1
|
Fu ZL, Yang Y, Ma L, Malmuthuge N, Guan LL, Bu DP. Dynamics of oxidative stress and immune responses in neonatal calves during diarrhea. J Dairy Sci 2024; 107:1286-1298. [PMID: 37776998 DOI: 10.3168/jds.2023-23630] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/08/2023] [Indexed: 10/02/2023]
Abstract
Oxidative stress is the imbalanced redox status between oxidant production and their scavengers leading to intestinal physiological dysfunction. However, the role of systemic and local oxidative status during neonatal calf diarrhea is not known. This study assessed systemic (serum) and local (fecal) oxidative status when calves either naturally developed diarrhea or naturally recovered. Healthy calves were enrolled in the study at d 18 of age, and their health status was monitored from the enrollment. Based on their enteric health status on d 21 and 28, calves were grouped as continuous diarrhea from d 21 to 28 (n = 14), diarrhea at d 21 but recovered at d 28 (DH group, n = 19), healthy at d 21 but developed diarrhea at d 28 (HD group, n = 15), and healthy throughout the study (HH group, n = 16). Serum and fecal samples were collected at d 21 and 28 from all calves in the morning 2 h after feeding. Dynamics of oxidative stress indicators including reactive oxygen species (ROS), malondialdehyde (MDA), H2O2, 8-hydroxy-2'-deoxyguanosine (8-OHDG), glutathione peroxidase, superoxide dismutase, catalase (CAT), and total antioxidant capacity and inflammatory indicators TNF-α, IL-1β, IL-4, IL-6, IL-10, and IFN-γ were evaluated using serum samples. In addition, fecal oxidative stress indicators ROS and MDA were measured. Serum ROS, MDA, 8-OHDG, as well as fecal ROS and MDA, were higher, whereas serum CAT and H2O2 were lower in diarrheic calves than those of healthy calves. Serum ROS, MDA, and 8OHDG and fecal ROS and MDA increased in the HD group from d 21 to 28 as they developed diarrhea. In contrast, all these oxidative stress markers decreased in the DH group from d 21 to 28 as they recovered. However, serum H2O2 had an opposite changing trend, which became lower in the HD group and higher in the DH group at d 28. In conclusion, both systemic and local oxidative stress markers and cytokine profiles altered as calves moved from being healthy to having diarrhea or vice versa. Serum ROS, MDA, and 8-OHDG can be used to develop biomarkers to screen calves prone to enteric infections during the preweaning period.
Collapse
Affiliation(s)
- Z L Fu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Y Yang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China; School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, A94 R704, Ireland
| | - L Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - N Malmuthuge
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, T1J 4B1, Canada
| | - L L Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada; Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4 Canada.
| | - D P Bu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
2
|
Huang Z. Evidence that Alzheimer's Disease Is a Disease of Competitive Synaptic Plasticity Gone Awry. J Alzheimers Dis 2024; 99:447-470. [PMID: 38669548 PMCID: PMC11119021 DOI: 10.3233/jad-240042] [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] [Indexed: 04/28/2024]
Abstract
Mounting evidence indicates that a physiological function of amyloid-β (Aβ) is to mediate neural activity-dependent homeostatic and competitive synaptic plasticity in the brain. I have previously summarized the lines of evidence supporting this hypothesis and highlighted the similarities between Aβ and anti-microbial peptides in mediating cell/synapse competition. In cell competition, anti-microbial peptides deploy a multitude of mechanisms to ensure both self-protection and competitor elimination. Here I review recent studies showing that similar mechanisms are at play in Aβ-mediated synapse competition and perturbations in these mechanisms underpin Alzheimer's disease (AD). Specifically, I discuss evidence that Aβ and ApoE, two crucial players in AD, co-operate in the regulation of synapse competition. Glial ApoE promotes self-protection by increasing the production of trophic monomeric Aβ and inhibiting its assembly into toxic oligomers. Conversely, Aβ oligomers, once assembled, promote the elimination of competitor synapses via direct toxic activity and amplification of "eat-me" signals promoting the elimination of weak synapses. I further summarize evidence that neuronal ApoE may be part of a gene regulatory network that normally promotes competitive plasticity, explaining the selective vulnerability of ApoE expressing neurons in AD brains. Lastly, I discuss evidence that sleep may be key to Aβ-orchestrated plasticity, in which sleep is not only induced by Aβ but is also required for Aβ-mediated plasticity, underlining the link between sleep and AD. Together, these results strongly argue that AD is a disease of competitive synaptic plasticity gone awry, a novel perspective that may promote AD research.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
3
|
Wang Z, Zhao X, Zhao G, Guo Y, Lu H, Mu W, Zhong J, Garcia-Barrio M, Zhang J, Chen YE, Chang L. PRDM16 deficiency in vascular smooth muscle cells aggravates abdominal aortic aneurysm. JCI Insight 2023; 8:e167041. [PMID: 37079380 PMCID: PMC10393233 DOI: 10.1172/jci.insight.167041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/19/2023] [Indexed: 04/21/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is usually asymptomatic until life-threatening complications occur, predominantly involving aortic rupture. Currently, no drug-based treatments are available, primarily due to limited understanding of AAA pathogenesis. The transcriptional regulator PR domain-containing protein 16 (PRDM16) is highly expressed in the aorta, but its functions in the aorta are largely unknown. By RNA-seq analysis, we found that vascular smooth muscle cell-specific (VSMC-specific) Prdm16-knockout (Prdm16SMKO) mice already showed extensive changes in the expression of genes associated with extracellular matrix (ECM) remodeling and inflammation in the abdominal aorta under normal housing conditions without any pathological stimuli. Human AAA lesions displayed lower PRDM16 expression. Periadventitial elastase application to the suprarenal region of the abdominal aorta aggravated AAA formation in Prdm16SMKO mice. During AAA development, VSMCs undergo apoptosis because of both intrinsic and environmental changes, including inflammation and ECM remodeling. Prdm16 deficiency promoted inflammation and apoptosis in VSMCs. A disintegrin and metalloproteinase 12 (ADAM12) is a gelatinase that can degrade various ECMs. We found that ADAM12 is a target of transcriptional repression by PRDM16. Adam12 knockdown reversed VSMC apoptosis induced by Prdm16 deficiency. Our study demonstrated that PRDM16 deficiency in VSMCs promoted ADAM12 expression and aggravates AAA formation, which may provide potential targets for AAA treatment.
Collapse
Affiliation(s)
- Zhenguo Wang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Xiangjie Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Key Laboratory of Animal Cellular and Genetics, Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, P.R. China
| | - Guizhen Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Pharmacology, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Wenjuan Mu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Juan Zhong
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Minerva Garcia-Barrio
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Kuret T, Kreft ME, Romih R, Veranič P. Cannabidiol as a Promising Therapeutic Option in IC/BPS: In Vitro Evaluation of Its Protective Effects against Inflammation and Oxidative Stress. Int J Mol Sci 2023; 24:ijms24055055. [PMID: 36902479 PMCID: PMC10003465 DOI: 10.3390/ijms24055055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Several animal studies have described the potential effect of cannabidiol (CBD) in alleviating the symptoms of interstitial cystitis/bladder pain syndrome (IC/BPS), a chronic inflammatory disease of the urinary bladder. However, the effects of CBD, its mechanism of action, and modulation of downstream signaling pathways in urothelial cells, the main effector cells in IC/BPS, have not been fully elucidated yet. Here, we investigated the effect of CBD against inflammation and oxidative stress in an in vitro model of IC/BPS comprised of TNFα-stimulated human urothelial cells SV-HUC1. Our results show that CBD treatment of urothelial cells significantly decreased TNFα-upregulated mRNA and protein expression of IL1α, IL8, CXCL1, and CXCL10, as well as attenuated NFκB phosphorylation. In addition, CBD treatment also diminished TNFα-driven cellular reactive oxygen species generation (ROS), by increasing the expression of the redox-sensitive transcription factor Nrf2, the antioxidant enzymes superoxide dismutase 1 and 2, and hem oxygenase 1. CBD-mediated effects in urothelial cells may occur by the activation of the PPARγ receptor since inhibition of PPARγ resulted in significantly diminished anti-inflammatory and antioxidant effects of CBD. Our observations provide new insights into the therapeutic potential of CBD through modulation of PPARγ/Nrf2/NFκB signaling pathways, which could be further exploited in the treatment of IC/BPS.
Collapse
|
5
|
Henrik SZŐKE, István BÓKKON, David M, Jan V, Ágnes K, Zoltán K, Ferenc F, Tibor K, László SL, Ádám D, Odilia M, Andrea K. The innate immune system and fever under redox control: A Narrative Review. Curr Med Chem 2022; 29:4324-4362. [DOI: 10.2174/0929867329666220203122239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/21/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022]
Abstract
ABSTRACT:
In living cells, redox potential is vitally important for normal physiological processes that are closely regulated by antioxidants, free amino acids and proteins that either have reactive oxygen and nitrogen species capture capability or can be compartmentalized. Although hundreds of experiments support the regulatory role of free radicals and their derivatives, several authors continue to claim that these perform only harmful and non-regulatory functions. In this paper we show that countless intracellular and extracellular signal pathways are directly or indirectly linked to regulated redox processes. We also briefly discuss how artificial oxidative stress can have important therapeutic potential and the possible negative effects of popular antioxidant supplements.
Next, we present the argument supported by a large number of studies that several major components of innate immunity, as well as fever, is also essentially associated with regulated redox processes. Our goal is to point out that the production of excess or unregulated free radicals and reactive species can be secondary processes due to the perturbed cellular signal pathways. However, researchers on pharmacology should consider the important role of redox mechanisms in the innate immune system and fever.
Collapse
Affiliation(s)
- SZŐKE Henrik
- Doctoral School of Health Sciences, University of Pécs, Pécs, Hungary
| | - BÓKKON István
- Neuroscience and Consciousness Research Department, Vision Research Institute,
Lowell, MA, USA
| | - martin David
- Department of Human Medicine, University Witten/Herdecke, Witten, Germany
| | - Vagedes Jan
- University Children’s Hospital, Tuebingen University, Tuebingen, Germany
| | - kiss Ágnes
- Doctoral School of Health Sciences, University of Pécs, Pécs, Hungary
| | - kovács Zoltán
- Doctoral School of Health Sciences, University of Pécs, Pécs, Hungary
| | - fekete Ferenc
- Department of Nyerges Gábor Pediatric Infectology, Heim Pál National Pediatric Institute, Budapest, Hungary
| | - kocsis Tibor
- Department of Clinical Governance, Hungarian National Ambulance Service, Budapest, Hungary
| | | | | | | | - kisbenedek Andrea
- Doctoral School of Health Sciences, University of Pécs, Pécs, Hungary
| |
Collapse
|
6
|
Lima TS, Mallya S, Jankeel A, Messaoudi I, Lodoen MB. Toxoplasma gondii Extends the Life Span of Infected Human Neutrophils by Inducing Cytosolic PCNA and Blocking Activation of Apoptotic Caspases. mBio 2021; 12:e02031-20. [PMID: 33500339 PMCID: PMC7858050 DOI: 10.1128/mbio.02031-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/01/2020] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii is an intracellular protozoan parasite that has the remarkable ability to infect and replicate in neutrophils, immune cells with an arsenal of antimicrobial effector mechanisms. We report that T. gondii infection extends the life span of primary human peripheral blood neutrophils by delaying spontaneous apoptosis, serum starvation-induced apoptosis, and tumor necrosis alpha (TNF-α)-mediated apoptosis. T. gondii blockade of apoptosis was associated with an inhibition of processing and activation of the apoptotic caspases caspase-8 and -3, decreased phosphatidylserine exposure on the plasma membrane, and reduced cell death. We performed a global transcriptome analysis of T. gondii-infected peripheral blood neutrophils using RNA sequencing (RNA-Seq) and identified gene expression changes associated with DNA replication and DNA repair pathways, which in mature neutrophils are indicative of changes in regulators of cell survival. Consistent with the RNA-Seq data, T. gondii infection upregulated transcript and protein expression of PCNA, which is found in the cytosol of human neutrophils, where it functions as a key inhibitor of apoptotic pro-caspases. Infection of neutrophils resulted in increased interaction of PCNA with pro-caspase-3. Inhibition of this interaction with an AlkB homologue 2 PCNA-interacting motif (APIM) peptide reversed the infection-induced delay in cell death. Taken together, these findings indicate a novel strategy by which T. gondii manipulates cell life span in primary human neutrophils, which may allow the parasite to maintain an intracellular replicative niche and avoid immune clearance.IMPORTANCEToxoplasma gondii is an obligate intracellular parasite that can cause life-threatening disease in immunocompromised individuals and in the developing fetus. Interestingly, T. gondii has evolved strategies to successfully manipulate the host immune system to establish a productive infection and evade host defense mechanisms. Although it is well documented that neutrophils are mobilized during acute T. gondii infection and infiltrate the site of infection, these cells can also be actively infected by T. gondii and serve as a replicative niche for the parasite. However, there is a limited understanding of the molecular processes occurring within T. gondii-infected neutrophils. This study reveals that T. gondii extends the life span of human neutrophils by inducing the expression of PCNA, which prevents activation of apoptotic caspases, thus delaying apoptosis. This strategy may allow the parasite to preserve its replicative intracellular niche.
Collapse
Affiliation(s)
- Tatiane S Lima
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Sharmila Mallya
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
7
|
Matsuoka Y, Al-Shareef H, Kogo M, Nakahara H. Effects of decreased Rac activity and malignant state on oral squamous cell carcinoma in vitro. PLoS One 2021; 16:e0212323. [PMID: 33444335 PMCID: PMC7808617 DOI: 10.1371/journal.pone.0212323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/16/2019] [Indexed: 11/19/2022] Open
Abstract
Rac proteins, members of the Rho family of small GTP-binding proteins, have been implicated in transducing a number of signals for various biological mechanisms, including cell cytoskeleton organization, transcription, proliferation, migration, and cancer cell motility. Among human cancers, Rac proteins are highly activated by either overexpression of the genes, up-regulation of the protein, or by mutations that allow the protein to elude normal regulatory signaling pathways. Rac proteins are involved in controlling cell survival and apoptosis. The effects of Rac inhibition by the Rac-specific small molecule inhibitor NSC23766 or by transfection of dominant negative Rac (Rac-DN) were examined on three human-derived oral squamous cell carcinoma cell lines that exhibit different malignancy grades, OSC-20 (grade 3), OSC-19 (grade 4C), and HOC313 (grade 4D). Upon suppression of Rac, OSC-19 and HOC313 cells showed significant decreases in Rac activity and resulted in condensation of the nuclei and up-regulation of c-Jun N-terminal kinase (JNK), leading to caspase-dependent apoptosis. In contrast, OSC-20 cells showed only a slight decrease in Rac activity, which resulted in slight activation of JNK and no change in the nuclei. Fibroblasts treated with NSC23766 also showed only a slight decrease in Rac activity with no change in the nuclei or JNK activity. Our results indicated that apoptosis elicited by the inhibition of Rac depended on the extent of decreased Rac activity and the malignant state of the squamous cell carcinoma. In addition, activation of JNK strongly correlated with apoptosis. Rac inhibition may represent a novel therapeutic approach for cancer treatment.
Collapse
Affiliation(s)
- Yudai Matsuoka
- The First Department of Oral & Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hani Al-Shareef
- Department of Oral & Maxillofacial Surgery, Osaka City University Graduate School of Medicine, Osaka, Osaka, Japan
| | - Mikihiko Kogo
- The First Department of Oral & Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hirokazu Nakahara
- The First Department of Oral & Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
- Department of Oral & Maxillofacial Surgery, Osaka City University Graduate School of Medicine, Osaka, Osaka, Japan
- * E-mail:
| |
Collapse
|
8
|
Li Y, Mooney EC, Xia XJ, Gupta N, Sahingur SE. A20 Restricts Inflammatory Response and Desensitizes Gingival Keratinocytes to Apoptosis. Front Immunol 2020; 11:365. [PMID: 32218782 PMCID: PMC7078700 DOI: 10.3389/fimmu.2020.00365] [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: 05/07/2019] [Accepted: 02/14/2020] [Indexed: 12/16/2022] Open
Abstract
The pathophysiology of periodontal disease involves a perturbed immune system to a dysbiotic microflora leading to unrestrained inflammation, collateral tissue damage, and various systemic complications. Gingival epithelial cells function as an important part of immunity to restrict microbial invasion and orchestrate the subsequent innate responses. A20 (TNFAIP3), an ubiquitin-editing enzyme, is one of the key regulators of inflammation and cell death in numerous tissues including gastrointestinal tract, skin, and lungs. Emerging evidence indicates A20 as an essential molecule in the oral mucosa as well. In this study, we characterized the role of A20 in human telomerase immortalized gingival keratinocytes (TIGKs) through loss and gain of function assays in preclinical models of periodontitis. Depletion of A20 through gene editing in TIGKs significantly increased IL-6 and IL-8 secretion in response to Porphyromonas gingivalis infection while A20 over-expression dampened the cytokine production compared to A20 competent cells through modulating NF-κB signaling pathway. In the subsequent experiments which assessed apoptosis, A20 depleted TIGKs displayed increased levels of cleaved caspase 3 and DNA fragmentation following P. gingivalis infection and TNF/CHX challenge compared to A20 competent cells. Consistently, there was reduced apoptosis in the cells overexpressing A20 compared to the control cells expressing GFP further substantiating the role of A20 in regulating gingival epithelial cell fate in response to exogenous insult. Collectively, our findings reveal first systematic evidence and demonstrate that A20 acts as a regulator of inflammatory response in gingival keratinocytes through its effect on NF-κB signaling and desensitizes cells to bacteria and cytokine induced apoptosis in the oral mucosa. As altered A20 levels can have profound effect on different cellular responses, future studies will determine whether A20-targeted therapies can be exploited to restrain periodontal inflammation and maintain oral mucosa tissue homeostasis.
Collapse
Affiliation(s)
- Yajie Li
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erin C Mooney
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, United States
| | - Xia-Juan Xia
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nitika Gupta
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sinem Esra Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
9
|
Evidence of necroptosis in osteoarthritic disease: investigation of blunt mechanical impact as possible trigger in regulated necrosis. Cell Death Dis 2019; 10:683. [PMID: 31527653 PMCID: PMC6746800 DOI: 10.1038/s41419-019-1930-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
Joint injuries are highly associated with cell death and development of posttraumatic osteoarthritis (PTOA). The present study focused on necroptosis as a possible modality of chondrocyte death after cartilage trauma and its relevance in OA disease in general. For this purpose, apoptosis- and necroptosis-associated markers were determined in highly degenerated (ICRS ≥ 3) as well as macroscopically intact cartilage tissue (ICRS ≤ 1) by means of real-time PCR and immunohistochemistry (IHC). Moreover, influence of blunt trauma and/or stimulation with cycloheximide (CHX), TNF-a, and caspase-inhibitor zVAD were investigated in cartilage explants (ICRS ≤ 1). Further characterization of necroptosis was performed in isolated chondrocytes. We found that gene expression levels of RIPK3 (4.2-fold, P < 0.0001) and MLKL (2.7-fold, P < 0.0001) were elevated in highly degenerated cartilage tissue, which was confirmed by IHC staining. After ex vivo trauma and/or CHX/TNF stimulation, addition of zVAD further enhanced expression of necroptosis-related markers as well as release of PGE2 and nitric oxide, which was in line with increased cell death and subsequent release of intracellular HMGB1 and dsDNA in CHX/TNF stimulated chondrocytes. However, trauma and/or chemically induced cell death and subsequent release of pro-inflammatory mediators could be largely attenuated by RIPK1-inhibitor necrostatin 1 or antioxidant N-acetylcysteine. Overall, the study provided clear evidence of necroptotic cell death in OA disease. Moreover, a possible link between cartilage injury and necroptotic processes was found, depending on oxidative stress and cytokine release. These results contribute to further understanding of cell death in PTOA and development of novel therapeutic approaches.
Collapse
|
10
|
TNF Family Cytokines Induce Distinct Cell Death Modalities in the A549 Human Lung Epithelial Cell Line when Administered in Combination with Ricin Toxin. Toxins (Basel) 2019; 11:toxins11080450. [PMID: 31374990 PMCID: PMC6723388 DOI: 10.3390/toxins11080450] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 01/10/2023] Open
Abstract
Ricin is a member of the ribosome-inactivating protein (RIP) family of toxins and is classified as a biothreat agent by the Centers for Disease Control and Prevention (CDC). Inhalation, the most potent route of toxicity, triggers an acute respiratory distress-like syndrome that coincides with near complete destruction of the lung epithelium. We previously demonstrated that the TNF-related apoptosis-inducing ligand (TRAIL; CD253) sensitizes human lung epithelial cells to ricin-induced death. Here, we report that ricin/TRAIL-mediated cell death occurs via apoptosis and involves caspases -3, -7, -8, and -9, but not caspase-6. In addition, we show that two other TNF family members, TNF-α and Fas ligand (FasL), also sensitize human lung epithelial cells to ricin-induced death. While ricin/TNF-α- and ricin/FasL-mediated killing of A549 cells was inhibited by the pan-caspase inhibitor, zVAD-fmk, evidence suggests that these pathways were not caspase-dependent apoptosis. We also ruled out necroptosis and pyroptosis. Rather, the combination of ricin plus TNF-α or FasL induced cathepsin-dependent cell death, as evidenced by the use of several pharmacologic inhibitors. We postulate that the effects of zVAD-fmk were due to the molecule’s known off-target effects on cathepsin activity. This work demonstrates that ricin-induced lung epithelial cell killing occurs by distinct cell death pathways dependent on the presence of different sensitizing cytokines, TRAIL, TNF-α, or FasL.
Collapse
|
11
|
Miguel CA, Raggio MC, Villar MJ, Gonzalez SL, Coronel MF. Anti-allodynic and anti-inflammatory effects of 17α-hydroxyprogesterone caproate in oxaliplatin-induced peripheral neuropathy. J Peripher Nerv Syst 2019; 24:100-110. [PMID: 30680838 DOI: 10.1111/jns.12307] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/08/2019] [Accepted: 01/22/2019] [Indexed: 12/11/2022]
Abstract
Chemotherapy-induced peripheral neuropathy is a disabling condition induced by several frequently used chemotherapeutic drugs including the front-line agent oxaliplatin (OXA). Symptoms are predominantly sensory with the development of neuropathic pain. Alternative dosing protocols and treatment discontinuation are the only available therapeutic strategies. The aim of our work was to evaluate the potential of a synthetic derivative of progesterone, 17α-hydroxyprogesterone caproate (HPGC), in the prevention and treatment of OXA-evoked painful neuropathy. We also evaluated glial activation at the dorsal root ganglia (DRG) and spinal cord levels as a possible target mechanism underlying HPGC actions. Male rats were injected with OXA and HPGC following a prophylactic (HPGCp) or therapeutic (HPGCt) scheme (starting either before or after chemotherapy). The development of hypersensitivity and allodynic pain and the expression of neuronal and glial activation markers were evaluated. When compared to control animals, those receiving OXA showed a significant decrease in paw mechanical and thermal thresholds, with the development of allodynia. Animals treated with HPGCp showed patterns of response similar to those detected in control animals, while those treated with HPGCt showed a suppression of both hypersensitivities after HPGC administration. We also observed a significant increase in the mRNA levels of activating transcription factor 3, the transcription factor (c-fos), glial fibrillary acidic protein, ionized calcium binding adaptor protein 1, interleukin 1 beta (IL-1β) and tumor necrosis factor alpha (TNFα) in DRG and spinal cord of OXA-injected animals, and significantly lower levels in rats receiving OXA and HPGC. These results show that HPGC administration reduces neuronal and glial activation markers and is able to both prevent and suppress OXA-induced allodynia, suggesting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Constanza A Miguel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental - CONICET, Buenos Aires, Argentina
| | - María C Raggio
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental - CONICET, Buenos Aires, Argentina
| | - Marcelo J Villar
- Instituto de Investigaciones en Medicina Traslacional, Universidad Austral - CONICET, Buenos Aires, Argentina
| | - Susana L Gonzalez
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental - CONICET, Buenos Aires, Argentina.,Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María F Coronel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental - CONICET, Buenos Aires, Argentina.,Facultad de Ciencias Biomédicas, Universidad Austral - CONICET, Buenos Aires, Argentina
| |
Collapse
|
12
|
Ogura K, Terasaki Y, Miyoshi-Akiyama T, Terasaki M, Moss J, Noda M, Yahiro K. Vibrio cholerae Cholix Toxin-Induced HepG2 Cell Death is Enhanced by Tumor Necrosis Factor-Alpha Through ROS and Intracellular Signal-Regulated Kinases. Toxicol Sci 2018; 156:455-468. [PMID: 28087840 DOI: 10.1093/toxsci/kfx009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cholix toxin (Cholix) from Vibrio cholerae is a potent virulence factor exhibiting ADP-ribosyltransferase activity on eukaryotic elongation factor 2 (eEF2) of host cells, resulting in the inhibition of protein synthesis. Administration of Cholix or its homologue Pseudomonas exotoxin A (PEA) to mice causes lethal hepatocyte damage. In this study, we demonstrate cytotoxicity of Cholix on human hepatocytes in the presence of tumor necrosis factor α (TNF-α), which has been reported to play a fatal role in PEA administered to mice. Compared with incubating HepG2 cells with Cholix alone, co-treatment with TNF-α and Cholix (TNF-α/Cholix) significantly enhanced the activation of caspases, cytochrome c release from mitochondria into cytoplasm, and poly-ADP-ribose polymerase (PARP) cleavage, while incubation with TNF-α alone or co-treatment with TNF-α/catalytically inactive Cholix did not. In the early stage of cell death, Cholix increased phosphorylation of mitogen-activated protein kinases (e.g., p38, ERK, JNK) and Akt, which was not affected by TNF-α alone. MAPK inhibitors (SP600125, SB20852, and U0126) suppressed PARP cleavage induced by TNF-α/Cholix. Protein kinase inhibitor Go6976 suppressed JNK phosphorylation and PARP cleavage by TNF-α/Cholix. In contrast, PKC activator PMA in the absence of TNF-α promoted Cholix-induced PARP cleavage. Reactive oxygen species (ROS) inhibitor, N-acetyl cysteine (NAC), suppressed TNF-α/Cholix-induced JNK and ERK phosphorylation, resulting in inhibition of PARP cleavage. These data suggest that ROS and JNK pathways are important mediators of TNF-α/Cholix-induced HepG2 cell death.
Collapse
Affiliation(s)
- Kohei Ogura
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuhiro Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1590
| | - Masatoshi Noda
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kinnosuke Yahiro
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
13
|
Zhao M, Tang S, Xin J, Wei Y, Liu D. Reactive oxygen species induce injury of the intestinal epithelium during hyperoxia. Int J Mol Med 2017; 41:322-330. [PMID: 29138796 PMCID: PMC5746288 DOI: 10.3892/ijmm.2017.3247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
Long-term therapeutic hyperoxia may exert serious toxic effects on intestinal epithelial cells in vitro and in vivo. The aim of the present study was to investigate the cause of this intestinal injury under conditions of hyperoxia. Caco-2 cells were treated with different concentrations of hydrogen peroxide (H2O2) and 85% hyperoxia for 24 h. higher rates of injury of Caco-2 cells were observed in the hyperoxia and H2O2 groups compared with the control group. The reactive oxygen species (ROS) level of the hyperoxia group was significantly higher compared with that of the 400 µM H2O2 group. The protein and gene levels of RelA, RelB, hypoxia‑inducible factor-1α, tumor necrosis factor-α and apoptosis signal‑regulating kinase 1 were significantly higher in the hyperoxia and H2O2 groups compared with those in the control group. In conclusion, during hyperoxia, intestinal epithelial cells were destroyed and the levels of ROS were increased. Therefore, ROS may play an important role in intestinal injury in a hyperoxic environment.
Collapse
Affiliation(s)
- Min Zhao
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Shimiao Tang
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Junchi Xin
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Yingliang Wei
- Department of Orthopaedic Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Dongyan Liu
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
14
|
Zhao M, Tang S, Xin J, Liu D. Influence of reactive oxygen species on secretory component in the intestinal epithelium during hyperoxia. Exp Ther Med 2017; 14:4033-4040. [PMID: 29075338 PMCID: PMC5648505 DOI: 10.3892/etm.2017.5027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 05/19/2017] [Indexed: 01/23/2023] Open
Abstract
Redox imbalance is established in various human diseases. Treatment of intestinal epithelial cells with hyperoxia for a prolonged period of time may cause serious effects on redox balance. Secretory component (SC) protein is secreted by intestinal epithelial cells, and has a vital role in mucosal immune systems and intestinal defense. The present study aimed to investigate the influence of reactive oxygen species (ROS) on intestinal epithelial cells and intestinal epithelial SC protein under hyperoxic conditions. Caco-2 cells were treated with increasing concentrations of hydrogen peroxide (H2O2) or 85% O2 (hyperoxia) for 24 h. Flow cytometry, immunohistochemistry staining, western blot analysis and reverse transcription-quantitative polymerase chain reaction were performed to detect the expression levels of SC protein. Significantly increased apoptosis and mortality rates were observed in hyperoxia- and H2O2-treated Caco-2 cells, as compared with the untreated control cells (P<0.05). Protein and mRNA expression levels of SC were significantly increased in hyperoxia- and H2O2-treated groups, as compared with the control group (P<0.05). During hyperoxia, intestinal epithelial cells were destroyed and ROS levels increased. Therefore, the results of the present study suggested that ROS might have an important role in intestinal injury in hyperoxic environments.
Collapse
Affiliation(s)
- Min Zhao
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Shimiao Tang
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Junchi Xin
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Dongyan Liu
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
15
|
Haga S, Kanno A, Ozawa T, Morita N, Asano M, Ozaki M. Detection of Necroptosis in Ligand-Mediated and Hypoxia-Induced Injury of Hepatocytes Using a Novel Optic Probe-Detecting Receptor-Interacting Protein (RIP)1/RIP3 Binding. Oncol Res 2017; 26:503-513. [PMID: 28770700 PMCID: PMC7844641 DOI: 10.3727/096504017x15005102445191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Liver injury is often observed in various pathological conditions including posthepatectomy state and cancer chemotherapy. It occurs mainly as a consequence of the combined necrotic and apoptotic types of cell death. In order to study liver/hepatocyte injury by the necrotic type of cell death, we studied signal-regulated necrosis (necroptosis) by developing a new optic probe for detecting receptor-interacting protein kinase 1 (RIP)/RIP3 binding, an essential process for necroptosis induction. In the mouse hepatocyte cell line, TIB-73 cells, TNF-α/cycloheximide (T/C) induced RIP1/3 binding only when caspase activity was suppressed by the caspase-specific inhibitor z-VAD-fmk (zVAD). T/C/zVAD-induced RIP1/3 binding was inhibited by necrostatin-1 (Nec-1), an allosteric inhibitor of RIP1. The reduced cell survival by T/C/zVAD was improved by Nec-1. These facts indicate that T/C induces necroptosis of hepatocytes when the apoptotic pathway is inhibited/unavailable. FasL also induced cell death, which was only partially inhibited by zVAD, indicating the possible involvement of necroptosis rather than apoptosis. FasL activated caspase 3 and, similarly, induced RIP1/3 binding when the caspases were inactivated. Interestingly, FasL-induced RIP1/3 binding was significantly suppressed by the antioxidants Trolox and N-acetyl cysteine (NAC), suggesting the involvement of reactive oxygen species (ROS) in FasL-induced necroptotic cellular processes. H₂O₂, by itself, induced RIP1/3 binding that was suppressed by Nec-1, but not by zVAD. Hypoxia induced RIP1/3 binding after reoxygenation, which was suppressed by Nec-1 or by the antioxidants. Cell death induced by hypoxia/reoxygenation (H/R) was also improved by Nec-1. Similar to H₂O₂, H/R did not require caspase inhibition for RIP1/3 binding, suggesting the involvement of a caspase-independent mechanism for non-ligand-induced and/or redox-mediated necroptosis. These data indicate that ROS can induce necroptosis and mediate the FasL- and hypoxia-induced necroptosis via a molecular mechanism that differs from a conventional caspase-dependent pathway. In conclusion, necroptosis is potentially involved in liver/hepatocyte injury induced by oxidative stress and FasL in the absence of apoptosis.
Collapse
Affiliation(s)
- Sanae Haga
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Akira Kanno
- Department of Environmental Applied Chemistry, Faculty of Engineering, University of ToyamaToyamaJapan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of TokyoTokyoJapan
| | - Naoki Morita
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)Sapporo, HokkaidoJapan
| | - Mami Asano
- Laboratory of Molecular and Functional Bio-Imaging, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| |
Collapse
|
16
|
Babu D, Leclercq G, Motterlini R, Lefebvre RA. Differential Effects of CORM-2 and CORM-401 in Murine Intestinal Epithelial MODE-K Cells under Oxidative Stress. Front Pharmacol 2017; 8:31. [PMID: 28228725 PMCID: PMC5296622 DOI: 10.3389/fphar.2017.00031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 01/16/2017] [Indexed: 12/14/2022] Open
Abstract
Carbon monoxide (CO)-releasing molecules (CO-RMs) are intensively studied to provide cytoprotective and anti-inflammatory effects of CO in inflammatory conditions including intestinal inflammation. The water-soluble CORM-A1 reduced apoptosis and NADPH oxidase (NOX)-derived reactive oxygen species (ROS) induced by tumor necrosis factor (TNF)-α/cycloheximide (CHX) in mouse MODE-K intestinal epithelial cells (IECs), without influencing TNF-α/CHX-induced mitochondrial superoxide anion (O2•–). The aim of the present study in the same model was to comparatively investigate the influence of lipid-soluble CORM-2 and water-soluble CORM-401, shown in vitro to release more CO under oxidative conditions. CORM-2 abolished TNF-α/CHX-induced total cellular ROS whereas CORM-401 partially reduced it, both partially reducing TNF-α/CHX-induced cell death. Only CORM-2 increased mitochondrial O2•– production after 2 h of incubation. CORM-2 reduced TNF-α/CHX-, rotenone- and antimycin-A-induced mitochondrial O2•– production; CORM-401 only reduced the effect of antimycin-A. Co-treatment with CORM-401 during 1 h exposure to H2O2 reduced H2O2 (7.5 mM)-induced ROS production and cell death, whereas CORM-2 did not. The study illustrates the importance of the chemical characteristics of different CO-RMs. The lipid solubility of CORM-2 might contribute to its interference with TNF-α/CHX-induced mitochondrial ROS signaling, at least in mouse IECs. CORM-401 is more effective than other CO-RMs under H2O2-induced oxidative stress conditions.
Collapse
Affiliation(s)
- Dinesh Babu
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| | - Georges Leclercq
- Department of Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| | - Roberto Motterlini
- INSERM U955, Faculty of Medicine, Equipe 12 and University Paris Est Créteil, France
| | - Romain A Lefebvre
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| |
Collapse
|
17
|
Babu D, Leclercq G, Motterlini R, Lefebvre RA. Differential Effects of CORM-2 and CORM-401 in Murine Intestinal Epithelial MODE-K Cells under Oxidative Stress. Front Pharmacol 2017. [PMID: 28228725 DOI: 10.3389/fphar.2017.00031/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
Carbon monoxide (CO)-releasing molecules (CO-RMs) are intensively studied to provide cytoprotective and anti-inflammatory effects of CO in inflammatory conditions including intestinal inflammation. The water-soluble CORM-A1 reduced apoptosis and NADPH oxidase (NOX)-derived reactive oxygen species (ROS) induced by tumor necrosis factor (TNF)-α/cycloheximide (CHX) in mouse MODE-K intestinal epithelial cells (IECs), without influencing TNF-α/CHX-induced mitochondrial superoxide anion ([Formula: see text]). The aim of the present study in the same model was to comparatively investigate the influence of lipid-soluble CORM-2 and water-soluble CORM-401, shown in vitro to release more CO under oxidative conditions. CORM-2 abolished TNF-α/CHX-induced total cellular ROS whereas CORM-401 partially reduced it, both partially reducing TNF-α/CHX-induced cell death. Only CORM-2 increased mitochondrial [Formula: see text] production after 2 h of incubation. CORM-2 reduced TNF-α/CHX-, rotenone- and antimycin-A-induced mitochondrial [Formula: see text] production; CORM-401 only reduced the effect of antimycin-A. Co-treatment with CORM-401 during 1 h exposure to H2O2 reduced H2O2 (7.5 mM)-induced ROS production and cell death, whereas CORM-2 did not. The study illustrates the importance of the chemical characteristics of different CO-RMs. The lipid solubility of CORM-2 might contribute to its interference with TNF-α/CHX-induced mitochondrial ROS signaling, at least in mouse IECs. CORM-401 is more effective than other CO-RMs under H2O2-induced oxidative stress conditions.
Collapse
Affiliation(s)
- Dinesh Babu
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| | - Georges Leclercq
- Department of Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| | - Roberto Motterlini
- INSERM U955, Faculty of Medicine, Equipe 12 and University Paris Est Créteil, France
| | - Romain A Lefebvre
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University Ghent, Belgium
| |
Collapse
|
18
|
Singh DP, Borse SP, Nivsarkar M. Clinical importance of nonsteroidal anti-inflammatory drug enteropathy: the relevance of tumor necrosis factor as a promising target. Transl Res 2016; 175:76-91. [PMID: 27083387 DOI: 10.1016/j.trsl.2016.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/18/2016] [Accepted: 03/21/2016] [Indexed: 12/13/2022]
Abstract
The pathogenesis of nonsteroidal anti-inflammatory drug (NSAID) enteropathy is still unclear, and consequently, there is no approved therapeutic strategy for ameliorating such damage. On the other hand, molecular treatment strategies targeting tumor necrosis factor (TNF) exerts beneficial effects on NSAID-induced intestinal lesions in rodents and rheumatoid arthritis patients. Thus, TNF appears to be a potential therapeutic target for both the prevention and treatment of NSAID enteropathy. However, the causative relationship between TNF and NSAID enteropathy is largely unknown. Currently approved anti-TNF agents are highly expensive and exhibit numerous side effects. Hence, in this review, the pivotal role of TNF in NSAID enteropathy has been summarized and plant-derived polyphenols have been suggested as useful alternative anti-TNF agents because of their ability to suppress TNF activated inflammatory pathways both in vitro and in vivo.
Collapse
Affiliation(s)
- Devendra Pratap Singh
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, Ahmedabad, Gujarat, India; Registered Ph.D Scholar (External) at Institute of Pharmacy, NIRMA University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, India
| | - Swapnil P Borse
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, Ahmedabad, Gujarat, India; Registered Ph.D Scholar (External) at Institute of Pharmacy, NIRMA University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, India
| | - Manish Nivsarkar
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, Ahmedabad, Gujarat, India.
| |
Collapse
|
19
|
Zhou S, Han Q, Wang R, Li X, Wang Q, Wang H, Wang J, Ma Y. PRDX2 protects hepatocellular carcinoma SMMC-7721 cells from oxidative stress. Oncol Lett 2016; 12:2217-2221. [PMID: 27602166 DOI: 10.3892/ol.2016.4899] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 06/02/2016] [Indexed: 12/31/2022] Open
Abstract
Peroxiredoxin2 (PRDX2) is a member of the peroxiredoxin family of antioxidant enzymes. A number of previous studies have indicated that PRDX2 may serve a cell type-dependent role in tumorigenesis. Recently, PRDX2 has been identified to be the new target of miR-122a, which has been demonstrated to be frequently downregulated in hepatocellular carcinoma (HCC). Thus, PRDX2 may have a pro-tumorigenic role in HCC. Because the role of PRDX2 in HCC has not yet been reported, it is of interest to explore how PRDX2 may affect reactive oxygen species (ROS)-mediated cell death in HCC cells. The present study analyzed the effects of PRDX2 knockdown or overexpression on hydrogen peroxide (H2O2)-induced cell death in HCC SMMC-7721 cells. Tumor necrosis factor-α (TNF-α)-induced cell death upon PRDX2 knockdown or overexpression was also examined in SMMC-7721 cells. It was found that PRDX2 knockdown augmented H2O2-induced cell death in SMMC-7721 cells, whereas PRDX2 overexpression exhibited opposite effects. By contrast, PRDX2 knockdown enhanced TNF-α-induced apoptosis, whereas PRDX2 overexpression reduced it, even though both treatments showed little effects on TNF-α-induced necrosis in SMMC-7721 cells. Further exploration confirmed PRDX2 knockdown led to enhanced ROS generation in response to H2O2. Taken together, the present study supports that PRDX2 serves a pro-tumorigenic role in HCC through, at least partially, limiting ROS-mediated apoptosis under oxidative stress.
Collapse
Affiliation(s)
- Silei Zhou
- Key Laboratory of Cellular and Molecular Immunology, Henan University, Kaifeng, Henan 475001, P.R. China; Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Quanli Han
- Department of Medical Oncology 2, Chinese PLA General Hospital & Chinese PLA Medical Academy, Beijing 100853, P.R. China
| | - Ru Wang
- Clinical Laboratory, 305 Hospital of PLA, Beijing 100017, P.R. China
| | - Xin Li
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Qingyang Wang
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Huizhong Wang
- Clinical Laboratory, 305 Hospital of PLA, Beijing 100017, P.R. China
| | - Jing Wang
- Department of Radiation Oncology, Chinese PLA General Hospital & Chinese PLA Medical Academy, Beijing 100853, P.R. China
| | - Yuanfang Ma
- Key Laboratory of Cellular and Molecular Immunology, Henan University, Kaifeng, Henan 475001, P.R. China
| |
Collapse
|
20
|
Keap1-Independent Regulation of Nrf2 Activity by Protein Acetylation and a BET Bromodomain Protein. PLoS Genet 2016; 12:e1006072. [PMID: 27233051 PMCID: PMC4883770 DOI: 10.1371/journal.pgen.1006072] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/30/2016] [Indexed: 12/30/2022] Open
Abstract
Mammalian BET proteins comprise a family of bromodomain-containing epigenetic regulators with complex functions in chromatin organization and gene regulation. We identified the sole member of the BET protein family in Drosophila, Fs(1)h, as an inhibitor of the stress responsive transcription factor CncC, the fly ortholog of Nrf2. Fs(1)h physically interacts with CncC in a manner that requires the function of its bromodomains and the acetylation of CncC. Treatment of cultured Drosophila cells or adult flies with fs(1)h RNAi or with the BET protein inhibitor JQ1 de-represses CncC transcriptional activity and engages protective gene expression programs. The mechanism by which Fs(1)h inhibits CncC function is distinct from the canonical mechanism that stimulates Nrf2 function by abrogating Keap1-dependent proteasomal degradation. Consistent with the independent modes of CncC regulation by Keap1 and Fs(1)h, combinations of drugs that can specifically target these pathways cause a strong synergistic and specific activation of protective CncC- dependent gene expression and boosts oxidative stress resistance. This synergism might be exploitable for the design of combinatorial therapies to target diseases associated with oxidative stress or inflammation. Nrf2-related transcription factors regulate gene expression programs that protect organisms against chemical or oxidative stress. Nrf2-activating drugs hold promise for the treatment of diseases that are connected to oxidative stress or inflammation. We identified Fs(1)h, a bromodomain-containing BET protein, as a negative regulator of Nrf2 function in Drosophila. BET proteins are involved in transcription regulation and chromatin organization and have been implicated in several diseases, including cancer. Fs(1)h interacts with acetylated lysines on CncC, the homolog of Nrf2 in Drosophila, and thereby prevents target gene activation. Nrf2 can be released from this inhibitory effect by small molecules that specifically interfere with the binding of BET proteins to acetylated targets. Fs(1)h regulates Nrf2 independently of Keap1, a well-studied Nrf2 regulator. Consequently, chemical inhibitors of Keap1 and of Fs(1)h can be combined to achieve synergistic activation of Nrf2 target genes and strongly boost oxidative stress tolerance in Drosophila. The Keap1-independent mechanism of Nrf2 regulation is conserved in mammals. We suggest that the synergistic effect of combinatorial Nrf2 targeting drugs may be effective for the treatment of different oxidative stress and inflammation-related diseases.
Collapse
|
21
|
Lee T, Lee E, Arrollo D, Lucas PC, Parameswaran N. Non-Hematopoietic β-Arrestin1 Confers Protection Against Experimental Colitis. J Cell Physiol 2015; 231:992-1000. [PMID: 26479868 DOI: 10.1002/jcp.25216] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/15/2015] [Indexed: 12/26/2022]
Abstract
β-Arrestins are multifunctional scaffolding proteins that modulate G protein-coupled receptor (GPCR)-dependent and -independent cell signaling pathways in various types of cells. We recently demonstrated that β-arrestin1 (β-arr1) deficiency strikingly attenuates dextran sodium sulfate (DSS)-induced colitis in mice. Since DSS-induced colitis is in part dependent on gut epithelial injury, we examined the role of β-arr1 in intestinal epithelial cells (IECs) using a colon epithelial cell line, SW480 cells. Surprisingly, we found that knockdown of β-arr1 in SW480 cells enhanced epithelial cell death via a caspase-3-dependent process. To understand the in vivo relevance and potential cell type-specific role of β-arr1 in colitis development, we generated bone marrow chimeras with β-arr1 deficiency in either the hematopoietic or non-hematopoietic compartment. Reconstituted chimeric mice were then subjected to DSS-induced colitis. Similar to our previous findings, β-arr1 deficiency in the hematopoietic compartment protected mice from DSS-induced colitis. However, consistent with the role of β-arr1 in epithelial apoptosis in vitro, non-hematopoietic β-arr1 deficiency led to an exacerbated colitis phenotype. To further understand signaling mechanisms, we examined the effect of β-arr1 on TNF-α-mediated NFκB and MAPK pathways. Our results demonstrate that β-arr1 has a critical role in modulating ERK, JNK and p38 MAPK pathways mediated by TNF-α in IECs. Together, our results show that β-arr1-dependent signaling in hematopoietic and non-hematopoietic cells differentially regulates colitis pathogenesis and further demonstrates that β-arr1 in epithelial cells inhibits TNF-α-induced cell death pathways.
Collapse
Affiliation(s)
- Taehyung Lee
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Eunhee Lee
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - David Arrollo
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
22
|
Babu D, Leclercq G, Goossens V, Remijsen Q, Vandenabeele P, Motterlini R, Lefebvre RA. Antioxidant potential of CORM-A1 and resveratrol during TNF-α/cycloheximide-induced oxidative stress and apoptosis in murine intestinal epithelial MODE-K cells. Toxicol Appl Pharmacol 2015; 288:161-78. [PMID: 26187750 DOI: 10.1016/j.taap.2015.07.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/03/2015] [Accepted: 07/07/2015] [Indexed: 12/26/2022]
Abstract
Targeting excessive production of reactive oxygen species (ROS) could be an effective therapeutic strategy to prevent oxidative stress-associated gastrointestinal inflammation. NADPH oxidase (NOX) and mitochondrial complexes (I and II) are the major sources of ROS production contributing to TNF-α/cycloheximide (CHX)-induced apoptosis in the mouse intestinal epithelial cell line, MODE-K. In the current study, the influence of a polyphenolic compound (resveratrol) and a water-soluble carbon monoxide (CO)-releasing molecule (CORM-A1) on the different sources of TNF-α/CHX-induced ROS production in MODE-K cells was assessed. This was compared with H2O2-, rotenone- or antimycin-A-induced ROS-generating systems. Intracellular total ROS, mitochondrial-derived ROS and mitochondrial superoxide anion (O2(-)) production levels were assessed. Additionally, the influence on TNF-α/CHX-induced changes in mitochondrial membrane potential (Ψm) and mitochondrial function was studied. In basal conditions, CORM-A1 did not affect intracellular total or mitochondrial ROS levels, while resveratrol increased intracellular total ROS but reduced mitochondrial ROS production. TNF-α/CHX- and H2O2-mediated increase in intracellular total ROS production was reduced by both resveratrol and CORM-A1, whereas only resveratrol attenuated the increase in mitochondrial ROS triggered by TNF-α/CHX. CORM-A1 decreased antimycin-A-induced mitochondrial O2(-) production without any influence on TNF-α/CHX- and rotenone-induced mitochondrial O2(-) levels, while resveratrol abolished all three effects. Finally, resveratrol greatly reduced and abolished TNF-α/CHX-induced mitochondrial depolarization and mitochondrial dysfunction, while CORM-A1 only mildly affected these parameters. These data indicate that the cytoprotective effect of resveratrol is predominantly due to mitigation of mitochondrial ROS, while CORM-A1 acts solely on NOX-derived ROS to protect MODE-K cells from TNF-α/CHX-induced cell death. This might explain the more pronounced cytoprotective effect of resveratrol.
Collapse
Affiliation(s)
- Dinesh Babu
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, Belgium.
| | - Georges Leclercq
- Department of Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Belgium
| | - Vera Goossens
- Inflammation Research Center, Molecular Signaling and Cell Death Unit, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Molecular Signaling and Cell Death Unit, Ghent University, Ghent, Belgium
| | - Quinten Remijsen
- Inflammation Research Center, Molecular Signaling and Cell Death Unit, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Molecular Signaling and Cell Death Unit, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Inflammation Research Center, Molecular Signaling and Cell Death Unit, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Molecular Signaling and Cell Death Unit, Ghent University, Ghent, Belgium
| | - Roberto Motterlini
- Inserm U955, Equipe 12 and University Paris-Est Créteil, Faculty of Medicine, F-94000 Créteil, France
| | - Romain A Lefebvre
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, Belgium
| |
Collapse
|
23
|
Babu D, Leclercq G, Goossens V, Vanden Berghe T, Van Hamme E, Vandenabeele P, Lefebvre RA. Mitochondria and NADPH oxidases are the major sources of TNF-α/cycloheximide-induced oxidative stress in murine intestinal epithelial MODE-K cells. Cell Signal 2015; 27:1141-58. [DOI: 10.1016/j.cellsig.2015.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 02/22/2015] [Indexed: 01/26/2023]
|
24
|
Zhao X, Fu J, Xu A, Yu L, Zhu J, Dai R, Su B, Luo T, Li N, Qin W, Wang B, Jiang J, Li S, Chen Y, Wang H. Gankyrin drives malignant transformation of chronic liver damage-mediated fibrosis via the Rac1/JNK pathway. Cell Death Dis 2015; 6:e1751. [PMID: 25950481 PMCID: PMC4669699 DOI: 10.1038/cddis.2015.120] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/17/2015] [Accepted: 03/24/2015] [Indexed: 02/07/2023]
Abstract
Hepatocarcinogenesis is a complex process involving chronic liver injury, inflammation, unregulated wound healing, subsequent fibrosis and carcinogenesis. To decipher the molecular mechanism underlying transition from chronic liver injury to dysplasia, we investigated the oncogenic role of gankyrin (PSMD10 or p28GANK) during malignant transformation in a transgenic mouse model. Here, we find that gankyrin increased in patients with cirrhosis. In addition to more severe liver fibrosis and tumorigenesis after DEN plus CCl4 treatment, hepatocyte-specific gankyrin-overexpressing mice (gankyrinhep) exhibited malignant transformation from liver fibrosis to tumors even under single CCl4 administration, whereas wild-type mice merely experienced fibrosis. Consistently, enhanced hepatic injury, severe inflammation and strengthened compensatory proliferation occurred in gankyrinhep mice during CCl4 performance. This correlated with augmented expressions of cell cycle-related genes and abnormal activation of Rac1/c-jun N-terminal kinase (JNK). Pharmacological inhibition of the Rac1/JNK pathway attenuated hepatic fibrosis and prevented CCl4-induced carcinogenesis in gankyrinhep mice. Together, these findings suggest that gankyrin promotes liver fibrosis/cirrhosis progression into hepatocarcinoma relying on a persistent liver injury and inflammatory microenvironment. Blockade of Rac1/JNK activation impeded gankyrin-mediated hepatocytic malignant transformation, indicating the combined inhibition of gankyrin and Rac1/JNK as a potential prevention mechanism for cirrhosis transition.
Collapse
Affiliation(s)
- X Zhao
- 1] Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China [2] International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - J Fu
- 1] International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China [2] National Center for Liver Cancer, Shanghai 200438, China
| | - A Xu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - L Yu
- 1] International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China [2] National Center for Liver Cancer, Shanghai 200438, China
| | - J Zhu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - R Dai
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - B Su
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - T Luo
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - N Li
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - W Qin
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - B Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - J Jiang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China
| | - S Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST, Department of Antomation, Tsinghua University, Beijing 100084, China
| | - Y Chen
- 1] International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China [2] National Center for Liver Cancer, Shanghai 200438, China
| | - H Wang
- 1] Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China [2] International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai 200438, China [3] National Center for Liver Cancer, Shanghai 200438, China [4] State Key Laboratory of Oncogenes and Related Genes, Cancer Institute of Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| |
Collapse
|
25
|
Porturas TP, Sun H, Buchlis G, Lou Y, Liang X, Cathopoulis T, Fayngerts S, Johnson DS, Wang Z, Chen YH. Crucial roles of TNFAIP8 protein in regulating apoptosis and Listeria infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:5743-50. [PMID: 25948813 DOI: 10.4049/jimmunol.1401987] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 03/30/2015] [Indexed: 01/01/2023]
Abstract
TNF-α-induced protein 8 (TNFAIP8 or TIPE) is a newly described regulator of cancer and infection. However, its precise roles and mechanisms of actions are not well understood. We report in this article that TNFAIP8 regulates Listeria monocytogenes infection by controlling pathogen invasion and host cell apoptosis in a RAC1 GTPase-dependent manner. TNFAIP8-knockout mice were resistant to lethal L. monocytogenes infection and had reduced bacterial load in the liver and spleen. TNFAIP8 knockdown in murine liver HEPA1-6 cells increased apoptosis, reduced bacterial invasion into cells, and resulted in dysregulated RAC1 activation. TNFAIP8 could translocate to plasma membrane and preferentially associate with activated RAC1-GTP. The combined effect of reduced bacterial invasion and increased sensitivity to TNF-α-induced clearance likely protected the TNFAIP8-knockout mice from lethal listeriosis. Thus, by controlling bacterial invasion and the death of infected cells through RAC1, TNFAIP8 regulates the pathogenesis of L. monocytogenes infection.
Collapse
Affiliation(s)
- Thomas P Porturas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - George Buchlis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Yunwei Lou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and Department of Immunology, Shandong University School of Medicine, Ji'nan 250012, People's Republic of China
| | - Xiaohong Liang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and Department of Immunology, Shandong University School of Medicine, Ji'nan 250012, People's Republic of China
| | - Terry Cathopoulis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Svetlana Fayngerts
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Derek S Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Zhaojun Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| |
Collapse
|
26
|
Kawarazaki W, Fujita T. Aberrant Rac1-mineralocorticoid receptor pathways in salt-sensitive hypertension. Clin Exp Pharmacol Physiol 2014; 40:929-36. [PMID: 24111570 DOI: 10.1111/1440-1681.12177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 09/11/2013] [Accepted: 09/17/2013] [Indexed: 12/17/2022]
Abstract
According to Guyton's model, impaired renal sodium excretion plays a key role in the increased salt sensitivity of blood pressure (BP). Several factors contribute to impaired renal sodium excretion, including the sympathetic nervous system, the renin-angiotensin system and aldosterone. Accumulating evidence suggests that abnormalities in aldosterone and its receptor (i.e. the mineralocorticoid receptor (MR)) are involved in the development of salt-sensitive (SS) hypertension. Patients with metabolic syndrome often exhibit hyperaldosteronism and are susceptible to SS hypertension. Aldosterone secretion from the adrenal glands is not suppressed in obese hypertensive rats fed a high-salt diet because of the abundant production of adipocyte-derived aldosterone-releasing factors, which are independent of the negative feedback regulation of aldosterone secretion by the renin-angiotensin-aldosterone system. Increased plasma aldosterone levels lead to SS hypertension via MR activation in the kidney. Renal MR activity is increased in Dahl salt-sensitive rats fed a high-salt diet, despite the appropriate suppression of plasma aldosterone levels. In this rat strain, activation of MR in the distal nephron causes salt-induced hypertension. This paradoxical response of the MR to salt loading can be attributed to activation of Rac1, a small GTPase. In the presence of aldosterone, activated Rac1 synergistically and directly activates MR in a ligand-independent manner. Thus, Rac1 activation in the kidney determines the salt sensitivity of BP. Together, the available evidence suggests that the aberrant Rac1-MR pathway plays a key role in the development of SS hypertension.
Collapse
Affiliation(s)
- Wakako Kawarazaki
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology=1, The University of Tokyo=1, Tokyo, Japan
| | | |
Collapse
|
27
|
Ray S, Zhao Y, Jamaluddin M, Edeh CB, Lee C, Brasier AR. Inducible STAT3 NH2 terminal mono-ubiquitination promotes BRD4 complex formation to regulate apoptosis. Cell Signal 2014; 26:1445-55. [PMID: 24657799 PMCID: PMC4067092 DOI: 10.1016/j.cellsig.2014.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/24/2014] [Accepted: 03/10/2014] [Indexed: 10/25/2022]
Abstract
Signal Transducers and Activator of Transcription-3 (STAT3) are latent transcription factors that are regulated by post-translational modifications (PTMs) in response to cellular activation by the IL-6 superfamily of cytokines to regulate cell cycle progression and/or apoptosis. Here we observe that STAT3 is inducibly mono-ubiquitinated and investigate its consequences. Using domain mapping and highly specific selected reaction monitoring-mass spectrometric assays, we identify lysine (K) 97 in its NH2-terminal domain as the major mono-ubiquitin conjugation site. We constructed a mono-ubiquitinated mimic consisting of a deubiquitinase-resistant monomeric ubiquitin fused to the NH2 terminus of STAT3 (ubiquitinated-STAT3 FP). In complex assays of ectopically expressed ubi-STAT3-FP, we observed enhanced complex formation with bromodomain-containing protein 4 (BRD4), a component of the activated positive transcriptional elongation factor (P-TEFb) complex. Chromatin immunoprecipitation experiments in STAT3(+/-) and STAT3(-/-) MEFs showed BRD4 recruitment to STAT3-dependent suppressor of cytokine signaling-3 gene (SOCS3). The effect of a selective small molecule inhibitor of BRD4, JQ1, to inhibit SOCS3 expression demonstrated the functional role of BRD4 for STAT3-dependent transcription. Additionally, ectopic ubiquitinated-STAT3 FP expression upregulated BCL2, BCL2L1, APEX1, SOD2, CCND1 and MYC expression indicating the role of ubiquitinated STAT3 in anti-apoptosis and cellular proliferation. Finally we observed that ubiquitinated-STAT3 FP suppressed TNFα-induced apoptotic cell death, indicating the functional importance of mono-ubiquitinated STAT3 in antiapoptotic gene expression. We conclude that STAT3 mono-ubiquitination is a key trigger in BRD4-dependent antiapoptotic and pro-proliferative gene expression programs. Thus, inhibiting the STAT3 mono-ubiquitination-BRD4 pathway may be a novel therapeutic target for the treatment of STAT3-dependent proliferative diseases.
Collapse
Affiliation(s)
- Sutapa Ray
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Yingxin Zhao
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Mohammad Jamaluddin
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Chukwudi B Edeh
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Chang Lee
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Allan R Brasier
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
28
|
Zhou X, Chen K, Lei H, Sun Z. Klotho gene deficiency causes salt-sensitive hypertension via monocyte chemotactic protein-1/CC chemokine receptor 2-mediated inflammation. J Am Soc Nephrol 2014; 26:121-32. [PMID: 24904083 DOI: 10.1681/asn.2013101033] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Klotho (KL) is a newly discovered aging suppressor gene. In mice, the KL gene extends the lifespan when overexpressed and shortens the lifespan when disrupted. This study investigated if KL deficiency affects BP and salt sensitivity using KL mutant heterozygous (+/-) mice and wild-type (WT) mice (9 weeks of age, 16 mice per group). Notably, systolic BP in KL(+/-) mice began to increase at the age of 15 weeks, reached a peak level at the age of 17 weeks, and remained elevated thereafter, whereas systolic BP remained consistent in WT mice. High salt (HS) intake further increased BP in KL(+/-) mice but did not affect BP in WT mice. Blockade of CC chemokine receptor 2 (CCR2), involved in monocyte chemotaxis, by a specific CCR2 antagonist (INCB3284) abolished the HS-induced increase in BP in KL(+/-) mice. Furthermore, HS loading substantially increased the expression of monocyte chemotactic protein-1 and the infiltration of macrophages and T cells in kidneys in KL(+/-) mice, and treatment with INCB3284 abolished these effects. Treatment of KL(+/-) mice with INCB3284 also attenuated the increased renal expressions of serum glucocorticoid-regulated kinase 1, thiazide-sensitive NaCl cotransporter, and ATP synthase β along with the renal structural damage and functional impairment induced by HS loading. In conclusion, KL deficiency caused salt-sensitive hypertension and renal damage by CCR2-mediated inflammation.
Collapse
Affiliation(s)
- Xiaoli Zhou
- Department of Cardiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kai Chen
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Han Lei
- Department of Cardiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
| | - Zhongjie Sun
- Department of Cardiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
29
|
Kobelt D, Aumann J, Schmidt M, Wittig B, Fichtner I, Behrens D, Lemm M, Freundt G, Schlag PM, Walther W. Preclinical study on combined chemo- and nonviral gene therapy for sensitization of melanoma using a human TNF-alpha expressing MIDGE DNA vector. Mol Oncol 2014; 8:609-19. [PMID: 24503218 DOI: 10.1016/j.molonc.2013.12.019] [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: 09/16/2013] [Revised: 12/09/2013] [Accepted: 12/30/2013] [Indexed: 12/26/2022] Open
Abstract
Nonviral gene therapy represents a realistic option for clinical application in cancer treatment. This preclinical study demonstrates the advantage of using the small-size MIDGE(®) DNA vector for improved transgene expression and therapeutic application. This is caused by significant increase in transcription efficiency, but not by increased intracellular vector copy numbers or gene transfer efficiency. We used the MIDGE-hTNF-alpha vector for high-level expression of hTNF-alpha in vitro and in vivo for a combined gene therapy and vindesine treatment in human melanoma models. The MIDGE vector mediated high-level hTNF-alpha expression leads to sensitization of melanoma cells towards vindesine. The increased efficacy of this combination is mediated by remarkable acceleration and increase of initiator caspase 8 and 9 and effector caspase 3 and 7 activation. In the therapeutic approach, the nonviral intratumoral in vivo jet-injection gene transfer of MIDGE-hTNF-alpha in combination with vindesine causes melanoma growth inhibition in association with increased apoptosis in A375 cell line or patient derived human melanoma xenotransplant (PDX) models. This study represents a proof-of-concept for an anticipated phase I clinical gene therapy trial, in which the MIDGE-hTNF-alpha vector will be used for efficient combined chemo- and nonviral gene therapy of malignant melanoma.
Collapse
Affiliation(s)
- Dennis Kobelt
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | - Jutta Aumann
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Experimental and Clinical Research Center, Charité University Medicine, Berlin, Germany
| | | | - Burghardt Wittig
- Foundation Institute Molecular Biology and Bioinformatics, Freie Universität Berlin, Berlin, Germany
| | - Iduna Fichtner
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Diana Behrens
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Margit Lemm
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Greta Freundt
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Experimental and Clinical Research Center, Charité University Medicine, Berlin, Germany
| | - Peter M Schlag
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Charité Comprehensive Cancer Center, Berlin, Germany
| | - Wolfgang Walther
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Experimental and Clinical Research Center, Charité University Medicine, Berlin, Germany.
| |
Collapse
|
30
|
Shulga N, Pastorino JG. Mitoneet mediates TNFα-induced necroptosis promoted by exposure to fructose and ethanol. J Cell Sci 2013; 127:896-907. [PMID: 24357718 DOI: 10.1242/jcs.140764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Fructose and ethanol are metabolized principally in the liver and are both known to contribute to the development of hepatic steatosis that can progress to hepatic steatohepatitis. The present study indentifies a synergistic interaction between fructose and ethanol in promoting hepatocyte sensitivity to TNFα-induced necroptosis. Concurrent exposure to fructose and ethanol induces the overexpression of the CDGSH iron-sulfur domain-containing protein 1 (CISD1 or mitoneet), which is localized to the outer mitochondrial membrane. The increased expression of mitoneet primes the hepatocyte for TNFα-induced cytotoxicity. Treatment with TNFα induces the translocation of a Stat3-Grim-19 complex to the mitochondria, which binds to mitoneet and promotes the rapid release of its 2Fe-2S cluster, causing an accumulation of mitochondrial iron. The dramatic increase of mitochondrial iron provokes a surge in formation of reactive oxygen species, resulting in mitochondrial injury and cell death. Additionally, mitoneet is constitutively expressed at high levels in L929 fibrosarcoma cells and is required for L929 cells to undergo TNFα-induced necroptosis in the presence of caspase inhibition, indicating the importance of mitoneet to the necroptotic form of cell death.
Collapse
Affiliation(s)
- Nataly Shulga
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | | |
Collapse
|
31
|
Bourgine PE, Pippenger BE, Todorov A, Tchang L, Martin I. Tissue decellularization by activation of programmed cell death. Biomaterials 2013; 34:6099-108. [DOI: 10.1016/j.biomaterials.2013.04.058] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 04/27/2013] [Indexed: 01/10/2023]
|
32
|
Erythropoietin protects epithelial cells from excessive autophagy and apoptosis in experimental neonatal necrotizing enterocolitis. PLoS One 2013; 8:e69620. [PMID: 23936061 PMCID: PMC3723879 DOI: 10.1371/journal.pone.0069620] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/12/2013] [Indexed: 12/17/2022] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of preterm infants. Increased intestinal epithelium permeability is an early event in NEC pathogenesis. Autophagy and apoptosis are induced by multiple stress pathways which may impact the intestinal barrier, and they have been associated with pathogenesis of diverse gastrointestinal diseases including inflammatory bowel disease. Using both in vitro and in vivo models, this study investigates autophagy and apoptosis under experimental NEC stresses. Furthermore this study evaluates the effect of erythropoietin (Epo), a component of breast milk previously shown to decrease the incidence of NEC and to preserve intestinal barrier function, on intestinal autophagy and apoptosis. It was found that autophagy and apoptosis are both rapidly up regulated in NEC in vivo as indicated by increased expression of the autophagy markers Beclin 1 and LC3II, and by evidence of apoptosis by TUNEL and cleaved caspase-3 staining. In the rat NEC experimental model, autophagy preceded the onset of apoptosis in intestine. In vitro studies suggested that Epo supplementation significantly decreased both autophagy and apoptosis via the Akt/mTOR signaling pathway and the MAPK/ERK pathway respectively. These results suggest that Epo protects intestinal epithelium from excessive autophagy and apoptosis in experimental NEC.
Collapse
|
33
|
Gumustekin M, Micili SC, Arici MA, Karaman M, Guneli ME, Tekmen I. The Effect of Insulin Treatment on Rac1 Expression in Diabetic Kidney. Ren Fail 2013; 35:396-402. [DOI: 10.3109/0886022x.2013.764256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
34
|
Talukder JR, Boyd B, Griffin A, Jaima A, Rajendran VM. Inflammatory cytokine TNF-α inhibits Na(+)-glutamine cotransport in intestinal epithelial cells. Can J Physiol Pharmacol 2012; 91:275-84. [PMID: 23627839 DOI: 10.1139/cjpp-2011-0488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glutamine (Gln), a preferred fuel source for enterocytes, is critical for intestinal epithelial cell integrity and barrier function. Chronic enteritis inhibits apical Na(+)-Gln cotransport. It is not known whether inflammatory cytokines that are secreted during inflammation inhibit Na(+)-Gln cotransport. Thus, this study aimed to examine whether TNF-α would affect apical Na(+)-Gln cotransport in intestinal epithelial cells. In this study, the presence of Na(+)-Gln cotransport was established by measuring Gln uptake in 10 days postconfluent IEC-6 cells grown on transwell plates. Cation, amino acid specificity, and siRNA transfection studies established that Na(+)-Gln cotransport is mediated via B(0)AT1. Immunoblotting and immunofluorescence studies established the apical membrane localization of B(0)AT1 in IEC-6 cells. Tumour necrosis factor α (TNF-α) inhibited Na(+)-Gln cotransport in a concentration- and time-dependent manner with an inhibitory concentration of 1.53 nmol·L(-1). Quantitative real-time PCR and Western blot analyses indicated that TNF-α did not alter B(0)AT1-specific transcripts or protein expression level. Kinetic studies revealed that TNF-α inhibited Na(+)-Gln cotransport by reducing the affinity of the cotransporters for Gln, and this effect was antagonized by genistein. Thus, we conclude that the TNF-α inhibition of Na(+)-Gln cotransport occurs at the post-translational level, and that the IEC-6 cell line is an excellent system to study the role of cytokines in Na(+)-Gln cotransport.
Collapse
|
35
|
Thomson ABR, Chopra A, Clandinin MT, Freeman H. Recent advances in small bowel diseases: Part I. World J Gastroenterol 2012; 18:3336-52. [PMID: 22807604 PMCID: PMC3396187 DOI: 10.3748/wjg.v18.i26.3336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 04/05/2012] [Accepted: 04/13/2012] [Indexed: 02/06/2023] Open
Abstract
As is the case in all parts of gastroenterology and hepatology, there have been many advances in our knowledge and understanding of small intestinal diseases. Over 1000 publications were reviewed for 2008 and 2009, and the important advances in basic science as well as clinical applications were considered. In Part I of this Editorial Review, seven topics are considered: intestinal development; proliferation and repair; intestinal permeability; microbiotica, infectious diarrhea and probiotics; diarrhea; salt and water absorption; necrotizing enterocolitis; and immunology/allergy. These topics were chosen because of their importance to the practicing physician.
Collapse
|
36
|
Rodrigues M, Turner O, Stolz D, Griffith LG, Wells A. Production of reactive oxygen species by multipotent stromal cells/mesenchymal stem cells upon exposure to fas ligand. Cell Transplant 2012; 21:2171-87. [PMID: 22526333 DOI: 10.3727/096368912x639035] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multipotent stromal cells (MSCs) can be differentiated into osteoblasts and chondrocytes, making these cells candidates to regenerate cranio-facial injuries and lesions in long bones. A major problem with cell replacement therapy, however, is the loss of transplanted MSCs at the site of graft. Reactive oxygen species (ROS) and nonspecific inflammation generated at the ischemic site have been hypothesized to lead to MSCs loss; studies in vitro show MSCs dying both in the presence of ROS or cytokines like FasL. We questioned whether MSCs themselves may be the source of these death inducers, specifically whether MSCs produce ROS under cytokine challenge. On treating MSCs with FasL, we observed increased ROS production within 2 h, leading to apoptotic death after 6 h of exposure to the cytokine. N-acetyl cysteine, an antioxidant, is able to protect MSCs from FasL-induced ROS production and subsequent ROS-dependent apoptosis, though the MSCs eventually succumb to ROS-independent death signaling. Epidermal growth factor (EGF), a cell survival factor, is able to protect cells from FasL-induced ROS production initially; however, the protective effect wanes with continued FasL exposure. In parallel, FasL induces upregulation of the uncoupling protein UCP2, the main uncoupling protein in MSCs, which is not abrogated by EGF; however, the production of ROS is followed by a delayed apoptotic cell death despite moderation by UCP2. FasL-induced ROS activates the stress-induced MAPK pathways JNK and p38MAPK as well as ERK, along with the activation of Bad, a proapoptotic protein, and suppression of survivin, an antiapoptotic protein; the latter two key modulators of the mitochondrial death pathway. FasL by itself also activates its canonical extrinsic death pathway noted by a time-dependent degradation of c-FLIP and activation of caspase 8. These data suggest that MSCs participate in their own demise due to nonspecific inflammation, holding implications for replacement therapies.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
37
|
Shibata S, Fujita T. Mineralocorticoid receptors in the pathophysiology of chronic kidney diseases and the metabolic syndrome. Mol Cell Endocrinol 2012; 350:273-80. [PMID: 21820485 DOI: 10.1016/j.mce.2011.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 06/29/2011] [Accepted: 07/09/2011] [Indexed: 01/19/2023]
Abstract
Recent studies indicate that aldosterone/mineralocorticoid receptor (MR) is a major contributor of chronic kidney disease (CKD) progression. Aldosterone/MR induces glomerular podocyte injury, causing the disruption of the glomerular filtration barrier and proteinuria. Conversely, MR antagonists substantially reduce proteinuria, which can be partly attributable to the protective effects on podocytes. Aldosterone excess, caused by adipocyte-derived aldosterone-releasing factors and other mechanisms, can be pathologically important in the renal complication of metabolic syndrome. A rat model of metabolic syndrome exhibits podocyte injury and proteinuria with serum aldosterone elevation, and the renal damage is prevented by MR blockade. Accumulating data also indicate that MR inhibition can confer renoprotection in a subgroup with low or normal aldosterone levels. We have recently identified the cross-talk between MR and small GTPase Rac1, providing one theoretical basis for the renoprotective effects of MR antagonists in non-high-aldosterone subjects. MR blockade can be a promising strategy for preventing CKD progression, and future clinical trials will conclusively determine the efficacy and tolerability of selective MR inhibition in CKD and metabolic syndrome.
Collapse
Affiliation(s)
- Shigeru Shibata
- Department of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | | |
Collapse
|
38
|
Apoptotic signaling in endothelial cells with neutrophil activation. Mol Cell Biochem 2011; 363:269-80. [DOI: 10.1007/s11010-011-1179-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 11/24/2011] [Indexed: 01/03/2023]
|
39
|
Overmeyer JH, Maltese WA. Death pathways triggered by activated Ras in cancer cells. Front Biosci (Landmark Ed) 2011; 16:1693-713. [PMID: 21196257 DOI: 10.2741/3814] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ras GTPases are best known for their ability to serve as molecular switches regulating cell growth, differentiation and survival. Gene mutations that result in expression of constitutively active forms of Ras have been linked to oncogenesis in animal models and humans. However, over the past two decades, evidence has gradually accumulated to support a paradoxical role for Ras proteins in the initiation of cell death pathways. In this review we survey the literature pointing to the ability of activated Ras to promote cell death under conditions where cancer cells encounter apoptotic stimuli or Ras is ectopically expressed. In some of these cases Ras acts through known effectors and well defined apoptotic death pathways. However, in other cases it appears that Ras operates by triggering novel non-apoptotic death mechanisms that are just beginning to be characterized. Understanding these mechanisms and the factors that go into changing the nature of Ras signaling from pro-survival to pro-death could set the stage for development of novel therapeutic approaches aimed at manipulating pro-death Ras signaling pathways in cancer.
Collapse
Affiliation(s)
- Jean H Overmeyer
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine, Toledo, Ohio 43614, USA
| | | |
Collapse
|
40
|
Chen YQ, Wang XX, Yao XM, Zhang DL, Yang XF, Tian SF, Wang NS. MicroRNA-195 promotes apoptosis in mouse podocytes via enhanced caspase activity driven by BCL2 insufficiency. Am J Nephrol 2011; 34:549-59. [PMID: 22123611 DOI: 10.1159/000333809] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 09/21/2011] [Indexed: 01/04/2023]
Abstract
BACKGROUND The apoptosis of podocytes is a characteristic event in diabetic nephropathy. The aim of this study was to investigate whether microRNAs (miRNAs) affect podocyte apoptosis in diabetic circumstances. METHODS Diabetic nephropathy was induced in DBA/2 mice by intraperitoneal injections of streptozotocin, and the levels of proteinuria were measured with ELISA. Apoptosis-related miRNAs were screened in isolated glomeruli. A conditionally immortalized mouse podocyte cell line was cultured in 25 mMD-glucose and either transfected with miRNA-195 (miR-195) mimics or inhibitors. The levels of BCL2 and caspase expression were determined using real-time RT-PCR and Western blot analysis, respectively. We also measured WT-1 and synaptopodin in podocytes. Apoptosis of podocytes was assessed with Hoechst 33258 nuclear staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and flow cytometry. RESULTS The expression of miR-195 was elevated in both diabetic mice with proteinuria and podocytes that were cultured in high glucose. Transfection with miR-195 reduced the protein levels of BCL2 and contributed to podocyte apoptosis via an increase in caspase-3. miR-195-treated podocytes underwent actin rearrangement and failed to synthesize sufficient levels of WT-1 and synaptopodin proteins, which suggests that the cells had suffered injuries similar to those observed in diabetic nephropathy in both humans and animal models. CONCLUSIONS Taken together, our findings demonstrate that miR-195 promotes apoptosis of podocytes under high-glucose conditions via enhanced caspase cascades for BCL2 insufficiency. This work thus presents a meaningful approach for deciphering mechanisms, by which miRNAs participate in diabetic renal injury.
Collapse
Affiliation(s)
- Yu-Qiang Chen
- Department of Nephrology and Rheumatology, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Potentiation of epithelial innate host responses by intercellular communication. PLoS Pathog 2010; 6:e1001194. [PMID: 21124989 PMCID: PMC2987820 DOI: 10.1371/journal.ppat.1001194] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 10/13/2010] [Indexed: 12/21/2022] Open
Abstract
The epithelium efficiently attracts immune cells upon infection despite the low number of pathogenic microbes and moderate levels of secreted chemokines per cell. Here we examined whether horizontal intercellular communication between cells may contribute to a coordinated response of the epithelium. Listeria monocytogenes infection, transfection, and microinjection of individual cells within a polarized intestinal epithelial cell layer were performed and activation was determined at the single cell level by fluorescence microscopy and flow cytometry. Surprisingly, chemokine production after L. monocytogenes infection was primarily observed in non-infected epithelial cells despite invasion-dependent cell activation. Whereas horizontal communication was independent of gap junction formation, cytokine secretion, ion fluxes, or nitric oxide synthesis, NADPH oxidase (Nox) 4-dependent oxygen radical formation was required and sufficient to induce indirect epithelial cell activation. This is the first report to describe epithelial cell-cell communication in response to innate immune activation. Epithelial communication facilitates a coordinated infectious host defence at the very early stage of microbial infection. All body surfaces are covered by a single layer of epithelial cells. Epithelial cells form a physical barrier to separate the underlying sterile tissue from the environment. In addition, epithelial cells actively sense bacterial and viral infection. The recognition of pathogenic microorganisms results in cell stimulation and the secretion of soluble mediators that attract professional immune cells to the site of infection. This first line host defence works very efficiently despite the often low number of pathogens and the limited amount of mediators secreted per epithelial cell. We therefore investigated whether infection of one individual epithelial cell would result in activation of other, non-infected cells within a confluent epithelial monolayer resulting in a more substantial host response. Indeed, using the model of the gut pathogen Listeria monocytogenes and monitoring infection and epithelial activation at a single cell level, we can clearly show that the epithelial response is mainly mediated by non-infected cells. Also, we identify oxygen radicals as potential mediators to facilitate horizontal epithelial communication upon immune stimulation. Our results thus provide a novel concept of a coordinated epithelial host response upon microbial infection facilitated by horizontal epithelial communication.
Collapse
|
42
|
Kilbey A, Terry A, Jenkins A, Borland G, Zhang Q, Wakelam MJO, Cameron ER, Neil JC. Runx regulation of sphingolipid metabolism and survival signaling. Cancer Res 2010; 70:5860-9. [PMID: 20587518 DOI: 10.1158/0008-5472.can-10-0726] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Runx genes (Runx1, 2, and 3) regulate cell fate in development and can operate as either oncogenes or tumor suppressors in cancer. The oncogenic potential of ectopic Runx expression has been shown in transgenic mice that develop lymphoma in potent synergy with overexpressed Myc, and in established fibroblasts that display altered morphology and increased tumorigenicity. Candidate oncogenic functions of overexpressed Runx genes include resistance to apoptosis in response to intrinsic and extrinsic stresses. In a search for gene targets responsible for this aspect of Runx phenotype, we have identified three key enzymes in sphingolipid metabolism (Sgpp1, Ugcg, and St3gal5/Siat9) as direct targets for Runx transcriptional regulation in a manner consistent with survival and apoptosis resistance. Consistent with these changes in gene expression, mass spectrometric analysis showed that ectopic Runx reduces intracellular long-chain ceramides in NIH3T3 fibroblasts and elevated extracellular sphingosine 1 phosphate. Runx expression also opposed the activation of c-Jun-NH(2)-kinase and p38(MAPK), key mediators of ceramide-induced death, and suppressed the onset of apoptosis in response to exogenous tumor necrosis factor alpha. The survival advantage conferred by ectopic Runx could be partially recapitulated by exogenous sphingosine 1 phosphate and was accompanied by reduced phosphorylation of p38(MAPK). These results reveal a novel link between transcription factor oncogenes and lipid signaling pathways involved in cancer cell survival and chemoresistance.
Collapse
Affiliation(s)
- Anna Kilbey
- Molecular Oncology Laboratory, Faculty of Veterinary Medicine, Institute of Comparative Medicine, University of Glasgow, Glasgow, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Obese persons with metabolic syndrome often have associated with salt-sensitive hypertension, microalbuminuria, and cardiac dysfunction, and the plasma aldosterone level in one-third of metabolic syndrome patients is clearly elevated. Hyperaldosteronism, which may be caused at least partially by certain adipocyte-derived factors, contributes to the development of proteinuria in obese hypertensive rats, and salt loading aggravates the proteinuria and induces cardiac diastolic dysfunction because of inadequate suppression of plasma aldosterone level. However, mineralocorticoid receptor (MR) antagonists prevent salt-induced renal and cardiac damage, suggesting that aldosterone excess and a high-salt diet exert an unfavorable synergistic action on the kidney and heart. In Dahl salt-sensitive rats, however, despite appropriate suppression of plasma aldosterone with a high-salt diet, salt loading paradoxically activated renal MR signaling, and the renal injury was markedly prevented by MR antagonists. Accordingly, we discovered an alternative pathway of MR activation in which Rac1, a small GTP-binding protein, activates MRs. Salt loading activates renal Rac1 in Dahl salt-sensitive rats, and Rac1 in turn induces MR activation, which results in renal injury, and the renal injury has been found to be prevented by Rac1 inhibitors. Moreover, several metabolic syndrome-related factors induce Rac1 activation, and one of them, hyperglycemia, activates MRs via Rac1 activation. Consistent with this, Rac1 inhibitors attenuated the proteinuria and renal injury in obese hypertensive animals. Thus, both salt and obesity activate Rac1 and cause MR activation. Abnormal activation of the aldosterone/MR pathway plays a key role in the development of salt-sensitive hypertension and renal injury in metabolic syndrome.
Collapse
Affiliation(s)
- Toshiro Fujita
- From the Department of Nephrology and Endocrinology, School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
44
|
Zhang QG, Wang R, Han D, Brann DW. Role of Rac1 GTPase in JNK signaling and delayed neuronal cell death following global cerebral ischemia. Brain Res 2009; 1265:138-47. [PMID: 19368836 PMCID: PMC3801190 DOI: 10.1016/j.brainres.2009.01.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 01/14/2009] [Accepted: 01/18/2009] [Indexed: 12/31/2022]
Abstract
The overall goal of this study was to determine the role of Rac1 in POSH/MLK/JNK signaling and delayed neuronal cell death following cerebral ischemia. Temporal studies revealed that Rac1 GTPase activation was significantly elevated in hippocampus CA1 at 10 min to 72 h after cerebral ischemia reperfusion, with peak levels 30 min to 6 h after reperfusion. Total Rac1 protein levels were not significantly changed following cerebral ischemia. Rac1 has been shown to interact with POSH (plenty of SH3s), a scaffold protein that binds to and regulates MLK3 and JNK activation. Co-immunoprecipitation (Co-IP) studies revealed that POSH-Rac1-MLK3 complex formation displayed a significant and prolonged elevation after reperfusion, with a correlative increase in phosphorylation/activation of MLK3 as compared to sham controls. Intracerebroventricular administration of Rac1 antisense oligonucleotides (AS-ODNs) significantly attenuated Rac1 levels and Rac1 activation at 30 min after reperfusion, with a correlated significant attenuation of POSH-MLK3-Rac1 complex formation and MLK3 activation in hippocampus CA1. Infusion of Rac1 AS-ODNs also significantly attenuated post-ischemic activation of JNK, downstream of MLK3, and strongly protected the hippocampus CA1 from ischemic damage. Missense oligos had no effect on any of the parameters measured. The Rac1 AS-ODNs results were further confirmed by administration of a Rac1 inhibitor (NSC23766), which markedly attenuated activation of Rac1 and JNK, and significantly attenuated apoptotic delayed neuronal cell death following cerebral ischemia. As a whole, these studies demonstrate an important role for Rac1 in activation of the prodeath MLK3-JNK kinase signaling pathway and delayed neuronal cell death following cerebral ischemia.
Collapse
Affiliation(s)
- Quan-Guang Zhang
- Developmental Neurobiology Program, Institute of Molecular Medicine and Genetics, and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| | - Ruimin Wang
- Research Center for Molecular Biology, North China Coal Medical University, Tangshan 063000, China
| | - Dong Han
- Developmental Neurobiology Program, Institute of Molecular Medicine and Genetics, and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| | - Darrell W. Brann
- Developmental Neurobiology Program, Institute of Molecular Medicine and Genetics, and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| |
Collapse
|
45
|
Susnow N, Zeng L, Margineantu D, Hockenbery DM. Bcl-2 family proteins as regulators of oxidative stress. Semin Cancer Biol 2008. [PMID: 19138742 DOI: 10.1016/j.semcancer.2008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Bcl-2 family of proteins includes pro- and anti-apoptotic factors acting at mitochondrial and microsomal membranes. An impressive body of published studies, using genetic and physical reconstitution experiments in model organisms and cell lines, supports a view of Bcl-2 proteins as the critical arbiters of apoptotic cell death decisions in most circumstances (excepting CD95 death receptor signaling in Type I cells). Evasion of apoptosis is one of the hallmarks of cancer [Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70], relevant to tumorigenesis as well as resistance to cytotoxic drugs, and deregulation of Bcl-2 proteins is observed in many cancers [Manion MK, Hockenbery DM. Targeting BCL-2-related proteins in cancer therapy. Cancer Biol Ther. 2003;2:S105-14; Olejniczak ET, Van Sant C, Anderson MG, Wang G, Tahir SK, Sauter G, et al. Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res. 2007;5:331-9]. The rekindled interest in aerobic glycolysis as a cancer trait raises interesting questions as to how metabolic changes in cancer cells are integrated with other essential alterations in cancer, e.g. promotion of angiogenesis and unbridled growth signals. Apoptosis induced by multiple different signals involves loss of mitochondrial homeostasis, in particular, outer mitochondrial membrane integrity, releasing cytochrome c and other proteins from the intermembrane space. This integrative process, controlled by Bcl-2 family proteins, is also influenced by the metabolic state of the cell. In this review, we consider the role of reactive oxygen species, a metabolic by-product, in the mitochondrial pathway of apoptosis, and the relationships between Bcl-2 functions and oxidative stress.
Collapse
Affiliation(s)
- Nathan Susnow
- Department of Medicine, University of Washington, Seattle, 98195-6424, United States
| | | | | | | |
Collapse
|
46
|
Bcl-2 family proteins as regulators of oxidative stress. Semin Cancer Biol 2008; 19:42-9. [PMID: 19138742 DOI: 10.1016/j.semcancer.2008.12.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 12/13/2008] [Indexed: 12/22/2022]
Abstract
The Bcl-2 family of proteins includes pro- and anti-apoptotic factors acting at mitochondrial and microsomal membranes. An impressive body of published studies, using genetic and physical reconstitution experiments in model organisms and cell lines, supports a view of Bcl-2 proteins as the critical arbiters of apoptotic cell death decisions in most circumstances (excepting CD95 death receptor signaling in Type I cells). Evasion of apoptosis is one of the hallmarks of cancer [Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70], relevant to tumorigenesis as well as resistance to cytotoxic drugs, and deregulation of Bcl-2 proteins is observed in many cancers [Manion MK, Hockenbery DM. Targeting BCL-2-related proteins in cancer therapy. Cancer Biol Ther. 2003;2:S105-14; Olejniczak ET, Van Sant C, Anderson MG, Wang G, Tahir SK, Sauter G, et al. Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res. 2007;5:331-9]. The rekindled interest in aerobic glycolysis as a cancer trait raises interesting questions as to how metabolic changes in cancer cells are integrated with other essential alterations in cancer, e.g. promotion of angiogenesis and unbridled growth signals. Apoptosis induced by multiple different signals involves loss of mitochondrial homeostasis, in particular, outer mitochondrial membrane integrity, releasing cytochrome c and other proteins from the intermembrane space. This integrative process, controlled by Bcl-2 family proteins, is also influenced by the metabolic state of the cell. In this review, we consider the role of reactive oxygen species, a metabolic by-product, in the mitochondrial pathway of apoptosis, and the relationships between Bcl-2 functions and oxidative stress.
Collapse
|
47
|
Yan Y, Dalmasso G, Nguyen HTT, Obertone TS, Charrier-Hisamuddin L, Sitaraman SV, Merlin D. Nuclear factor-kappaB is a critical mediator of Ste20-like proline-/alanine-rich kinase regulation in intestinal inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1013-28. [PMID: 18787102 PMCID: PMC2543070 DOI: 10.2353/ajpath.2008.080339] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/11/2008] [Indexed: 01/04/2023]
Abstract
Inflammatory bowel disease (IBD) is thought to result from commensal flora, aberrant cellular stress, and genetic factors. Here we show that the expression of colonic Ste20-like proline-/alanine-rich kinase (SPAK) that lacks a PAPA box and an F-alpha helix loop is increased in patients with IBD. The same effects were observed in a mouse model of dextran sodium sulfate-induced colitis and in Caco2-BBE cells treated with the pro-inflammatory cytokine tumor necrosis factor (TNF)-alpha. The 5'-flanking region of the SPAK gene contains two transcriptional start sites, three transcription factor Sp1-binding sites, and one transcription factor nuclear factor (NF)-kappaB-binding site, but no TATA elements. The NF-kappaB-binding site was essential for stimulated SPAK promoter activity by TNF-alpha, whereas the Sp1-binding sites were important for basal promoter activity. siRNA-induced knockdown of NF-kappaB, but not of Sp1, reduced TNF-alpha-induced SPAK expression. Nuclear run-on and mRNA decay assays demonstrated that TNF-alpha directly increased SPAK mRNA transcription without affecting SPAK mRNA stability. Furthermore, up-regulation of NF-kappaB expression and demethylation of the CpG islands induced by TNF-alpha also played roles in the up-regulation of SPAK expression. In conclusion, our data indicate that during inflammatory conditions, TNF-alpha is a key regulator of SPAK expression. The development of compounds that can either modulate or disrupt the activity of SPAK-mediated pathways is therefore important for the control and attenuation of downstream pathological responses, particularly in IBD.
Collapse
Affiliation(s)
- Yutao Yan
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|