel and water soluble bis(hydroxymethyl) alkanoate curcuminoid derivative, MTH-3, on human breast adenocarcinoma MDA-MB-231 cells. This study investigated the effect of MTH-3 on cell viability, cell cycle and induction of autophagy and apoptosis in MDA-MB-231 cells. After 24-h treatment with MTH-3, a concentration-dependent decrease in MDA-MB-231 cell viability was observed, and the IC50 value was 5.37±1.22 μM. MTH-3 significantly triggered G2/M phase arrest and apoptosis in MDA-MB-231 cells. Within a 24-h treatment, MTH-3 decreased the CDK1 activity by decreasing CDK1 and cyclin B1 protein levels. MTH-3-induced apoptosis was further confirmed by morphological assessment and Annexin V/PI staining assay. Induction of apoptosis caused by MTH-3 was accompanied by an apparent increase of DR3, DR5 and FADD and, as well as a marked decrease of Bcl-2 and Bcl-xL protein expression. MTH-3 also decreased the protein levels of Ero1, PDI, PERK and calnexin, as well as increased the expression of IRE1α, CHOP and Bip that consequently led to ER stress and MDA-MB-231 cell apoptosis. In addition, MTH-3-treated cells were involved in the autophagic process and cleavage of LC3B was observed. MTH-3 enhanced the protein levels of LC3B, Atg5, Atg7, Atg12, p62 and Beclin-1 in MDA-MB-231 cells. Finally, DNA microarray was carried out to investigate the level changes of gene expression modulated by MTH-3 in MDA-MB-231 cells. Taken together, our results suggest that MTH-3 might be a novel therapeutic agent for the treatment of triple-negative breast cancer in the near future.
Collapse
Affiliation(s)
- Ling-Chu Chang
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 404, R.O.C
| | - Min-Tsang Hsieh
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 404, R.O.C
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, R.O.C
| | - Chi-Cheng Lu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan, R.O.C
| | - Fuu-Jen Tsai
- Human Genetic Center, China Medical University Hospital, Taichung 404, R.O.C
| | - Je-Wei Tsao
- School of Pharmacy, China Medical University, Taichung 404, R.O.C
| | - Yu-Jen Chiu
- Division of Reconstructive and Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Sheng-Chu Kuo
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 404, R.O.C
| | - Kuo-Hsiung Lee
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 404, R.O.C
| |
Collapse
|
30
|
Wu YH, Lin HR, Lee YH, Huang PH, Wei HC, Stern A, Chiu DTY. A novel fine tuning scheme of miR-200c in modulating lung cell redox homeostasis. Free Radic Res 2017; 51:591-603. [PMID: 28675952 DOI: 10.1080/10715762.2017.1339871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oxidative stress induces miR-200c, the predominant microRNA (miRNA) in lung tissues; however, the antioxidant role and biochemistry of such induction have not been clearly defined. Therefore, a lung adenocarcinoma cell line (A549) and a normal lung fibroblast (MRC-5) were used as models to determine the effects of miR-200c expression on lung antioxidant response. Hydrogen peroxide (H2O2) upregulated miR-200c, whose overexpression exacerbated the decrease in cell proliferation, retarded the progression of cells in the G2/M-phase, and increased oxidative stress upon H2O2 stimulation. The expression of three antioxidant proteins, superoxide dismutase (SOD)-2, haem oxygenase (HO)-1, and sirtuin (SIRT) 1, was reduced upon H2O2 stimulation in miR-200c-overexpressed A549 cells. This phenomenon of increased oxidative stress and antioxidant protein downregulation also occurs simultaneously in miR-200c overexpressed MRC-5 cells. Molecular analysis revealed that miR-200c inhibited the gene expression of HO-1 by directly targeting its 3'-untranslated region. The downregulation of SOD2 and SIRT1 by miR-200c was mediated through zinc finger E-box-binding homeobox 2 (ZEB2) and extracellular signal-regulated kinase 5 (ERK5) pathways, respectively, where knockdown of ZEB2 or ERK5 decreased the expression of SOD2 or SIRT1 in A549 cells. LNA anti-miR-200c transfection in A549 cells inhibited the endogenous miR-200c expression, resulting in increased expressions of antioxidant proteins, reduced oxidative stress and recovered cell proliferation upon H2O2 stimulation. These findings indicate that miR-200c fine-tuned the antioxidant response of the lung cells to oxidative stress through several pathways, and thus this study provides novel information concerning the role of miR-200c in modulating redox homeostasis of lung.
Collapse
Affiliation(s)
- Yi-Hsuan Wu
- a Research Center for Chinese Herbal Medicine, College of Human Ecology , Chang Gung University of Science and Technology , Taoyuan , Taiwan.,b Department of Medical Biotechnology and Laboratory Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan
| | - Hsin-Ru Lin
- b Department of Medical Biotechnology and Laboratory Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan.,c Molecular Medicine Research Center, Chang Gung University , Taoyuan , Taiwan
| | - Ying-Hsuan Lee
- b Department of Medical Biotechnology and Laboratory Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan
| | - Pin-Hao Huang
- b Department of Medical Biotechnology and Laboratory Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan
| | - Huei-Chung Wei
- b Department of Medical Biotechnology and Laboratory Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan
| | - Arnold Stern
- d New York University School of Medicine , New York , NY , USA
| | - Daniel Tsun-Yee Chiu
- a Research Center for Chinese Herbal Medicine, College of Human Ecology , Chang Gung University of Science and Technology , Taoyuan , Taiwan.,b Department of Medical Biotechnology and Laboratory Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan.,e Healthy Aging Research Center, Chang Gung University , Taoyuan , Taiwan.,f Department of Pediatric Hematology/Oncology , Linkou Chang Gung Memorial Hospital , Taoyuan , Taiwan
| |
Collapse
|
31
|
Bukowska-Strakova K, Ciesla M, Szade K, Nowak WN, Straka R, Szade A, Tyszka-Czochara M, Najder K, Konturek A, Siedlar M, Dulak J, Jozkowicz A. Reprint of: Heme oxygenase 1 affects granulopoiesis in mice through control of myelocyte proliferation. Immunobiology 2017; 222:846-857. [PMID: 28576353 DOI: 10.1016/j.imbio.2017.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/29/2022]
Abstract
Heme oxygenase-1 (HO-1) is stress-inducible, cytoprotective enzyme degrading heme to carbon monoxide (CO), biliverdin and Fe2+. We showed that HO-1 knock-out mice (HO-1-/-) have a twofold higher level of granulocytes than wild type (WT) mice, despite decreased concentration of granulocyte colony-stimulating factor (G-CSF) in the blood and reduced surface expression of G-CSF receptor on the hematopoietic precursors. This suggests the effect of HO-1 on granulopoiesis. Here we aimed to determine the stage of granulopoiesis regulated by HO-1. The earliest stages of hematopoiesis were not biased toward myeloid differentiation in HO-1-/- mice. Within committed granulocytic compartment, in WT mice, HO-1 was up-regulated starting from myelocyte stage. This was concomitant with up-regulation of miR-155, which targets Bach1, the HO-1 repressor. In HO-1-/- mice granulopoiesis was accelerated between myelocyte and metamyelocyte stage. There was a higher fraction of proliferating myelocytes, with increased nuclear expression of pro-proliferative C/EBPβ (CCAAT/enhancer binding protein beta) protein, especially its active LAP (liver-enriched activator proteins) isoform. Also our mathematical model confirmed shortening the myelocyte cyclic-time and prolonged mitotic expansion in absence of HO-1. It seems that changes in C/EBPβ expression and activity in HO-1-/- myelocytes can be associated with reduced level of its direct repressor miR-155 or with decreased concentration of CO, known to reduce nuclear translocation of C/EBPs. Mature HO-1-/- granulocytes were functionally competent as determined by oxidative burst capacity. In conclusion, HO-1 influences granulopoiesis through regulation of myelocyte proliferation. It is accompanied by changes in expression of transcriptionally active C/EBPβ protein. As HO-1 expression vary in human and is up-regulated in response to chemotherapy, it can potentially influence chemotherapy-induced neutropenia.
Collapse
Affiliation(s)
- Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Ciesla
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Witold Norbert Nowak
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Robert Straka
- AGH University of Science and Technology, Faculty of Metal Engineering and Industrial Computer Science, Department of Heat Engineering and Environment Protection, Poland
| | - Agata Szade
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Malgorzata Tyszka-Czochara
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Karolina Najder
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Konturek
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
32
|
Splenic Ly6C hi monocytes contribute to adverse late post-ischemic left ventricular remodeling in heme oxygenase-1 deficient mice. Basic Res Cardiol 2017; 112:39. [PMID: 28534119 PMCID: PMC5440541 DOI: 10.1007/s00395-017-0629-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022]
Abstract
Heme oxygenase-1 (Hmox1) is a stress-inducible protein crucial in heme catabolism. The end products of its enzymatic activity possess anti-oxidative, anti-apoptotic and anti-inflammatory properties. Cardioprotective effects of Hmox1 were demonstrated in experimental models of myocardial infarction (MI). Nevertheless, its importance in timely resolution of post-ischemic inflammation remains incompletely understood. The aim of this study was to determine the role of Hmox1 in the monocyte/macrophage-mediated cardiac remodeling in a mouse model of MI. Hmox1 knockout (Hmox1-/-) and wild-type (WT, Hmox1+/+) mice were subjected to a permanent ligation of the left anterior descending coronary artery. Significantly lower incidence of left ventricle (LV) free wall rupture was noted between 3rd and 5th day after MI in Hmox1-/- mice resulting in their better overall survival. Then, starting from 7th until 21st day post-MI a more potent deterioration of LV function was observed in Hmox1-/- than in the surviving Hmox1+/+ mice. This was accompanied by higher numbers of Ly6Chi monocytes in peripheral blood, as well as higher expression of monocyte chemoattractant protein-1 and adhesion molecules in the hearts of MI-operated Hmox1-/- mice. Consequently, a greater post-MI monocyte-derived myocardial macrophage infiltration was noted in Hmox1-deficient individuals. Splenectomy decreased the numbers of circulating inflammatory Ly6Chi monocytes in blood, reduced the numbers of proinflammatory cardiac macrophages and significantly improved the post-MI LV function in Hmox1-/- mice. In conclusion, Hmox1 deficiency has divergent consequences in MI. On the one hand, it improves early post-MI survival by decreasing the occurrence of cardiac rupture. Afterwards, however, the hearts of Hmox1-deficient mice undergo adverse late LV remodeling due to overactive and prolonged post-ischemic inflammatory response. We identified spleen as an important source of these cardiovascular complications in Hmox1-/- mice.
Collapse
|
33
|
Otterbein LE, Foresti R, Motterlini R. Heme Oxygenase-1 and Carbon Monoxide in the Heart: The Balancing Act Between Danger Signaling and Pro-Survival. Circ Res 2017; 118:1940-1959. [PMID: 27283533 DOI: 10.1161/circresaha.116.306588] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Understanding the processes governing the ability of the heart to repair and regenerate after injury is crucial for developing translational medical solutions. New avenues of exploration include cardiac cell therapy and cellular reprogramming targeting cell death and regeneration. An attractive possibility is the exploitation of cytoprotective genes that exist solely for self-preservation processes and serve to promote and support cell survival. Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. The remarkable cardioprotective effects ascribed to heme oxygenase-1 are best evidenced by its ability to regulate inflammatory processes, cellular signaling, and mitochondrial function ultimately mitigating myocardial tissue injury and the progression of vascular-proliferative disease. We discuss here new insights into the role of heme oxygenase-1 and heme on cardiovascular health, and importantly, how they might be leveraged to promote heart repair after injury.
Collapse
Affiliation(s)
- Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Roberta Foresti
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| | - Roberto Motterlini
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| |
Collapse
|
34
|
Espinoza JA, González PA, Kalergis AM. Modulation of Antiviral Immunity by Heme Oxygenase-1. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:487-493. [PMID: 28082120 DOI: 10.1016/j.ajpath.2016.11.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/05/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022]
Abstract
Heme oxygenase-1 (HO-1) is a stress-inducible, anti-inflammatory, and cytoprotective enzyme expressed in most cell types in the organism. Under several stress stimuli, HO-1 expression and activity is up-regulated to catalyze the rate-limiting enzymatic step of heme degradation into carbon monoxide, free iron, and biliverdin. Besides its effects on cell metabolism, HO-1 is also capable of modulating host innate and adaptive immune responses in response to sepsis, transplantation, and autoimmunity, and preventing oxidative damage associated with inflammation. In addition, recent studies have reported that HO-1 can exert a significant antiviral activity against a wide variety of viruses, including HIV, hepatitis C virus, hepatitis B virus, enterovirus 71, influenza virus, respiratory syncytial virus, dengue virus, and Ebola virus, among others. Herein, we address the current understanding of the functional significance of HO-1 against a variety of viruses and its potential as a therapeutic strategy to prevent and control viral infections. Furthermore, we review the most important features of the immunoregulatory functions for this enzyme.
Collapse
Affiliation(s)
- Janyra A Espinoza
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM, Combined Research Unit 1064, Nantes University Hospital Nantes, Institute for Transplantation-Urology-Nephrology, Université de Nantes, Faculty of Medicine, Nantes, France; Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
35
|
Jez M, Ciesla M, Stepniewski J, Langrzyk A, Muchova L, Vitek L, Jozkowicz A, Dulak J. Valproic acid downregulates heme oxygenase-1 independently of Nrf2 by increasing ubiquitination and proteasomal degradation. Biochem Biophys Res Commun 2017; 485:160-166. [PMID: 28189672 DOI: 10.1016/j.bbrc.2017.02.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 01/01/2023]
Abstract
AIMS Heme oxygenase-1 (HO-1; HMOX1 in human, Hmox1 in mice) is an antioxidative enzyme affecting wide range of sub-cellular processes. It was shown to modulate tumor growth or vascular-related diseases, thus being putative molecular target for tailored therapies. Therefore it is of importance to elucidate novel compounds regulating HO-1 activity/expression and to delineate mechanisms of their action. In the present study we aimed to understand mode of action of valproic acid (VA), an antiepileptic drug, on HO-1 expression. RESULTS We demonstrated that HO-1 expression is decreased by VA at protein but not mRNA level in human alveolar rhabdomyosarcoma cell line CW9019. Nrf2 transcription factor, the activator of HO-1 expression through ARE sequence, was excluded as a mediator of HO-1 decrease, as VA downregulated Bach1, a Nrf2 repressor, concomitantly upregulating ARE activation. Also miRNA-dependent inhibition was excluded as a mechanism of HMOX1 regulation. However, co-immunoprecipitation assay showed a higher level of ubiquitinated HO-1 after VA treatment. Accordingly, MG132, an inhibitor of proteasomal degradation, reversed the effect of VA on HO-1 suggesting that decrease in HO-1 expression by VA is through protein stability. The inhibitory effect of VA on HO-1 was also observed in murine cells including embryonic fibroblasts isolated from Nrf2-deficient mice, what confirms Nrf2-independent effect of the compound. Importantly, VA decreased also HO-1 expression and activity in murine skeletal muscles in vivo. CONCLUSION Our data indicate that VA downregulates HO-1 by acting through ubiquitin-proteasomal pathway leading to decrease in protein level.
Collapse
Affiliation(s)
- Mateusz Jez
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Ciesla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stepniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | | - Lucie Muchova
- Fourth Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Libor Vitek
- Fourth Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Kardio-Med Silesia, M. Curie-Skłodowskiej 10c, Zabrze, Poland.
| |
Collapse
|
36
|
Bolisetty S, Zarjou A, Agarwal A. Heme Oxygenase 1 as a Therapeutic Target in Acute Kidney Injury. Am J Kidney Dis 2017; 69:531-545. [PMID: 28139396 DOI: 10.1053/j.ajkd.2016.10.037] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/22/2016] [Indexed: 01/06/2023]
Abstract
A common clinical condition, acute kidney injury (AKI) significantly influences morbidity and mortality, particularly in critically ill patients. The pathophysiology of AKI is complex and involves multiple pathways, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Recent evidence suggests that a single insult to the kidney significantly enhances the propensity to develop chronic kidney disease. Therefore, the generation of effective therapies against AKI is timely. In this context, the cytoprotective effects of heme oxygenase 1 (HO-1) in animal models of AKI are well documented. HO-1 modulates oxidative stress, autophagy, and inflammation and regulates the progression of cell cycle via direct and indirect mechanisms. These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). This review highlights recent advances in the molecular mechanisms of HO-1-mediated cytoprotection and discusses the translational potential of HO-1 induction in AKI.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL; Birmingham Veterans Administration Medical Center, Birmingham, AL.
| |
Collapse
|
37
|
Bukowska-Strakova K, Ciesla M, Szade K, Nowak WN, Straka R, Szade A, Tyszka-Czochara M, Najder K, Konturek A, Siedlar M, Dulak J, Jozkowicz A. Heme oxygenase 1 affects granulopoiesis in mice through control of myelocyte proliferation. Immunobiology 2016; 222:506-517. [PMID: 27817989 DOI: 10.1016/j.imbio.2016.10.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 12/12/2022]
Abstract
Heme oxygenase-1 (HO-1) is stress-inducible, cytoprotective enzyme degrading heme to carbon monoxide (CO), biliverdin and Fe2+. We showed that HO-1 knock-out mice (HO-1-/-) have a twofold higher level of granulocytes than wild type (WT) mice, despite decreased concentration of granulocyte colony-stimulating factor (G-CSF) in the blood and reduced surface expression of G-CSF receptor on the hematopoietic precursors. This suggests the effect of HO-1 on granulopoiesis. Here we aimed to determine the stage of granulopoiesis regulated by HO-1. The earliest stages of hematopoiesis were not biased toward myeloid differentiation in HO-1-/- mice. Within committed granulocytic compartment, in WT mice, HO-1 was up-regulated starting from myelocyte stage. This was concomitant with up-regulation of miR-155, which targets Bach1, the HO-1 repressor. In HO-1-/- mice granulopoiesis was accelerated between myelocyte and metamyelocyte stage. There was a higher fraction of proliferating myelocytes, with increased nuclear expression of pro-proliferative C/EBPβ (CCAAT/enhancer binding protein beta) protein, especially its active LAP (liver-enriched activator proteins) isoform. Also our mathematical model confirmed shortening the myelocyte cyclic-time and prolonged mitotic expansion in absence of HO-1. It seems that changes in C/EBPβ expression and activity in HO-1-/- myelocytes can be associated with reduced level of its direct repressor miR-155 or with decreased concentration of CO, known to reduce nuclear translocation of C/EBPs. Mature HO-1-/- granulocytes were functionally competent as determined by oxidative burst capacity. In conclusion, HO-1 influences granulopoiesis through regulation of myelocyte proliferation. It is accompanied by changes in expression of transcriptionally active C/EBPβ protein. As HO-1 expression vary in human and is up-regulated in response to chemotherapy, it can potentially influence chemotherapy-induced neutropenia.
Collapse
Affiliation(s)
- Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Ciesla
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Witold Norbert Nowak
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Robert Straka
- AGH University of Science and Technology, Faculty of Metal Engineering and Industrial Computer Science, Department of Heat Engineering and Environment Protection, Poland
| | - Agata Szade
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Malgorzata Tyszka-Czochara
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Karolina Najder
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Konturek
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty Of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
38
|
Zou C, Zou C, Cheng W, Li Q, Han Z, Wang X, Jin J, Zou J, Liu Z, Zhou Z, Zhao W, Du Z. Heme oxygenase-1 retards hepatocellular carcinoma progression through the microRNA pathway. Oncol Rep 2016; 36:2715-2722. [PMID: 27571925 DOI: 10.3892/or.2016.5056] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/25/2016] [Indexed: 11/06/2022] Open
Abstract
Heme metabolism system is involved in microRNA (miRNA) biogenesis. The complicated interplay between heme oxygenase-1 (HO-1) and miRNA has been observed in various tissues and diseases, including human malignancy. In the present study, our data showed that stable HO-1 overexpression in hepatocellular carcinoma (HCC) cells downregulated several oncomiRs. The most stably downregulated are miR-30d and miR-107. Iron, one of HO-1 catalytic products, was an important mediator in this regulation. Cell function analysis demonstrated that HO-1 inhibited the proliferation and metastasis of HepG2 cells, whereas miR-30d/miR-107 improved the proliferative and migratory ability of HepG2 cells. The beneficial effect of HO-1 in HCC inhibition could be reversed by upregulating miR-30d and miR-107. Akt and ERK pathways may be involved in the regulation of HO-1/miR-30d/miR-107 in HCC. These data indicate that HO-1 significantly suppresses HCC progression by regulating the miR-30d/miR-107 level, suggesting miR-30d/miR-107 regulation as a new molecular mechanism of HO-1 anticancer effect.
Collapse
Affiliation(s)
- Chaoxia Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Chendan Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Wanpeng Cheng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Qiang Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhongjing Han
- Department of Hemopathology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang, P.R. China
| | - Xiaona Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Jianfeng Jin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Jiaqi Zou
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhiyan Liu
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhongqiu Zhou
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Weiming Zhao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhimin Du
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
39
|
Casas E, Cai G, Kuehn LA, Register KB, McDaneld TG, Neill JD. Association of MicroRNAs with Antibody Response to Mycoplasma bovis in Beef Cattle. PLoS One 2016; 11:e0161651. [PMID: 27537842 PMCID: PMC4990326 DOI: 10.1371/journal.pone.0161651] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/09/2016] [Indexed: 12/16/2022] Open
Abstract
The objective of this study was to identify microRNAs associated with a serum antibody response to Mycoplasma bovis in beef cattle. Serum from sixteen beef calves was collected at three points: in summer after calves were born, in fall at weaning, and in the following spring. All sera collected in the summer were ELISA-negative for anti-M. bovis. By the fall, eight animals were seropositive for IgG (positive group), while eight remained negative (negative group). By spring, all animals in both groups were seropositive. MicroRNAs were extracted from sera and sequenced on the Illumina HiSeq next-generation sequencer. A total of 1,374,697 sequences mapped to microRNAs in the bovine genome. Of these, 82% of the sequences corresponded to 27 microRNAs, each represented by a minimum of 10,000 sequences. There was a statistically significant interaction between ELISA response and season for bta-miR-24-3p (P = 0.0268). All sera collected at the initial summer had a similar number of copies of this microRNA (P = 0.773). In the fall, the positive group had an increased number of copies when compared to the negative group (P = 0.021), and this grew more significant by the following spring (P = 0.0001). There were 21 microRNAs associated (P< 0.05) with season. These microRNAs could be evaluated further as candidates to potentially improve productivity in cattle. The microRNAs bta-let-7b, bta-miR- 24-3p, bta-miR- 92a, and bta-miR-423-5p, were significatly associated with ELISA status (P< 0.05). These microRNAs have been recognized as playing a role in the host defense against bacteria in humans, mice, and dairy cattle. Further studies are needed to establish if these microRNAs could be used as diagnostic marker or indicator of exposure, or whether intervention strategies could be developed as an alternative to antibiotics for controlling disease due to M. bovis.
Collapse
Affiliation(s)
- Eduardo Casas
- USDA, ARS, National Animal Disease Center, Ames, IA 50010, United States of America
- * E-mail:
| | - Guohong Cai
- USDA, ARS, National Animal Disease Center, Ames, IA 50010, United States of America
| | - Larry A. Kuehn
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, United States of America
| | - Karen B. Register
- USDA, ARS, National Animal Disease Center, Ames, IA 50010, United States of America
| | - Tara G. McDaneld
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, United States of America
| | - John D. Neill
- USDA, ARS, National Animal Disease Center, Ames, IA 50010, United States of America
| |
Collapse
|
40
|
Lee D, Kook SH, Ji H, Lee SA, Choi KC, Lee KY, Lee JC. N-acetyl cysteine inhibits H2O2-mediated reduction in the mineralization of MC3T3-E1 cells by down-regulating Nrf2/HO-1 pathway. BMB Rep 2016; 48:636-41. [PMID: 26303969 PMCID: PMC4911206 DOI: 10.5483/bmbrep.2015.48.11.112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Indexed: 12/16/2022] Open
Abstract
There are controversial findings regarding the roles of nuclear factor
(erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway on bone
metabolism under oxidative stress. We investigated how Nrf2/HO-1 pathway affects
osteoblast differentiation of MC3T3-E1 cells in response to hydrogen peroxide
(H2O2), N-acetyl cysteine (NAC), or
both. Exposing the cells to H2O2 decreased the alkaline
phosphatase activity, calcium accumulation, and expression of osteoblast
markers, such as osteocalcin and runt-related transcription factor-2. In
contrast, H2O2 treatment increased the expression of Nrf2
and HO-1 in the cells. Treatment with hemin, a chemical HO-1 inducer, mimicked
the inhibitory effect of H2O2 on osteoblast
differentiation by increasing the HO-1 expression and decreasing the osteogenic
marker genes. Pretreatment with NAC restored all changes induced by
H2O2 to near normal levels in the cells. Collectively,
our findings suggest that H2O2-mediated activation of
Nrf2/HO-1 pathway negatively regulates the osteoblast differentiation, which is
inhibited by NAC. [BMB Reports 2015; 48(11): 636-641]
Collapse
Affiliation(s)
- Daewoo Lee
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju 54896, Korea
| | - Sung-Ho Kook
- Institute of Oral Biosciences and School of Dentistry; Department of Bioactive Material Sciences and Institute of Molecular Biology and Genetics, Chonbuk National University, Jeonju 54896, Korea
| | - Hyeok Ji
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju 54896, Korea
| | - Seung-Ah Lee
- Department of Nursing, Chonnam Techno College, Chonnam 57500, Korea
| | - Ki-Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, RDA, Cheonan 31002, Korea
| | - Kyung-Yeol Lee
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju 54896, Korea
| | - Jeong-Chae Lee
- Institute of Oral Biosciences and School of Dentistry; Department of Bioactive Material Sciences and Institute of Molecular Biology and Genetics, Chonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
41
|
Almeida AS, Soares NL, Vieira M, Gramsbergen JB, Vieira HLA. Carbon Monoxide Releasing Molecule-A1 (CORM-A1) Improves Neurogenesis: Increase of Neuronal Differentiation Yield by Preventing Cell Death. PLoS One 2016; 11:e0154781. [PMID: 27144388 PMCID: PMC4856303 DOI: 10.1371/journal.pone.0154781] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 04/19/2016] [Indexed: 11/19/2022] Open
Abstract
Cerebral ischemia and neurodegenerative diseases lead to impairment or death of neurons in the central nervous system. Stem cell based therapies are promising strategies currently under investigation. Carbon monoxide (CO) is an endogenous product of heme degradation by heme oxygenase (HO) activity. Administration of CO at low concentrations produces several beneficial effects in distinct tissues, namely anti-apoptotic and anti-inflammatory. Herein the CO role on modulation of neuronal differentiation was assessed. Three different models with increasing complexity were used: human neuroblastoma SH-S5Y5 cell line, human teratocarcinoma NT2 cell line and organotypic hippocampal slice cultures (OHSC). Cell lines were differentiated into post-mitotic neurons by treatment with retinoic acid (RA) supplemented with CO-releasing molecule A1 (CORM-A1). CORM-A1 positively modulated neuronal differentiation, since it increased final neuronal production and enhanced the expression of specific neuronal genes: Nestin, Tuj1 and MAP2. Furthermore, during neuronal differentiation process, there was an increase in proliferative cell number (ki67 mRNA expressing cells) and a decrease in cell death (lower propidium iodide (PI) uptake, limitation of caspase-3 activation and higher Bcl-2 expressing cells). CO supplementation did not increase the expression of RA receptors. In the case of SH-S5Y5 model, small amounts of reactive oxygen species (ROS) generation emerges as important signaling molecules during CO-promoted neuronal differentiation. CO's improvement of neuronal differentiation yield was validated using OHSC as ex vivo model. CORM-A1 treatment of OHSC promoted higher levels of cells expressing the neuronal marker Tuj1. Still, CORM-A1 increased cell proliferation assessed by ki67 expression and also prevented cell death, which was followed by increased Bcl-2 expression, decreased levels of active caspase-3 and PI uptake. Likewise, ROS signaling emerged as key factors in CO's increasing number of differentiated neurons in OHSC. In conclusion, CO's increasing number of differentiated neurons is a novel biological role disclosed herein. CO improves neuronal yield due to its capacity to reduce cell death, promoting an increase in proliferative population. However, one cannot disregard a direct CO's effect on specific cellular processes of neuronal differentiation. Further studies are needed to evaluate how CO can potentially modulate cell mechanisms involved in neuronal differentiation. In summary, CO appears as a promising therapeutic molecule to stimulate endogenous neurogenesis or to improve in vitro neuronal production for cell therapy strategies.
Collapse
Affiliation(s)
- Ana S. Almeida
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
- Instituto de Tecnologia Química e Biológica (ITQB), Universidade Nova de Lisboa, Apartado 127, 2781-901 Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal
| | - Nuno L. Soares
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - Melissa Vieira
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - Jan Bert Gramsbergen
- Institute of Molecular Medicine, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Helena L. A. Vieira
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal
| |
Collapse
|
42
|
Ryter SW, Choi AMK. Targeting heme oxygenase-1 and carbon monoxide for therapeutic modulation of inflammation. Transl Res 2016; 167:7-34. [PMID: 26166253 PMCID: PMC4857893 DOI: 10.1016/j.trsl.2015.06.011] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 12/19/2022]
Abstract
The heme oxygenase-1 (HO-1) enzyme system remains an attractive therapeutic target for the treatment of inflammatory conditions. HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXα (BV), the latter which is converted to bilirubin-IXα (BR). HO-1 may function as a pleiotropic regulator of inflammatory signaling programs through the generation of its biologically active end products, namely CO, BV and BR. CO, when applied exogenously, can affect apoptotic, proliferative, and inflammatory cellular programs. Specifically, CO can modulate the production of proinflammatory or anti-inflammatory cytokines and mediators. HO-1 and CO may also have immunomodulatory effects with respect to regulating the functions of antigen-presenting cells, dendritic cells, and regulatory T cells. Therapeutic strategies to modulate HO-1 in disease include the application of natural-inducing compounds and gene therapy approaches for the targeted genetic overexpression or knockdown of HO-1. Several compounds have been used therapeutically to inhibit HO activity, including competitive inhibitors of the metalloporphyrin series or noncompetitive isoform-selective derivatives of imidazole-dioxolanes. The end products of HO activity, CO, BV and BR may be used therapeutically as pharmacologic treatments. CO may be applied by inhalation or through the use of CO-releasing molecules. This review will discuss HO-1 as a therapeutic target in diseases involving inflammation, including lung and vascular injury, sepsis, ischemia-reperfusion injury, and transplant rejection.
Collapse
Affiliation(s)
- Stefan W Ryter
- Joan and Sanford I. Weill Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY.
| | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY
| |
Collapse
|
43
|
Kim M, Kim Y, Lee S, Kuk M, Kim AY, Kim W, Kweon OK. Comparison of viability and antioxidant capacity between canine adipose-derived mesenchymal stem cells and heme oxygenase-1-overexpressed cells after freeze-thawing. J Vet Med Sci 2015; 78:619-25. [PMID: 26725542 PMCID: PMC4873853 DOI: 10.1292/jvms.15-0361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Allogenic adipose-derived mesenchymal stem cells (Ad-MSCs) are an alternative source for
cytotherapy owing to their antioxidant and anti-inflammatory effects. Frozen-thawed
allogenic Ad-MSCs can be used instantly for this purpose. However, the viability and
function of frozen-thawed Ad-MSCs have not been clearly evaluated. The purpose of this
study was to compare the viability and function of Ad-MSCs and heme oxygenase-1
(HO-1)-overexpressed Ad-MSCs in vitro after freeze-thawing. The
viability, proliferation, antioxidant capacity and mRNA gene expression of growth factors
were evaluated. Frozen-thawed cells showed significantly lower viability than fresh cells
(77% for Ad-MSCs and 71% for HO-1 Ad-MSCs, P<0.01). However, the
proliferation rate of frozen-thawed Ad-MSCs increased and did not differ from that of
fresh Ad-MSCs after 3 days of culture. In contrast, the proliferation rate of
HO-1-overexpressed Ad-MSCs was lower than that of Ad-MSCs. The mRNA expression levels of
TGF-β, HGF and VEGF did not differ
between fresh and frozen-thawed Ad-MSCs, but COX-2 and
IL-6 had significantly higher mRNA expression in frozen cells than
fresh cells (P<0.05). Fresh Ad-MSCs exhibited higher
HO-1 mRNA expression than frozen-thawed Ad-MSCs, and fresh
HO-1-overexpressed Ad-MSCs exhibited higher than fresh Ad-MSCs
(P<0.05). However, there was no significant difference between fresh
and frozen HO-1-overexpressed Ad-MSCs. The antioxidant capacity of HO-1-overexpressed
Ad-MSCs was significantly higher than that of Ad-MSCs. Cryopreservation of Ad-MSCs
negatively affects viability and antioxidant capacity, and HO-1-overexpressed Ad-MSCs
might be useful to maximize the effect of Ad-MSCs for cytotherapy.
Collapse
Affiliation(s)
- Mijung Kim
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, 151-742, Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Loboda A, Jozkowicz A, Dulak J. HO-1/CO system in tumor growth, angiogenesis and metabolism - Targeting HO-1 as an anti-tumor therapy. Vascul Pharmacol 2015; 74:11-22. [PMID: 26392237 DOI: 10.1016/j.vph.2015.09.004] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/02/2015] [Accepted: 09/16/2015] [Indexed: 02/08/2023]
Abstract
Heme oxygenase-1 (HO-1, hmox-1) catalyzes the rate-limiting step in the heme degradation processes. Out of three by-products of HO-1 activity, biliverdin, iron ions and carbon monoxide (CO), the latter was mostly shown to mediate many beneficial HO-1 effects, including protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. Mounting evidence suggests that HO-1/CO systemmay be of special benefit in protection inmany pathological conditions, like atherosclerosis or myocardial infarction. By contrast, the augmented expression of HO-1 in tumor tissues may have detrimental effect as HO-1 accelerates the formation of tumor neovasculature and provides the selective advantage for tumor cells to overcome the increased oxidative stress during tumorigenesis and during treatment. The inhibition of HO-1 has been proposed as an anti-cancer therapy, however, because of non-specific effects of known HO-1 inhibitors, the discovery of ideal drug lowering HO-1 expression/activity is still an open question. Importantly, in several types of cancer HO-1/CO system exerts opposite activities, making the possible treatment more complicated. All together indicates the complex role for HO-1/CO in various in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| |
Collapse
|
45
|
Kook SH, Kim KA, Ji H, Lee D, Lee JC. Irradiation inhibits the maturation and mineralization of osteoblasts via the activation of Nrf2/HO-1 pathway. Mol Cell Biochem 2015; 410:255-66. [PMID: 26346162 DOI: 10.1007/s11010-015-2559-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/03/2015] [Indexed: 01/18/2023]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) regulates the induction of antioxidant gene expression and protects cells against oxidative injury. However, there are controversial findings regarding the roles of Nrf2 on bone metabolism under oxidative stress. The role of Nrf2 on the differentiation of radiation-exposed osteoblasts is also unclear. We investigated whether Nrf2 negatively or positively affects osteoblast differentiation in response to irradiation. Irradiation inhibited osteoblast differentiation of MC3T3-E1 cells in a dose-dependent manner. This inhibition was evidenced by the irradiation-mediated decreases in bone-like nodule formation, alkaline phosphatase (ALP) activity, calcium accumulation, and expression of osteoblast markers, such as ALP, osteocalcin, osteopontin, bone sialoprotein, osterix, and Runx2. These reductions were accompanied by increased induction of Nrf2 and heme oxygenase-1 (HO-1), accumulation of cellular oxidants, and depletion of antioxidant defense enzymes. siRNA-mediated silencing of Nrf2 markedly reversed the negative effect of irradiation on osteoblast differentiation of the cells, leading to a decrease in HO-1 and an increase in Runx2 levels. Irradiation-mediated decreases in the levels of Runx2 and osteocalcin mRNA, but not of Nrf2 protein, were also significantly inhibited by HO-1 inhibitor, zinc protoporphyrin IX. Furthermore, N-acetyl cysteine restored all of the changes induced by irradiation to near-normal levels in the cells. These results demonstrate that irradiation inhibits osteoblast differentiation and mineralization of MC3T3-E1 cells through the oxidative stress-mediated activation of Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Sung-Ho Kook
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea
- Research Center of Bioactive Materials and Institute of Molecular Biology and Genetics, Chonbuk National University, Jeonju, 561-756, South Korea
| | - Kyoung-A Kim
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea
- Department of Oral and Maxillofacial Radiology and Research Institute of Clinical Medicine, Chonbuk National University, Jeonju, 561-756, South Korea
| | - Hyeok Ji
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea
| | - Daewoo Lee
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea
| | - Jeong-Chae Lee
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea.
- Research Center of Bioactive Materials and Institute of Molecular Biology and Genetics, Chonbuk National University, Jeonju, 561-756, South Korea.
- Department of Orthodontics and Institute of Oral Biosciences, Research Center of Bioactive Materials, Chonbuk National University, Jeonju, 561-756, South Korea.
| |
Collapse
|
46
|
MicroRNA miR-24-3p promotes porcine reproductive and respiratory syndrome virus replication through suppression of heme oxygenase-1 expression. J Virol 2015; 89:4494-503. [PMID: 25653454 DOI: 10.1128/jvi.02810-14] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most economically important viruses affecting the swine industry worldwide. Our previous research showed that PRRSV downregulates the expression of heme oxygenase-1 (HO-1), a pivotal cytoprotective enzyme, postinfection and that overexpression of HO-1 inhibits PRRSV replication. MicroRNAs regulate gene expression at the posttranscriptional level and have recently been demonstrated to play vital roles in pathogen-host interactions. The present study sought to determine whether microRNAs modulate HO-1 expression and, by doing so, regulate PRRSV replication. Using bioinformatic prediction and experimental verification, we demonstrate that HO-1 expression is regulated by miR-24-3p. A direct interaction between miR-24-3p and HO-1 mRNA was confirmed using a number of approaches. Overexpression of miR-24-3p significantly decreased HO-1 mRNA and protein levels. PRRSV infection induced miR-24-3p expression to facilitate viral replication. The suppressive effect of HO-1 induction by protoporphyrin IX cobalt chloride (CoPP; a classical inducer of HO-1 expression) on PRRSV replication in MARC-145 cells and primary porcine alveolar macrophages could also be reversed by overexpression of miR-24-3p. Collectively, these results suggested that miR-24-3p promotes PRRSV replication through suppression of HO-1 expression, which not only provides new insights into virus-host interactions during PRRSV infection but also suggests potential new antiviral strategies against PRRSV infection. IMPORTANCE MicroRNAs (miRNAs) play vital roles in viral infections by regulating the expression of viral or host genes at the posttranscriptional level. Heme oxygenase-1 (HO-1), a pivotal cytoprotective enzyme, has antiviral activity for a number of viruses, such as Ebola virus, hepatitis C virus, human immunodeficiency virus, and our focus, PRRSV, which causes great economic losses each year in the swine industry worldwide. Here, we show that PRRSV infection induces host miRNA miR-24-3p expression and that miR-24-3p regulates HO-1 expression through both mRNA degradation and translation repression. Suppression of HO-1 expression by miR-24-3p facilitates PRRSV replication. This work lends credibility to the hypothesis that an arterivirus can manipulate cellular miRNAs to enhance virus replication by regulating antiviral responses following viral infection. Therefore, our findings provide new insights into the pathogenesis of PRRSV.
Collapse
|
47
|
Dulak J, Jozkowicz A. Novel faces of heme oxygenase-1: mechanisms and therapeutic potentials. Antioxid Redox Signal 2014; 20:1673-6. [PMID: 24341409 PMCID: PMC3961789 DOI: 10.1089/ars.2013.5761] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Not only double, Janus face, but numerous appearances characterize heme oxygenase-1 (HO-1), an inducible enzyme which main role is to degrade heme. Recently, the noncanonical functions of HO-1 have particularly attracted researchers' attention. Indeed, understanding the enzyme-independent activities of HO-1 can provide additional chances for translational application of research on HO-1. In this Forum, eight reviews and two original articles describe a plethora of mechanisms in which this pleiotropic protein is involved. Further understanding of HO-1 functions is of particular significance for elucidating the pathology of various human diseases and providing rationale for novel therapies.
Collapse
Affiliation(s)
- Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University , Krakow, Poland
| | | |
Collapse
|