Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Ann Clin Transl Neurol ; 9(1): 50-66, 2022 01.
Article in English | MEDLINE | ID: mdl-35014217

ABSTRACT

OBJECTIVE: Dual leucine zipper kinase (DLK), which regulates the c-Jun N-terminal kinase pathway involved in axon degeneration and apoptosis following neuronal injury, is a potential therapeutic target in amyotrophic lateral sclerosis (ALS). This first-in-human study investigated safety, tolerability, and pharmacokinetics (PK) of oral GDC-0134, a small-molecule DLK inhibitor. Plasma neurofilament light chain (NFL) levels were explored in GDC-0134-treated ALS patients and DLK conditional knockout (cKO) mice. METHODS: The study included placebo-controlled, single and multiple ascending-dose (SAD; MAD) stages, and an open-label safety expansion (OLE) with adaptive dosing for up to 48 weeks. RESULTS: Forty-nine patients were enrolled. GDC-0134 (up to 1200 mg daily) was well tolerated in the SAD and MAD stages, with no serious adverse events (SAEs). In the OLE, three study drug-related SAEs occurred: thrombocytopenia, dysesthesia (both Grade 3), and optic ischemic neuropathy (Grade 4); Grade ≤2 sensory neurological AEs led to dose reductions/discontinuations. GDC-0134 exposure was dose-proportional (median half-life = 84 h). Patients showed GDC-0134 exposure-dependent plasma NFL elevations; DLK cKO mice also exhibited plasma NFL compared to wild-type littermates. INTERPRETATION: This trial characterized GDC-0134 safety and PK, but no adequately tolerated dose was identified. NFL elevations in GDC-0134-treated patients and DLK cKO mice raised questions about interpretation of biomarkers affected by both disease and on-target drug effects. The safety profile of GDC-0134 was considered unacceptable and led to discontinuation of further drug development for ALS. Further work is necessary to understand relationships between neuroprotective and potentially therapeutic effects of DLK knockout/inhibition and NFL changes in patients with ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neurofilament Proteins/blood , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Adult , Aged , Amyotrophic Lateral Sclerosis/blood , Animals , Biomarkers/blood , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , MAP Kinase Kinase Kinases/deficiency , Male , Mice , Mice, Knockout , Middle Aged , Outcome Assessment, Health Care , Protein Kinase Inhibitors/pharmacokinetics
2.
Front Immunol ; 12: 738490, 2021.
Article in English | MEDLINE | ID: mdl-34691044

ABSTRACT

Tumor progression locus 2 (Tpl2) is a serine-threonine kinase known to promote inflammation in response to various pathogen-associated molecular patterns (PAMPs), inflammatory cytokines and G-protein-coupled receptors and consequently aids in host resistance to pathogens. We have recently shown that Tpl2-/- mice succumb to infection with a low-pathogenicity strain of influenza (x31, H3N2) by an unknown mechanism. In this study, we sought to characterize the cytokine and immune cell profile of influenza-infected Tpl2-/- mice to gain insight into its host protective effects. Although Tpl2-/- mice display modestly impaired viral control, no virus was observed in the lungs of Tpl2-/- mice on the day of peak morbidity and mortality suggesting that morbidity is not due to virus cytopathic effects but rather to an overactive antiviral immune response. Indeed, increased levels of interferon-ß (IFN-ß), the IFN-inducible monocyte chemoattractant protein-1 (MCP-1, CCL2), Macrophage inflammatory protein 1 alpha (MIP-1α; CCL3), MIP-1ß (CCL4), RANTES (CCL5), IP-10 (CXCL10) and Interferon-γ (IFN-γ) was observed in the lungs of influenza-infected Tpl2-/- mice at 7 days post infection (dpi). Elevated cytokine and chemokines were accompanied by increased infiltration of the lungs with inflammatory monocytes and neutrophils. Additionally, we noted that increased IFN-ß correlated with increased CCL2, CXCL1 and nitric oxide synthase (NOS2) expression in the lungs, which has been associated with severe influenza infections. Bone marrow chimeras with Tpl2 ablation localized to radioresistant cells confirmed that Tpl2 functions, at least in part, within radioresistant cells to limit pro-inflammatory response to viral infection. Collectively, this study suggests that Tpl2 tempers inflammation during influenza infection by constraining the production of interferons and chemokines which are known to promote the recruitment of detrimental inflammatory monocytes and neutrophils.


Subject(s)
Cytokine Release Syndrome/metabolism , Cytokines/blood , Influenza A Virus, H3N2 Subtype/pathogenicity , Lung/metabolism , MAP Kinase Kinase Kinases/deficiency , Monocytes/metabolism , Neutrophils/metabolism , Orthomyxoviridae Infections/metabolism , Proto-Oncogene Proteins/deficiency , Animals , Biomarkers/blood , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/genetics , Disease Models, Animal , Female , Host-Pathogen Interactions , Influenza A Virus, H3N2 Subtype/immunology , Lung/immunology , Lung/virology , MAP Kinase Kinase Kinases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/virology , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/virology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Proto-Oncogene Proteins/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Time Factors
3.
Toxicol Appl Pharmacol ; 418: 115494, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33722668

ABSTRACT

Tumor progression locus 2 (Tpl2, gene name MAP3K8), a mitogen-activated protein kinase, is widely expressed in immune and non-immune cells to integrate tumor necrosis factor (TNF), toll-like receptors (TLRs), and interleukin-1 (IL1) receptor signaling to regulate inflammatory response. Given its central role in inflammatory response, Tpl2 is an attractive small molecule drug target. However, the role of Tpl2 as an oncogene or tumor suppressor gene remains controversial, and its function outside immune cells is not understood. We therefore utilized a Tpl2 kinase dead (Tpl2-KD) mouse model in an 18-month aging study to further elucidate Tpl2 effects on lifespan and chronic disease. Histopathological studies revealed the incidence and severity of spontaneous tumors and non-neoplastic lesions were comparable between wild type and Tpl2-KD mice. The only finding was that male Tpl2-KD mice had higher bodyweight and an increased incidence of liver steatosis, suggesting a sex-specific role for Tpl2 in hepatic lipid metabolism. In conclusion, loss of Tpl2 kinase activity did not lead to increased tumorigenesis over aging in mice but affected likely alterations in lipid metabolism in male animals.


Subject(s)
Fatty Liver/enzymology , Inflammation/enzymology , Liver/enzymology , MAP Kinase Kinase Kinases/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Age Factors , Animals , Fatty Liver/genetics , Fatty Liver/pathology , Female , Genotype , Inflammation/genetics , Lipid Metabolism , Liver/pathology , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Sex Factors
4.
Cell ; 182(2): 404-416.e14, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610081

ABSTRACT

Problems arising during translation of mRNAs lead to ribosome stalling and collisions that trigger a series of quality control events. However, the global cellular response to ribosome collisions has not been explored. Here, we uncover a function for ribosome collisions in signal transduction. Using translation elongation inhibitors and general cellular stress conditions, including amino acid starvation and UV irradiation, we show that ribosome collisions activate the stress-activated protein kinase (SAPK) and GCN2-mediated stress response pathways. We show that the MAPKKK ZAK functions as the sentinel for ribosome collisions and is required for immediate early activation of both SAPK (p38/JNK) and GCN2 signaling pathways. Selective ribosome profiling and biochemistry demonstrate that although ZAK generally associates with elongating ribosomes on polysomal mRNAs, it specifically auto-phosphorylates on the minimal unit of colliding ribosomes, the disome. Together, these results provide molecular insights into how perturbation of translational homeostasis regulates cell fate.


Subject(s)
Ribosomes/metabolism , Stress, Physiological , ATP-Binding Cassette Transporters/metabolism , Anisomycin/pharmacology , Apoptosis/drug effects , DNA Damage/radiation effects , Enzyme Activation , Humans , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Phosphorylation , Polyribosomes/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Ultraviolet Rays , eIF-2 Kinase/metabolism
5.
Trends Immunol ; 40(9): 799-808, 2019 09.
Article in English | MEDLINE | ID: mdl-31401161

ABSTRACT

Mammalian TPL-2 kinase (MAP3K8) mediates Toll-like receptor activation of ERK1/2 and p38α MAP kinases and is critical for regulating immune responses to pathogens. TPL-2 also has an important adaptor function, maintaining stability of associated ABIN-2 ubiquitin-binding protein. Consequently, phenotypes detected in Map3k8-/- mice can be caused by lack of TPL-2, ABIN-2, or both proteins. Recent studies show that increased inflammation of Map3k8-/- mice in allergic airway inflammation and colitis results from reduced ABIN-2 signaling, rather than blocked TPL-2 signaling. However, Map3k8-/- mice have been employed extensively to evaluate the potential of TPL-2 as an anti-inflammatory drug target. We posit that Map3k8D270A/D270A mice, expressing catalytically inactive TPL-2 and physiologic ABIN-2, should be used to evaluate the potential effects of TPL-2 inhibitors in disease.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Inflammation/immunology , MAP Kinase Kinase Kinases/immunology , Proto-Oncogene Proteins/immunology , Signal Transduction/immunology , Animals , Humans , MAP Kinase Kinase Kinases/deficiency , Mice , Mice, Knockout , Proto-Oncogene Proteins/deficiency
6.
Clin Exp Pharmacol Physiol ; 46(9): 854-860, 2019 09.
Article in English | MEDLINE | ID: mdl-31192472

ABSTRACT

Mixed-lineage kinase 3 (MLK3), the mitogen-activated protein kinase kinase kinase (MAP3K), has been recognized as a player in tumorigenesis and oncogenic signalling, yet its detailed functions and signalling in cervical cancer have not been fully elucidated. Here, we identify that cervical cancer cells display higher mRNA and protein levels of MLK3 than normal cervical epithelial squamous cells. In HeLa and SiHa cell, MLK3 knockdown using siRNA remarkably suppressed cell survival and promoted cell apoptosis, with increased expression of the apoptosis-related protein Bax and reduced Bcl-2. Moreover, MLK3 knockdown promoted cell autophagy, demonstrated by increased ratio of autophagy-related proteins LC3II/LC3I and decreased p62 expression in MLK3 depletion cells. Furthermore, MLK3 knockdown remarkably abolished Notch-1 expression in cervical cancer cells. By co-treating Hela cells with MLK3 specific siRNA and pcDNA3.1-Notch-1 overexpression plasmid or autophagy inhibitor 3-MA, we found that MLK3 played its role in cervical cancer cells via the Notch-1/autophagy network. Our results demonstrate the importance of MLK3 in cervical cancer progression via modulating the Notch-1/autophagy network, and suggest that MLK3 is a promising therapeutic target for cervical cancer.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Receptor, Notch1/metabolism , Uterine Cervical Neoplasms/pathology , Cell Proliferation/genetics , Female , HeLa Cells , Humans , Signal Transduction/genetics , Mitogen-Activated Protein Kinase Kinase Kinase 11
7.
Stem Cells ; 37(6): 766-778, 2019 06.
Article in English | MEDLINE | ID: mdl-30786091

ABSTRACT

Aberrant wound healing presents as inappropriate or insufficient tissue formation. Using a model of musculoskeletal injury, we demonstrate that loss of transforming growth factor-ß activated kinase 1 (TAK1) signaling reduces inappropriate tissue formation (heterotopic ossification) through reduced cellular differentiation. Upon identifying increased proliferation with loss of TAK1 signaling, we considered a regenerative approach to address insufficient tissue production through coordinated inactivation of TAK1 to promote cellular proliferation, followed by reactivation to elicit differentiation and extracellular matrix production. Although the current regenerative medicine paradigm is centered on the effects of drug treatment ("drug on"), the impact of drug withdrawal ("drug off") implicit in these regimens is unknown. Because current TAK1 inhibitors are unable to phenocopy genetic Tak1 loss, we introduce the dual-inducible COmbinational Sequential Inversion ENgineering (COSIEN) mouse model. The COSIEN mouse model, which allows us to study the response to targeted drug treatment ("drug on") and subsequent withdrawal ("drug off") through genetic modification, was used here to inactivate and reactivate Tak1 with the purpose of augmenting tissue regeneration in a calvarial defect model. Our study reveals the importance of both the "drug on" (Cre-mediated inactivation) and "drug off" (Flp-mediated reactivation) states during regenerative therapy using a mouse model with broad utility to study targeted therapies for disease. Stem Cells 2019;37:766-778.


Subject(s)
Bone Regeneration/drug effects , Fractures, Bone/genetics , MAP Kinase Kinase Kinases/genetics , Mesenchymal Stem Cells/enzymology , Osteoblasts/enzymology , Wound Healing/genetics , Animals , Bone Regeneration/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Female , Founder Effect , Fractures, Bone/drug therapy , Fractures, Bone/enzymology , Fractures, Bone/pathology , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/deficiency , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Skull/drug effects , Skull/injuries , Skull/metabolism , Wound Healing/drug effects
8.
Br J Pharmacol ; 175(22): 4239-4252, 2018 11.
Article in English | MEDLINE | ID: mdl-30153328

ABSTRACT

BACKGROUND AND PURPOSE: Angiotensin-converting enzyme (ACE), an important part of the renin-angiotensin system, is implicated in stimulating the fibrotic processes in the heart, lung, liver and kidney, while an ACE inhibitor (ACEI) promotes physiological tissue repair in these organs. The mechanism is closely related to TGF-ß1 pathways. However, the reported effects of applying ACEIs during scar formation are unclear. Hence, we explored the anti-fibrotic effects of an ACEI and the molecular mechanisms involved in a mouse scar model. EXPERIMENTAL APPROACH: After a full-thickness skin wound operation, ACE wild-type mice were randomly assigned to receive either ramipril, losartan or hydralazine p.o. ACE knockout (KO) mice and negative control mice only received vehicle (water). Wound/scar widths during wound healing and histological examinations were recorded at the final day. The ability of ACEI to reduce fibrosis via TGF-ß1 signalling was evaluated in vitro and in vivo. KEY RESULTS: ACE KO mice and mice that received ramipril showed narrower wound/scar width, reduced fibroblast proliferation, decreased collagen and TGF-ß1 expression. ACEI attenuated the phosphorylation of small mothers against decapentaplegic (Smad2/3) and TGF-ß-activated kinase 1 (TAK1) both in vitro and in vivo. The expression of ACE-related peptides varied in murine models with different drug treatments. CONCLUSIONS AND IMPLICATIONS: ACEI showed anti-fibrotic properties in scar formation by mediating downstream peptides to suppress TGF-ß1/Smad and TGF-ß1/TAK1 pathways. These findings suggest that dual inhibition of Smad and TAK1 signalling by ACEI is a useful strategy for the development of new anti-fibrotic agents.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Animals , Disease Models, Animal , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Knockout , Smad Proteins/metabolism
9.
Elife ; 72018 07 03.
Article in English | MEDLINE | ID: mdl-29968565

ABSTRACT

Neuropathic pain resulting from nerve injury can become persistent and difficult to treat but the molecular signaling responsible for its development remains poorly described. Here, we identify the neuronal stress sensor dual leucine zipper kinase (DLK; Map3k12) as a key molecule controlling the maladaptive pathways that lead to pain following injury. Genetic or pharmacological inhibition of DLK reduces mechanical hypersensitivity in a mouse model of neuropathic pain. Furthermore, DLK inhibition also prevents the spinal cord microgliosis that results from nerve injury and arises distant from the injury site. These striking phenotypes result from the control by DLK of a transcriptional program in somatosensory neurons regulating the expression of numerous genes implicated in pain pathogenesis, including the immune gene Csf1. Thus, activation of DLK is an early event, or even the master regulator, controlling a wide variety of pathways downstream of nerve injury that ultimately lead to chronic pain.


Subject(s)
Gliosis/genetics , Hyperalgesia/genetics , MAP Kinase Kinase Kinases/genetics , Neuralgia/genetics , Peripheral Nerve Injuries/genetics , Sensory Receptor Cells/enzymology , Animals , Disease Models, Animal , Female , Gene Expression Regulation , Gliosis/enzymology , Gliosis/pathology , Gliosis/prevention & control , Hyperalgesia/enzymology , Hyperalgesia/pathology , Hyperalgesia/prevention & control , MAP Kinase Kinase Kinases/deficiency , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Transgenic , Microglia/enzymology , Microglia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/enzymology , Neuralgia/pathology , Neuralgia/prevention & control , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Sciatic Nerve/enzymology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sensory Receptor Cells/pathology , Signal Transduction , Spinal Cord/enzymology , Spinal Cord/pathology , Touch , Transcription, Genetic
10.
Sci Rep ; 8(1): 8475, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855498

ABSTRACT

Bone morphogenetic proteins (BMPs) activate the canonical Smad1/5/8 and non-canonical Tak1-MAPK pathways via BMP receptors I and II to regulate skeletal development and bone remodeling. Specific ablation of Bmpr1a in immature osteoblasts, osteoblasts, or osteocytes results in an increase in cancellous bone mass, yet opposite results have been reported regarding the underlying mechanisms. Moreover, the role for BMPRIA-mediated signaling in bone marrow mesenchymal stromal cells (BM-MSCs) has not been explored. Here, we specifically ablated Bmpr1a in BM-MSCs in adult mice to study the function of BMPR1A in bone remodeling and found that the mutant mice showed an increase in cancellous and cortical bone mass, which was accompanied by a decrease in bone formation rate and a greater decrease in bone resorption. Decreased bone formation was associated with a defect in BM-MSC osteogenic differentiation whereas decreased bone resorption was associated with a decrease in RANKL production and osteoclastogenesis. However, ablation of Tak1, a critical non-canonical signaling molecule downstream of BMP receptors, in BM-MSCs at adult stage did not affect bone remodeling. These results suggest that BMP signaling through BMPRIA controls BM-MSC osteogenic differentiation/bone formation and RANKL expression/osteoclastogenesis in adult mice independent of Tak1 signaling.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation , RANK Ligand/metabolism , Animals , Bone Marrow Cells/cytology , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Lineage , Homeodomain Proteins/metabolism , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , RANK Ligand/genetics , Signal Transduction
11.
J Neuroinflammation ; 15(1): 148, 2018 May 17.
Article in English | MEDLINE | ID: mdl-29776451

ABSTRACT

BACKGROUND: Activation of transforming growth factor-ß-activated kinase 1 (TAK1) occurs after stroke and leads to an exacerbation of brain injury. TAK1 is involved in innate and adaptive immune responses, but it has divergent inflammatory effects that are dependent on the cell type in which it is activated. There is a robust infiltration of myeloid cells after stroke; however, the contribution of myeloid TAK1 to cerebral ischemia is currently unknown. We hypothesized that myeloid-specific deletion of TAK1 would protect against ischemic brain injury. METHODS: Myeloid TAK1ΔM and wild-type (WT) mice were subjected to middle cerebral artery occlusion (MCAo). Brain-infiltrating and splenic immune cells were evaluated at 3 days after stroke. Assessment of infarct size and behavioral deficits were performed on days 3 and 7 post-stroke. RESULTS: Infarcts were significantly smaller in TAK1ΔM mice (p < 0.01), and behavioral deficits were less severe despite equivalent reduction in cerebral blood flow. Flow cytometry demonstrated an increase in the frequency of splenic monocytes and neutrophils (p < 0.05) and a decrease in splenic CD3+ T (p < 0.01) and CD19+ B (p = 0.06) cells in TAK1ΔM mice compared to WT at baseline. Three days after stroke, a significant increase in the number of brain-infiltrating immune cell was observed in both TAK1ΔM (p < 0.05) and WT (p < 0.001) mice compared to their respective shams. However, there was a significant decrease in the infiltrating CD45hi immune cell counts (p < 0.05), with a pronounced reduction in infiltrating monocytes (p < 0.001) in TAK1ΔM after stroke compared to WT stroke mice. Additionally, a significant reduction in CD49d+ monocytes was seen in the brains of TAK1ΔM stroke mice compared to wild-type mice. Importantly, TAK1ΔM MCAo mice had smaller infarcts and improved behavioral outcomes at day 7 post-stroke. CONCLUSION: Our results showed that deletion of myeloid TAK1 resulted in smaller infarcts and improved functional outcomes at the peak of inflammation (day 3) and a reduction in brain-infiltrating immune cells that were primarily monocytes. Myeloid TAK1 deletion was also protective at 7 days post MCAo, reflecting a detrimental role of myeloid TAK1 in the progression of ischemic injury.


Subject(s)
Infarction, Middle Cerebral Artery/pathology , MAP Kinase Kinase Kinases/deficiency , Monocytes/pathology , Myeloid Cells/physiology , Neutrophils/physiology , Recovery of Function/genetics , Animals , Antigens, CD/metabolism , Cerebrovascular Circulation/genetics , Disease Models, Animal , Flow Cytometry , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/physiopathology , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Muramidase/genetics , Muramidase/metabolism , Neutrophil Infiltration/genetics
12.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29844241

ABSTRACT

Tumor progression locus 2 (TPL2), a serine/threonine protein kinase, is a major inflammatory mediator in immune cells. The predominant inflammatory actions of TPL2 depend on the activation of mitogen-activated protein kinases (MAPK) and the upregulated production of the cytokines tumor necrosis factor alpha (TNF-α) and interleukin 1ß (IL-1ß) in macrophages and dendritic cells in response to lipopolysaccharide (LPS). Significant increases in TNF-α, IL-6, IL-ß, and IL-8 levels in patients with Clostridium difficile infection (CDI) have been reported. Both TNF-α and IL-6 have been postulated to play key roles in the systemic inflammatory response in CDI, and IL-8 is essential for the development of local intestinal inflammatory responses in CDI. The objective of this study was to elucidate the role of TPL2 in the pathogenesis of CDI. We found that TPL2 was significantly activated in human and mouse intestinal tissues upon C. difficile toxin exposure or CDI. We further demonstrated that TPL2 knockout (TPL2-KO) mice were significantly more resistant to CDI than wild-type mice, with significantly reduced production of TNF-α, IL-6, IL-1ß, KC (a mouse homologue of IL-8), and myeloperoxidase (MPO) in the ceca and colons of TPL2-KO mice. Finally, we found that TPL2 inhibition by a specific inhibitor or TPL2 gene ablation significantly reduced TcdB-induced production of TNF-α, IL-6, IL-ß, and KC by inhibiting the activation of p38, extracellular signal-regulated kinase (ERK), and c-Jun NH2-terminal kinase (JNK). Taken together, our data suggest that TPL2 represents a potential therapeutic target for CDI treatment.


Subject(s)
Clostridium Infections/pathology , Inflammation/pathology , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Blotting, Western , Cecum/pathology , Colon/pathology , Cytokines/analysis , Disease Susceptibility , Humans , MAP Kinase Kinase Kinases/deficiency , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/analysis , Proto-Oncogene Proteins/deficiency , Signal Transduction
13.
Am J Physiol Renal Physiol ; 315(2): F336-F344, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29667914

ABSTRACT

Transforming growth factor-ß (TGFß) plays a central role in renal scarring, controlling extracellular matrix deposition by interstitial cells and mesangial cells. TGFß signals through Smad and mitogen-activated protein kinase (MAPK) pathways. To understand the role of MAPK in interstitial and mesangial cells, we genetically inactivated TGFß-activated kinase-1 ( Map3k7) using Foxd1+/cre. Embryonic kidney development was unperturbed in mutants, but spontaneous scarring of the kidney ensued during the first postnatal week, with retention of embryonic nephrogenic rests and accumulation of collagen IV in the mesangium. MAPK signaling in the mesangium of mutant mice was skewed, with depressed p38 but elevated c-Jun NH2-terminal kinase (JNK) activation at postnatal day 3. Despite normal expression of platelet-derived growth factor receptor-ß (PDGFRß) in the mesangium of mutants at birth, expression was lost concomitantly with the increase in JNK activation, and studies in isolated mesangial cells revealed that JNK negatively regulates Pdgfrß. In summary, we show that MAP3K7 balances MAPK signaling in mesangial cells, suppressing postnatal JNK activation. We propose that the balance of MAPK signaling is essential for appropriate postnatal regulation of mesangial PDGFRß expression.


Subject(s)
Forkhead Transcription Factors/metabolism , Glomerulonephritis/enzymology , MAP Kinase Kinase Kinases/metabolism , Mesangial Cells/enzymology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Cells, Cultured , Collagen Type IV/metabolism , Disease Models, Animal , Enzyme Activation , Fibrosis , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Silencing , Genetic Predisposition to Disease , Glomerulonephritis/genetics , Glomerulonephritis/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Mesangial Cells/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Int J Cancer ; 142(1): 81-91, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28875549

ABSTRACT

Hepatocyte death is associated with liver inflammation, fibrosis and hepatocellular carcinoma (HCC). Damaged cells trigger inflammation through activation of Toll-like receptors (TLRs). Although the role of TLR4 in HCC development has been reported, the role of TLR9 in the development of HCC remains elusive. To investigate the role of TLR4 and TLR9 signaling in liver inflammation-fibrosis-cancer axis, we took advantage of mice with hepatic deletion of transforming growth factor-ß-activated kinase 1 (Tak1ΔHep) that develop spontaneous liver injury, inflammation, fibrosis, and HCC, recapitulating the pathology of human HCC. We generated double knockout mice lacking genes of our interest with hepatic Tak1. Tak1ΔHep mice and Tlr4-deficient Tak1ΔHep mice had similar serum ALT levels, but Tlr4-deficient Tak1ΔHep mice exhibited significantly reduced macrophage infiltration, myofibroblast activation and tumor formation. Ablation of TLR9 reduced spontaneous liver injury, inflammation, fibrosis, and cancer development in Tak1ΔHep mice. In addition, the common adaptor, myeloid differentiation factor 88 (MyD88)-deficient Tak1ΔHep mice also attenuated liver injury, macrophage recruitment, collagen deposition, and tumor growth compared with control Tak1ΔHep mice. Genetic ablation of TNF receptor type I (TNFR) in Tak1ΔHep mice remarkably reduced liver inflammation-fibrosis-cancer axis. Surprisingly, disruption of interleukin-1 receptor (IL-1R) had no effect on liver injury and tumor formation, although Il1r-deficient Tak1ΔHep showed attenuated macrophage infiltration and collagen deposition. In conclusion, TLR4- and TLR9-MyD88 are driving forces of progression to HCC accompanied by liver inflammation and fibrosis in Tak1ΔHep mice. Importantly, TLR4 and TLR9 downstream TNFR, but not IL-1R signaling is crucial for the development of HCC in Tak1ΔHep mice.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Hepatocytes/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , MAP Kinase Kinase Kinases/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology
15.
Infect Immun ; 85(10)2017 10.
Article in English | MEDLINE | ID: mdl-28760932

ABSTRACT

Tumor progression locus 2 (Tpl2) is a serine-threonine kinase that regulates Th1 differentiation, secretion of the inflammatory cytokine gamma interferon (IFN-γ), and host defense against the intracellular pathogens Toxoplasma gondii, Listeria monocytogenes, and Mycobacterium tuberculosis However, relatively little is known about the contribution of Tpl2 to Th17 differentiation and immune cell function during infection with an extracellular pathogen. The goal of this study was to determine whether Tpl2 influences the immune response generated to the extracellular bacterium Citrobacter rodentium, which induces a mixed Th1 and Th17 response. During peak infection with C. rodentium, Tpl2-/- mice experienced greater bacterial burdens with evidence of dissemination to the liver and spleen but ultimately cleared the bacteria within 3 weeks postinfection, similar to the findings for wild-type mice. Tpl2-/- mice also recruited fewer neutrophils and monocytes to the colon during peak infection, which correlated with increased bacterial burdens. In mixed bone marrow chimeras, Tpl2 was shown to play a T cell-intrinsic role in promoting both IFN-γ and interleukin-17A production during infection with C. rodentium However, upon CD4 T cell transfer into Rag-/- mice, Tpl2-/- CD4 T cells were as protective as wild-type CD4 T cells against the dissemination of bacteria and mortality. These data indicate that the enhanced bacterial burdens in Tpl2-/- mice are not caused primarily by impairments in CD4 T cell function but result from defects in innate immune cell recruitment and function.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Bacterial Load , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Interferon-gamma/immunology , Interleukin-17/immunology , Intestines/immunology , Intestines/microbiology , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics
16.
Comp Med ; 67(4): 344-349, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28830581

ABSTRACT

Staphylococcus xylosus is a commensal bacterium found on the skin and mucosal surfaces of SPF mice. S. xylosus is rarely pathogenic, most often causing skin lesions and dermatitis in immunocompromised mice, particularly those with impaired NADPH oxidase function. Here we report spontaneous infection with S. xylosus in Rag1-/-Tpl2-/- mice. Infection was characterized by the presence of alopecia, crusts, and scaly skin. S. xylosus was detected in the feces, skin, lymph nodes, and lungs of Rag1-/-Tpl2-/- mice and led to mortality or euthanasia due to humane endpoints. C57BL/6 mice were culture-positive for S. xylosus on the skin, and Rag1-/- and Tpl2-/- mice were culture-positive on the skin and occasionally in the feces. However, S. xylosus did not cause clinical symptoms in C57BL/6, Rag1-/-, or Tpl2-/- mice. Compared with those in Rag1-/- mice, relative concentrations of circulating monocytes, but not neutrophils or lymphocytes, were increased in Rag1-/-Tpl2-/- mice, consistent with their increased incidence of clinical symptoms. Overall, this case study suggests a novel role for Tpl2 in T-cell-independent host resistance to the otherwise commensal organism S. xylosus.


Subject(s)
Dermatitis/veterinary , Homeodomain Proteins/genetics , Immunocompromised Host , MAP Kinase Kinase Kinases/genetics , Opportunistic Infections/veterinary , Proto-Oncogene Proteins/genetics , Skin/microbiology , Staphylococcal Skin Infections/veterinary , Staphylococcus/pathogenicity , Animals , Bacterial Translocation , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/microbiology , Feces/microbiology , Genetic Predisposition to Disease , Host-Pathogen Interactions , MAP Kinase Kinase Kinases/deficiency , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/microbiology , Opportunistic Infections/genetics , Opportunistic Infections/immunology , Opportunistic Infections/microbiology , Phenotype , Proto-Oncogene Proteins/deficiency , Skin/immunology , Skin/pathology , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcus/classification , Staphylococcus/immunology
17.
Arterioscler Thromb Vasc Biol ; 37(2): 237-246, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27856455

ABSTRACT

OBJECTIVE: Map3k8 (Cot/Tpl2) activates the MKK1/2-ERK1/2, MAPK pathway downstream from interleukin-1R, tumor necrosis factor-αR, NOD-2R (nucleotide-binding oligomerization domain-like 2R), adiponectinR, and Toll-like receptors. Map3k8 plays a key role in innate and adaptive immunity and influences inflammatory processes by modulating the functions of different cell types. However, its role in atherogenesis remains unknown. In this study, we analyzed the role of this kinase in this pathology. APPROACH AND RESULTS: We show here that Map3k8 deficiency results in smaller numbers of Ly6ChighCD11clow and Ly6ClowCD11chigh monocytes in ApoE-/- mice fed a high-fat diet (HFD). Map3k8-/-ApoE-/- monocytes displayed high rates of apoptosis and reduced amounts of Nr4a1, a transcription factor known to modulate apoptosis in Ly6ClowCD11chigh monocytes. Map3k8-/-ApoE-/- splenocytes and macrophages showed irregular patterns of cytokine and chemokine expression. Map3k8 deficiency altered cell adhesion and migration in vivo and decreased CCR2 expression, a determinant chemokine receptor for monocyte mobilization, on circulating Ly6ChighCD11clow monocytes. Map3k8-/-ApoE-/- mice fed an HFD showed decreased cellular infiltration in the atherosclerotic plaque, with low lipid content. Lesions had similar size after Map3k8+/+ApoE-/- bone marrow transplant into Map3k8-/-ApoE-/- and Map3k8+/+ApoE-/- mice fed an HFD, whereas smaller plaques were observed after the transplantation of bone marrow lacking both ApoE and Map3k8. CONCLUSIONS: Map3k8 decreases apoptosis of monocytes and enhances CCR2 expression on Ly6ChighCD11clow monocytes of ApoE-/- mice fed an HFD. These findings explain the smaller aortic lesions in ApoE-/- mice with Map3k8-/-ApoE-/- bone marrow cells fed an HFD, supporting further studies of Map3k8 as an antiatherosclerotic target.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , MAP Kinase Kinase Kinases/metabolism , Monocytes/metabolism , Plaque, Atherosclerotic , Proto-Oncogene Proteins/metabolism , Animals , Antigens, Ly/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , CD11c Antigen/metabolism , Cell Adhesion , Chemotaxis, Leukocyte , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Genetic Predisposition to Disease , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Macrophages, Peritoneal/metabolism , Male , Mice, Knockout , Monocytes/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenotype , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptors, CCR2/metabolism , Signal Transduction , Spleen/metabolism
18.
JCI Insight ; 1(7): e81175, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27699262

ABSTRACT

Obesity is an increasing health problem worldwide, and nonsurgical strategies to treat obesity have remained rather inefficient. We here show that acute loss of TGF-ß-activated kinase 1 (TAK1) in adipocytes results in an increased rate of apoptotic adipocyte death and increased numbers of M2 macrophages in white adipose tissue. Mice with adipocyte-specific TAK1 deficiency have reduced adipocyte numbers and are resistant to obesity induced by a high-fat diet or leptin deficiency. In addition, adipocyte-specific TAK1-deficient mice under a high-fat diet showed increased energy expenditure, which was accompanied by enhanced expression of the uncoupling protein UCP1. Interestingly, acute induction of adipocyte-specific TAK1 deficiency in mice already under a high-fat diet was able to stop further weight gain and improved glucose tolerance. Thus, loss of TAK1 in adipocytes reduces the total number of adipocytes, increases browning of white adipose tissue, and may be an attractive strategy to treat obesity, obesity-dependent diabetes, and other associated complications.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Apoptosis , MAP Kinase Kinase Kinases/deficiency , Obesity/genetics , Adipocytes/cytology , Animals , Diet, High-Fat , MAP Kinase Kinase Kinases/genetics , Macrophages/cytology , Mice , Mice, Inbred C57BL , Uncoupling Protein 1/metabolism
19.
Toxins (Basel) ; 8(9)2016 09 02.
Article in English | MEDLINE | ID: mdl-27598200

ABSTRACT

Ricin activates the proinflammatory ribotoxic stress response through the mitogen activated protein 3 kinase (MAP3K) ZAK, resulting in activation of mitogen activated protein kinases (MAPKs) p38 and JNK1/2. We had a novel zak-/- mouse generated to study the role of ZAK signaling in vivo during ricin intoxication. To characterize this murine strain, we intoxicated zak-/- and zak+/+ bone marrow-derived murine macrophages with ricin, measured p38 and JNK1/2 activation by Western blot, and measured zak, c-jun, and cxcl-1 expression by qRT-PCR. To determine whether zak-/- mice differed from wild-type mice in their in vivo response to ricin, we performed oral ricin intoxication experiments with zak+/+ and zak-/- mice, using blinded histopathology scoring of duodenal tissue sections to determine differences in tissue damage. Unlike macrophages derived from zak+/+ mice, those derived from the novel zak-/- strain fail to activate p38 and JNK1/2 and have decreased c-jun and cxcl-1 expression following ricin intoxication. Furthermore, compared with zak+/+ mice, zak-/- mice have decreased duodenal damage following in vivo ricin challenge. zak-/- mice demonstrate a distinct ribotoxic stress-associated phenotype in response to ricin and therefore provide a new animal model for in vivo studies of ZAK signaling.


Subject(s)
Duodenum/drug effects , MAP Kinase Kinase Kinases/deficiency , Macrophages/drug effects , Ricin/toxicity , Stress, Physiological/drug effects , Animals , Cells, Cultured , Chemokine CXCL1/metabolism , Duodenum/enzymology , Duodenum/pathology , Enzyme Activation , Genotype , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Macrophages/enzymology , Macrophages/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...