1
|
Amosu MM, Jankowski AM, McCright JC, Yang BE, Grano de Oro Fernandez J, Moore KA, Gadde HS, Donthi M, Kaluzienski ML, Maisel K. Plasmacytoid Dendritic Cells Mediate CpG-ODN-induced Increase in Survival in a Mouse Model of Lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2024; 71:519-533. [PMID: 38990702 DOI: 10.1165/rcmb.2023-0410oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/11/2024] [Indexed: 07/13/2024] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a devastating disease primarily found in women of reproductive age that leads to cystic destruction of the lungs. Recent work has shown that LAM causes immunosuppression and that checkpoint inhibitors can be used as LAM treatment. Toll-like receptor (TLR) agonists can also reactivate immunity, and the TLR9 agonist CpG oligodeoxynucleotide (CpG-ODN) has been effective in treating lung cancer in animal models. In this study, we investigated the use of TLR9 agonist CpG-ODN as LAM immunotherapy in combination with checkpoint inhibitor anti-PD1 and standard of care rapamycin, and determined the immune mechanisms underlying therapeutic efficacy. We used survival studies, flow cytometry, ELISA, and histology to assess immune response and survival after intranasal treatment with CpG-ODN in combination with rapamycin or anti-PD1 therapy in a mouse model of metastatic LAM. We found that local administration of CpG-ODN enhances survival in a mouse model of LAM. We found that a lower dose led to longer survival, likely because of fewer local side effects, but increased LAM nodule count and size compared with the higher dose. CpG-ODN treatment also reduced regulatory T cells and increased the number of T-helper type 17 cells as well as cytotoxic T cells. These effects appear to be mediated in part by plasmacytoid dendritic cells because depletion of plasmacytoid dendritic cells reduces survival and abrogates T-helper type 17 cell response. Finally, we found that CpG-ODN treatment is effective in early-stage and progressive disease and is additive with anti-PD1 therapy and rapamycin. In summary, we have found that TLR9 agonist CpG-ODN can be used as LAM immunotherapy and effectively synergizes with rapamycin and anti-PD1 therapy in LAM.
Collapse
Affiliation(s)
- Mayowa M Amosu
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Ashleigh M Jankowski
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Jacob C McCright
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Bennett E Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | | | - Kaitlyn A Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Havish S Gadde
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Mehul Donthi
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Michele L Kaluzienski
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
2
|
Reed EB, Orbeta S, Miao BA, Sitikov A, Chen B, Levitan I, Solway J, Mutlu GM, Fang Y, Mongin AA, Dulin NO. Anoctamin-1 is induced by TGF-β and contributes to lung myofibroblast differentiation. Am J Physiol Lung Cell Mol Physiol 2024; 326:L111-L123. [PMID: 38084409 PMCID: PMC11279757 DOI: 10.1152/ajplung.00155.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by progressive scarring of the lungs and resulting in deterioration in lung function. Transforming growth factor-β (TGF-β) is one of the most established drivers of fibrotic processes. TGF-β promotes the transformation of tissue fibroblasts to myofibroblasts, a key finding in the pathogenesis of pulmonary fibrosis. We report here that TGF-β robustly upregulates the expression of the calcium-activated chloride channel anoctamin-1 (ANO1) in human lung fibroblasts (HLFs) at mRNA and protein levels. ANO1 is readily detected in fibrotic areas of IPF lungs in the same area with smooth muscle α-actin (SMA)-positive myofibroblasts. TGF-β-induced myofibroblast differentiation (determined by the expression of SMA, collagen-1, and fibronectin) is significantly inhibited by a specific ANO1 inhibitor, T16Ainh-A01, or by siRNA-mediated ANO1 knockdown. T16Ainh-A01 and ANO1 siRNA attenuate profibrotic TGF-β signaling, including activation of RhoA pathway and AKT, without affecting initial Smad2 phosphorylation. Mechanistically, TGF-β treatment of HLFs results in a significant increase in intracellular chloride levels, which is prevented by T16Ainh-A01 or by ANO1 knockdown. The downstream mechanism involves the chloride-sensing "with-no-lysine (K)" kinase (WNK1). WNK1 siRNA significantly attenuates TGF-β-induced myofibroblast differentiation and signaling (RhoA pathway and AKT), whereas the WNK1 kinase inhibitor WNK463 is largely ineffective. Together, these data demonstrate that 1) ANO1 is a TGF-β-inducible chloride channel that contributes to increased intracellular chloride concentration in response to TGF-β; and 2) ANO1 mediates TGF-β-induced myofibroblast differentiation and fibrotic signaling in a manner dependent on WNK1 protein but independent of WNK1 kinase activity.NEW & NOTEWORTHY This study describes a novel mechanism of differentiation of human lung fibroblasts (HLFs) to myofibroblasts: the key process in the pathogenesis of pulmonary fibrosis. Transforming growth factor-β (TGF-β) drives the expression of calcium-activated chloride channel anoctmin-1 (ANO1) leading to an increase in intracellular levels of chloride. The latter recruits chloride-sensitive with-no-lysine (K) kinase (WNK1) to activate profibrotic RhoA and AKT signaling pathways, possibly through activation of mammalian target of rapamycin complex-2 (mTORC2), altogether promoting myofibroblast differentiation.
Collapse
Affiliation(s)
- Eleanor B Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Shaina Orbeta
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, United States
| | - Bernadette A Miao
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Albert Sitikov
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Julian Solway
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Yun Fang
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, United States
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
3
|
Evans JF, McCormack FX, Sonenberg N, Krymskaya VP. Lost in translation: a neglected mTOR target for lymphangioleiomyomatosis. Eur Respir Rev 2023; 32:230100. [PMID: 37758276 PMCID: PMC10523142 DOI: 10.1183/16000617.0100-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/24/2023] [Indexed: 09/30/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a cystic lung disease of women resulting from mutations in tuberous sclerosis complex (TSC) genes that suppress the mammalian target of rapamycin complex 1 (mTORC1) pathway. mTORC1 activation enhances a plethora of anabolic cellular functions, mainly via the activation of mRNA translation through stimulation of ribosomal protein S6 kinase (S6K1)/ribosomal protein S6 (S6) and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1)/eukaryotic translation initiation factor 4E (eIF4E). Rapamycin (sirolimus), an allosteric inhibitor of mTORC1, stabilises lung function in many but not all LAM patients and, upon cessation of the drug, disease progression resumes. At clinically tolerable concentrations, rapamycin potently inhibits the ribosomal S6K1/S6 translation ribosome biogenesis and elongation axis, but not the translation 4E-BP1/eIF4E initiation axis. In this mini-review, we propose that inhibition of mTORC1-driven translation initiation is an obvious but underappreciated therapeutic strategy in LAM, TSC and other mTORC1-driven diseases.
Collapse
Affiliation(s)
- Jilly F Evans
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Vera P Krymskaya
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Elia D, Cassandro R, Caminati A, Luisi F, Harari S. Lymphangioleiomyomatosis. Presse Med 2023; 52:104173. [PMID: 37696446 DOI: 10.1016/j.lpm.2023.104173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) is an ultra-rare, slowly progressive neoplastic cystic disease, belonging to the group of PEComas. It can occur sporadically or associated to tuberous sclerosis complex disease and affects mainly women in child-birth age. Dyspnoea is the most frequent symptom referred to the time of diagnosis, however spontaneous pneumothorax may be a typical presentation associated to extrathoracic manifestations, such as renal angiomyolipomas. In the last decade, important advances in understanding molecular mechanisms underlying the LAM pathogenesis have been reached. It has allowed to obtain improvements in the research of novel biomarkers, treatment and a better management of the disease.
Collapse
Affiliation(s)
- Davide Elia
- UO di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, MultiMedica IRCCS, Via San Vittore 12, 20123 Milan, Italy
| | - Roberto Cassandro
- UO di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, MultiMedica IRCCS, Via San Vittore 12, 20123 Milan, Italy
| | - Antonella Caminati
- UO di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, MultiMedica IRCCS, Via San Vittore 12, 20123 Milan, Italy
| | - Francesca Luisi
- UO di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, MultiMedica IRCCS, Via San Vittore 12, 20123 Milan, Italy
| | - Sergio Harari
- UO di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, MultiMedica IRCCS, Via San Vittore 12, 20123 Milan, Italy; Department of Clinical Sciences and Community Health, - Università degli Studi di Milano, 20123 Milan, Italy.
| |
Collapse
|
5
|
Zhu QY, He ZM, Cao WM, Li B. The role of TSC2 in breast cancer: a literature review. Front Oncol 2023; 13:1188371. [PMID: 37251941 PMCID: PMC10213421 DOI: 10.3389/fonc.2023.1188371] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
TSC2 is a tumor suppressor gene as well as a disease-causing gene for autosomal dominant disorder tuberous sclerosis complex (TSC). Research has found that some tumor tissues have lower TSC2 expression levels than normal tissues. Furthermore, low expression of TSC2 is associated with poor prognosis in breast cancer. TSC2 acts as a convergence point of a complex network of signaling pathways and receives signals from the PI3K, AMPK, MAPK, and WNT pathways. It also regulates cellular metabolism and autophagy through inhibition of a mechanistic target of rapamycin complex, which are processes relevant to the progression, treatment, and prognosis of breast cancer. In-depth study of TSC2 functions provides significant guidance for clinical applications in breast cancer, including improving the treatment efficacy, overcoming drug resistance, and predicting prognosis. In this review, protein structure and biological functions of TSC2 were described and recent advances in TSC2 research in different molecular subtypes of breast cancer were summarized.
Collapse
Affiliation(s)
- Qiao-Yan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Zhe-Min He
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Bei Li
- Department of Geriatric, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
An In Vitro Analysis of TKI-Based Sequence Therapy in Renal Cell Carcinoma Cell Lines. Int J Mol Sci 2023; 24:ijms24065648. [PMID: 36982721 PMCID: PMC10058472 DOI: 10.3390/ijms24065648] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
The tyrosine kinase inhibitor (TKI) cabozantinib might impede the growth of the sunitinib-resistant cell lines by targeting MET and AXL overexpression in metastatic renal cell carcinoma (mRCC). We studied the role of MET and AXL in the response to cabozantinib, particularly following long-term administration with sunitinib. Two sunitinib-resistant cell lines, 786-O/S and Caki-2/S, and the matching 786-O/WT and Caki-2/WT cells were exposed to cabozantinib. The drug response was cell-line-specific. The 786-O/S cells were less growth-inhibited by cabozantinib than 786-O/WT cells (p-value = 0.02). In 786-O/S cells, the high level of phosphorylation of MET and AXL was not affected by cabozantinib. Despite cabozantinib hampering the high constitutive phosphorylation of MET, the Caki-2 cells showed low sensitivity to cabozantinib, and this was independent of sunitinib pretreatment. In both sunitinib-resistant cell lines, cabozantinib increased Src-FAK activation and impeded mTOR expression. The modulation of ERK and AKT was cell-line-specific, mirroring the heterogeneity among the patients. Overall, the MET- and AXL-driven status did not affect cell responsiveness to cabozantinib in the second-line treatment. The activation of Src-FAK might counteract cabozantinib activity and contribute to tumor survival and may be considered an early indicator of therapy response.
Collapse
|
7
|
Brazill JM, Shin D, Magee K, Majumdar A, Shen IR, Cavalli V, Scheller EL. Knockout of TSC2 in Nav1.8+ neurons predisposes to the onset of normal weight obesity. Mol Metab 2023; 68:101664. [PMID: 36586433 PMCID: PMC9841058 DOI: 10.1016/j.molmet.2022.101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Obesity and nutrient oversupply increase mammalian target of rapamycin (mTOR) signaling in multiple cell types and organs, contributing to the onset of insulin resistance and complications of metabolic disease. However, it remains unclear when and where mTOR activation mediates these effects, limiting options for therapeutic intervention. The objective of this study was to isolate the role of constitutive mTOR activation in Nav1.8-expressing peripheral neurons in the onset of diet-induced obesity, bone loss, and metabolic disease. METHODS In humans, loss of function mutations in tuberous sclerosis complex 2 (TSC2) lead to maximal constitutive activation of mTOR. To mirror this in mice, we bred Nav1.8-Cre with TSC2fl/fl animals to conditionally delete TSC2 in Nav1.8-expressing neurons. Male and female mice were studied from 4- to 34-weeks of age and a subset of animals were fed a high-fat diet (HFD) for 24-weeks. Assays of metabolism, body composition, bone morphology, and behavior were performed. RESULTS By lineage tracing, Nav1.8-Cre targeted peripheral sensory neurons, a subpopulation of postganglionic sympathetics, and several regions of the brain. Conditional knockout of TSC2 in Nav1.8-expressing neurons (Nav1.8-TSC2KO) selectively upregulated neuronal mTORC1 signaling. Male, but not female, Nav1.8-TSC2KO mice had a 4-10% decrease in body size at baseline. When challenged with HFD, both male and female Nav1.8-TSC2KO mice resisted diet-induced gains in body mass. However, this did not protect against HFD-induced metabolic dysfunction and bone loss. In addition, despite not gaining weight, Nav1.8-TSC2KO mice fed HFD still developed high body fat, a unique phenotype previously referred to as 'normal weight obesity'. Nav1.8-TSC2KO mice also had signs of chronic itch, mild increases in anxiety-like behavior, and sex-specific alterations in HFD-induced fat distribution that led to enhanced visceral obesity in males and preferential deposition of subcutaneous fat in females. CONCLUSIONS Knockout of TSC2 in Nav1.8+ neurons increases itch- and anxiety-like behaviors and substantially modifies fat storage and metabolic responses to HFD. Though this prevents HFD-induced weight gain, it masks depot-specific fat expansion and persistent detrimental effects on metabolic health and peripheral organs such as bone, mimicking the 'normal weight obesity' phenotype that is of growing concern. This supports a mechanism by which increased neuronal mTOR signaling can predispose to altered adipose tissue distribution, adipose tissue expansion, impaired peripheral metabolism, and detrimental changes to skeletal health with HFD - despite resistance to weight gain.
Collapse
Affiliation(s)
- Jennifer M Brazill
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University, Saint Louis, MO, USA.
| | - David Shin
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University, Saint Louis, MO, USA.
| | - Kristann Magee
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University, Saint Louis, MO, USA.
| | - Anurag Majumdar
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University, Saint Louis, MO, USA.
| | - Ivana R Shen
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University, Saint Louis, MO, USA.
| | - Valeria Cavalli
- Department of Neuroscience, Washington University, Saint Louis, MO, USA; Center of Regenerative Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA.
| | - Erica L Scheller
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University, Saint Louis, MO, USA; Center of Regenerative Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Department of Cell Biology and Physiology, Washington University, Saint Louis, MO, USA; Department of Biomedical Engineering, Washington University, Saint Louis, MO, USA.
| |
Collapse
|
8
|
Shen Y, Goncharov DA, Pena A, Baust J, Barragan AC, Ray A, Rode A, Bachman TN, Chang B, Jiang L, Dieffenbach P, Fredenburgh LE, Rojas M, DeLisser H, Mora AL, Kudryashova TV, Goncharova EA. Cross-talk between TSC2 and the extracellular matrix controls pulmonary vascular proliferation and pulmonary hypertension. Sci Signal 2022; 15:eabn2743. [PMID: 36473049 PMCID: PMC9869933 DOI: 10.1126/scisignal.abn2743] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increased proliferation and survival of cells in small pulmonary arteries (PAs) drive pulmonary arterial hypertension (PAH). Because cell growth mediated by the mTOR-containing mTORC1 complex is inhibited by tuberous sclerosis complex 2 (TSC2), we investigated the role of this GTPase-activating protein in PAH pathology. TSC2 abundance was decreased in remodeled small PAs and PA vascular smooth muscle cells (PAVSMCs) from patients with PAH or from rodent pulmonary hypertension (PH) models, as well as PAVSMCs maintained on substrates that reproduced pathology-induced stiffness. Accordingly, mice with smooth muscle-specific reduction in TSC2 developed PH. At the molecular level, decreased TSC2 abundance led to stiffness-induced PAVSMC proliferation, increased abundance of the mechanosensitive transcriptional coactivators YAP/TAZ, and enhanced mTOR kinase activity. Moreover, extracellular matrix (ECM) produced by TSC2-deficient PAVSMCs stimulated the proliferation of nondiseased PA adventitial fibroblasts and PAVSMCs through fibronectin and its receptor, the α5β1 integrin. Reconstituting TSC2 in PAVSMCs from patients with PAH through overexpression or treatment with the SIRT1 activator SRT2104 decreased YAP/TAZ abundance, mTOR activity, and ECM production, as well as inhibited proliferation and induced apoptosis. In two rodent models of PH, SRT2104 treatment restored TSC2 abundance, attenuated pulmonary vascular remodeling, and ameliorated PH. Thus, TSC2 in PAVSMCs integrates ECM composition and stiffness with pro-proliferative and survival signaling, and restoring TSC2 abundance could be an attractive therapeutic option to treat PH.
Collapse
Affiliation(s)
- Yuanjun Shen
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA 95616
| | - Dmitry A. Goncharov
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA 95616
| | - Andressa Pena
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Jeffrey Baust
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Andres Chavez Barragan
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Arnab Ray
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Analise Rode
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Timothy N. Bachman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Baojun Chang
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Lifeng Jiang
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA 95616
| | - Paul Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
| | - Laura E. Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Current affiliation: Regeneron Pharmaceuticals, Tarrytown, NY
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University College of Medicine, Columbus, OH, USA 43210
| | - Horace DeLisser
- Department of Pathology and Laboratory Medicine, Pulmonary Vascular Disease Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA 19104
| | - Ana L. Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University College of Medicine, Columbus, OH, USA 43210
| | - Tatiana V. Kudryashova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA 95616
| | - Elena. A. Goncharova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA 95616
| |
Collapse
|
9
|
Voisin A, Gaillard A, Balbous A, Leveziel N. Proteins Associated with Phagocytosis Alteration in Retinal Pigment Epithelial Cells Derived from Age-Related Macular Degeneration Patients. Antioxidants (Basel) 2022; 11:antiox11040713. [PMID: 35453399 PMCID: PMC9028973 DOI: 10.3390/antiox11040713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
Age-related macular degeneration (AMD) is partially characterized by retinal pigment epithelial (RPE) cell dysfunction. This study focused on phagocytosis activity and its involvement in AMD. Phagocytic activity was analyzed by flow cytometry using porcine photoreceptor outer segment (POS) and fluorescent beads in basal and under oxidative stress condition induced by Fe-NTA in fifteen hiPSC-RPE cell lines (six controls, six atrophic AMD and three exudative AMD). Oxidative stress exposure inhibited phagocytosis in the same manner for control, atrophic AMD (AMDa) and exudative AMD (AMDe) cell lines. However, altered phagocytosis in basal condition in hiPSC-RPE AMDa/e was observed compared to control cell lines. Gene expression after 3 or 24 h of POS incubation was analyzed by RNA-Seq based transcriptomic profiling. Differential gene expression was observed by RNA seq after 3 and 24 h POS exposure. We have focused on the genes involved in mTOR/PI3K-AKT/MEK-ERK pathway. We investigated differences in gene expression by analyzing the expression levels and activity of the corresponding proteins by Western blot. We showed the involvement of three proteins essential for phagocytosis activity: fak, tuberin and rictor. These findings demonstrate that hiPSC-RPE AMDa/e cells have a typical disease phenotype characterized by alteration of the main function of RPE cells, phagocytosis activity.
Collapse
Affiliation(s)
- Audrey Voisin
- Laboratoire de Neurosciences Expérimentales et Cliniques, Equipe Thérapie Cellulaire dans les Pathologies Cérébrales, INSERM, Université de Poitiers, F-86073 Poitiers, France; (A.G.); (A.B.); (N.L.)
- CHU Poitiers, F-86021 Poitiers, France
- Correspondence:
| | - Afsaneh Gaillard
- Laboratoire de Neurosciences Expérimentales et Cliniques, Equipe Thérapie Cellulaire dans les Pathologies Cérébrales, INSERM, Université de Poitiers, F-86073 Poitiers, France; (A.G.); (A.B.); (N.L.)
| | - Anaïs Balbous
- Laboratoire de Neurosciences Expérimentales et Cliniques, Equipe Thérapie Cellulaire dans les Pathologies Cérébrales, INSERM, Université de Poitiers, F-86073 Poitiers, France; (A.G.); (A.B.); (N.L.)
- CHU Poitiers, F-86021 Poitiers, France
| | - Nicolas Leveziel
- Laboratoire de Neurosciences Expérimentales et Cliniques, Equipe Thérapie Cellulaire dans les Pathologies Cérébrales, INSERM, Université de Poitiers, F-86073 Poitiers, France; (A.G.); (A.B.); (N.L.)
- CHU Poitiers, F-86021 Poitiers, France
| |
Collapse
|
10
|
Landh E, Wang R, Moir LM, Traini D, Young PM, Ong HX. Prospective nanoparticle treatments for lymphangioleiomyomatosis. Expert Opin Drug Deliv 2022; 19:75-86. [DOI: 10.1080/17425247.2022.2029401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Emelie Landh
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW 2037, Australia
- Discipline of Pharmacology, Faculty of Medicine and Health, Sydney, Australia
| | - Roger Wang
- Discipline of Pharmacology, Faculty of Medicine and Health, Sydney, Australia
| | - Lyn M. Moir
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW 2037, Australia
- Discipline of Pharmacology, Faculty of Medicine and Health, Sydney, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW 2037, Australia
- Discipline of Pharmacology, Faculty of Medicine and Health, Sydney, Australia
| | - Paul M. Young
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW 2037, Australia
- Discipline of Pharmacology, Faculty of Medicine and Health, Sydney, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW 2037, Australia
- Discipline of Pharmacology, Faculty of Medicine and Health, Sydney, Australia
| |
Collapse
|
11
|
McCarthy C, Gupta N, Johnson SR, Yu JJ, McCormack FX. Lymphangioleiomyomatosis: pathogenesis, clinical features, diagnosis, and management. THE LANCET. RESPIRATORY MEDICINE 2021; 9:1313-1327. [PMID: 34461049 DOI: 10.1016/s2213-2600(21)00228-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/02/2021] [Accepted: 05/07/2021] [Indexed: 01/15/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a slowly progressive, low-grade, metastasising neoplasm of women, characterised by infiltration of the lung parenchyma with abnormal smooth muscle-like cells, resulting in cystic lung destruction. The invading cell in LAM arises from an unknown source and harbours mutations in tuberous sclerosis complex (TSC) genes that result in constitutive activation of the mechanistic target of rapamycin (mTOR) pathway, dysregulated cellular proliferation, and a programme of frustrated lymphangiogenesis, culminating in disordered lung remodelling and respiratory failure. Over the past two decades, all facets of LAM basic and clinical science have seen important advances, including improved understanding of molecular mechanisms, novel diagnostic and prognostic biomarkers, effective treatment strategies, and comprehensive clinical practice guidelines. Further research is needed to better understand the natural history of LAM; develop more powerful diagnostic, prognostic, and predictive biomarkers; optimise the use of inhibitors of mTOR complex 1 in the treatment of LAM; and explore novel approaches to the development of remission-inducing therapies.
Collapse
Affiliation(s)
- Cormac McCarthy
- Department of Respiratory Medicine, St Vincent's University Hospital, University College Dublin, Dublin, Ireland.
| | - Nishant Gupta
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Simon R Johnson
- Division of Respiratory Medicine, University of Nottingham, NIHR Respiratory Biomedical Research Centre, Nottingham, UK
| | - Jane J Yu
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Francis X McCormack
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
12
|
Pungsrinont T, Kallenbach J, Baniahmad A. Role of PI3K-AKT-mTOR Pathway as a Pro-Survival Signaling and Resistance-Mediating Mechanism to Therapy of Prostate Cancer. Int J Mol Sci 2021; 22:11088. [PMID: 34681745 PMCID: PMC8538152 DOI: 10.3390/ijms222011088] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Androgen deprivation therapy (ADT) and androgen receptor (AR)-targeted therapy are the gold standard options for treating prostate cancer (PCa). These are initially effective, as localized and the early stage of metastatic disease are androgen- and castration-sensitive. The tumor strongly relies on systemic/circulating androgens for activating AR signaling to stimulate growth and progression. However, after a certain point, the tumor will eventually develop a resistant stage, where ADT and AR antagonists are no longer effective. Mechanistically, it seems that the tumor becomes more aggressive through adaptive responses, relies more on alternative activated pathways, and is less dependent on AR signaling. This includes hyperactivation of PI3K-AKT-mTOR pathway, which is a central signal that regulates cell pro-survival/anti-apoptotic pathways, thus, compensating the blockade of AR signaling. The PI3K-AKT-mTOR pathway is well-documented for its crosstalk between genomic and non-genomic AR signaling, as well as other signaling cascades. Such a reciprocal feedback loop makes it more complicated to target individual factor/signaling for treating PCa. Here, we highlight the role of PI3K-AKT-mTOR signaling as a resistance mechanism for PCa therapy and illustrate the transition of prostate tumor from AR signaling-dependent to PI3K-AKT-mTOR pathway-dependent. Moreover, therapeutic strategies with inhibitors targeting the PI3K-AKT-mTOR signal used in clinic and ongoing clinical trials are discussed.
Collapse
Affiliation(s)
| | | | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany; (T.P.); (J.K.)
| |
Collapse
|
13
|
Querfurth H, Lee HK. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol Neurodegener 2021; 16:44. [PMID: 34215308 PMCID: PMC8252260 DOI: 10.1186/s13024-021-00428-5] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Novel targets to arrest neurodegeneration in several dementing conditions involving misfolded protein accumulations may be found in the diverse signaling pathways of the Mammalian/mechanistic target of rapamycin (mTOR). As a nutrient sensor, mTOR has important homeostatic functions to regulate energy metabolism and support neuronal growth and plasticity. However, in Alzheimer's disease (AD), mTOR alternately plays important pathogenic roles by inhibiting both insulin signaling and autophagic removal of β-amyloid (Aβ) and phospho-tau (ptau) aggregates. It also plays a role in the cerebrovascular dysfunction of AD. mTOR is a serine/threonine kinase residing at the core in either of two multiprotein complexes termed mTORC1 and mTORC2. Recent data suggest that their balanced actions also have implications for Parkinson's disease (PD) and Huntington's disease (HD), Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS). Beyond rapamycin; an mTOR inhibitor, there are rapalogs having greater tolerability and micro delivery modes, that hold promise in arresting these age dependent conditions.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA.
| | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Bauer PK, Flicker M, Fabian E, Flick H, Brcic L, Liegl-Atzwanger B, Janisch M, Fuchsjäger M, Olschewski H, Krejs GJ. Clinical-Pathological Conference Series from the Medical University of Graz : Case No 170: A 33-year-old psychologist with severe dyspnea and right-sided chylothorax. Wien Klin Wochenschr 2021; 133:65-72. [PMID: 33119872 PMCID: PMC7840639 DOI: 10.1007/s00508-020-01753-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Philipp K Bauer
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Martin Flicker
- Department of Internal Medicine, State Hospital Hochsteiermark, Leoben, Austria
| | - Elisabeth Fabian
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Holger Flick
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Michael Janisch
- Division of General Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Michael Fuchsjäger
- Division of General Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Horst Olschewski
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Guenter J Krejs
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| |
Collapse
|
15
|
Evans JF, Obraztsova K, Lin SM, Krymskaya VP. CrossTORC and WNTegration in Disease: Focus on Lymphangioleiomyomatosis. Int J Mol Sci 2021; 22:ijms22052233. [PMID: 33668092 PMCID: PMC7956553 DOI: 10.3390/ijms22052233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) and wingless-related integration site (Wnt) signal transduction networks are evolutionarily conserved mammalian growth and cellular development networks. Most cells express many of the proteins in both pathways, and this review will briefly describe only the key proteins and their intra- and extracellular crosstalk. These complex interactions will be discussed in relation to cancer development, drug resistance, and stem cell exhaustion. This review will also highlight the tumor-suppressive tuberous sclerosis complex (TSC) mutated, mTOR-hyperactive lung disease of women, lymphangioleiomyomatosis (LAM). We will summarize recent advances in the targeting of these pathways by monotherapy or combination therapy, as well as future potential treatments.
Collapse
|
16
|
A Systematic Review of Lymphangioleiomyomatosis on Diagnosis and Molecular Mechanism. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6612776. [PMID: 33628792 PMCID: PMC7892222 DOI: 10.1155/2021/6612776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/19/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Objective Lymphangioleiomyomatosis (LAM) is a rare low-grade metastatic tumor; however, LAM patients were always found in young age with difficulty for diagnosis. Our study is aimed at observing the clinical characteristics of patients with lymphangiomatosis, including the clinical manifestations, imaging findings, histopathological features, and immunophenotype. Methods We did a systematic review on LAM/PLAM cases, especially on male cases, and collected the clinical features and molecular mechanisms of PLAM based on previous findings. Results Diagnosis criteria were summarized by combining CT scans, MRI, immunohistochemistry results, and gene sequencing results for effectively distinguishing between PLAM and similar diseases. Moreover, our study illustrated the molecular mechanism of PLAM as well as the signaling pathway involved in the disease initials. In addition, a male case was reported with differential diagnosis on the clinical manifestations, microscopic features, immunophenotypes, and genotypes. Conclusion Our review will definitely improve the understanding of diagnosis and treatment in PLAM cases.
Collapse
|
17
|
Finding new edges: systems approaches to MTOR signaling. Biochem Soc Trans 2021; 49:41-54. [PMID: 33544134 PMCID: PMC7924996 DOI: 10.1042/bst20190730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 11/17/2022]
Abstract
Cells have evolved highly intertwined kinase networks to finely tune cellular homeostasis to the environment. The network converging on the mechanistic target of rapamycin (MTOR) kinase constitutes a central hub that integrates metabolic signals and adapts cellular metabolism and functions to nutritional changes and stress. Feedforward and feedback loops, crosstalks and a plethora of modulators finely balance MTOR-driven anabolic and catabolic processes. This complexity renders it difficult — if not impossible — to intuitively decipher signaling dynamics and network topology. Over the last two decades, systems approaches have emerged as powerful tools to simulate signaling network dynamics and responses. In this review, we discuss the contribution of systems studies to the discovery of novel edges and modulators in the MTOR network in healthy cells and in disease.
Collapse
|
18
|
Song X, Cai H, Yang C, Xue X, Wang J, Mo Y, Zhu M, Zhu G, Ye L, Jin M. Possible Novel Therapeutic Targets in Lymphangioleiomyomatosis Treatment. Front Med (Lausanne) 2020; 7:554134. [PMID: 33072782 PMCID: PMC7542236 DOI: 10.3389/fmed.2020.554134] [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: 04/21/2020] [Accepted: 08/13/2020] [Indexed: 12/16/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare systemic neoplastic disease that exclusively happens in women. Studies focusing on LAM and tuberous sclerosis complex (TSC) have made great progress in understanding the pathogenesis and searching for treatment. The inactive mutation of TSC1 or TSC2 is found in patients with LAM to activate the crucial mammalian target of rapamycin (mTOR) signaling pathway and result in enhanced cell proliferation and migration. However, it does not explain every step of tumorigenesis in LAM. Because cessation of rapamycin would break the stabilization of lung function or improved quality of life and lead to disease recurrent, continued studies on the pathogenesis of LAM are necessary to identify novel targets and new treatment. Researchers have found several aberrant regulations that affect the mTOR pathway such as its upstream or downstream molecules and compensatory pathways in LAM. Some therapeutic targets have been under study in clinical trials. New methods like genome-wide association studies have located a novel gene related to LAM. Herein, we review the current knowledge regarding pathogenesis and treatment of LAM and summarize novel targets of therapeutic potential recently.
Collapse
Affiliation(s)
- Xixi Song
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Cai
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chengyu Yang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomin Xue
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuqing Mo
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengchan Zhu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guiping Zhu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Ye
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meiling Jin
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Landh E, M Moir L, Bradbury P, Traini D, M Young P, Ong HX. Properties of rapamycin solid lipid nanoparticles for lymphatic access through the lungs & part I: the effect of size. Nanomedicine (Lond) 2020; 15:1927-1945. [PMID: 32820673 DOI: 10.2217/nnm-2020-0077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Lymphangioleiomyomatosis (LAM) is characterized by growth of smooth muscle-like cells in the lungs that spread to other organs via lymphatic vessels. Current oral rapamycin treatment is limited by low bioavailability of approximately 15%. Aim: The effect of inhaled rapamycin solid lipid nanoparticles (Rapa-SLNs) size on its penetration through the lymphatics. Method: Three Rapa-SLN formulations (200-1000 nm) were produced and assessed for particle characteristics and further for toxicity and performance in vitro. Results: Rapa-SLNs of 200 nm inhibited proliferation in TSC2-negative mouse embryonic fibroblast cells and penetrated the respiratory epithelium and lymphatic endothelium significantly faster compared with free rapamycin and larger Rapa-SLNs. Conclusion: Rapa-SLN approximately 200 nm allows efficient entry of rapamycin into the lymphatic system and is therefore a promising treatment for LAM patients.
Collapse
Affiliation(s)
- Emelie Landh
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Lyn M Moir
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Peta Bradbury
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| |
Collapse
|
20
|
Yang T, Yang WX. The dynamics and regulation of microfilament during spermatogenesis. Gene 2020; 744:144635. [PMID: 32244053 DOI: 10.1016/j.gene.2020.144635] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/28/2020] [Accepted: 03/30/2020] [Indexed: 12/22/2022]
Abstract
Spermatogenesis is a highly complex physiological process which contains spermatogonia proliferation, spermatocyte meiosis and spermatid morphogenesis. In the past decade, actin binding proteins and signaling pathways which are critical for regulating the actin cytoskeleton in testis had been found. In this review, we summarized 5 actin-binding proteins that have been proven to play important roles in the seminiferous epithelium. Lack of them perturbs spermatids polarity and the transport of spermatids. The loss of Arp2/3 complex, Formin1, Eps8, Palladin and Plastin3 cause sperm release failure suggesting their irreplaceable role in spermatogenesis. Actin regulation relies on multiple signal pathways. The PI3K/Akt signaling pathway positively regulate the mTOR pathway to promote actin reorganization in seminiferous epithelium. Conversely, TSC1/TSC2 complex, the upstream of mTOR, is activated by the LKB1/AMPK pathway to inhibit cell proliferation, differentiation and migration. The increasing researches focus on the function of actin binding proteins (ABPs), however, their collaborative regulation of actin patterns and potential regulatory signaling networks remains unclear. We reviewed ABPs that play important roles in mammalian spermatogenesis and signal pathways involved in the regulation of microfilaments. We suggest that more relevant studies should be performed in the future.
Collapse
Affiliation(s)
- Tong Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
21
|
Lu Y, Liu X, Zhang E, Kopras EJ, Smith EP, Astreinidis A, Li C, Leung YK, Ho SM, Yu JJ. Estrogen activates pyruvate kinase M2 and increases the growth of TSC2-deficient cells. PLoS One 2020; 15:e0228894. [PMID: 32078667 PMCID: PMC7032738 DOI: 10.1371/journal.pone.0228894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/24/2020] [Indexed: 01/15/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a devastating lung disease caused by inactivating gene mutations in either TSC1 or TSC2 that result in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). As LAM occurs predominantly in women during their reproductive age and is exacerbated by pregnancy, the female hormonal environment, and in particular estrogen, is implicated in LAM pathogenesis and progression. However, detailed underlying molecular mechanisms are not well understood. In this study, utilizing human pulmonary LAM specimens and cell culture models of TSC2-deficient LAM patient-derived and rat uterine leiomyoma-derived cells, we tested the hypothesis that estrogen promotes the growth of mTORC1-hyperactive cells through pyruvate kinase M2 (PKM2). Estrogen increased the phosphorylation of PKM2 at Ser37 and induced the nuclear translocation of phospho-PKM2. The estrogen receptor antagonist Faslodex reversed these effects. Restoration of TSC2 inhibited the phosphorylation of PKM2 in an mTORC1 inhibitor-insensitive manner. Finally, accumulation of phosphorylated PKM2 was evident in pulmonary nodule from LAM patients. Together, our data suggest that female predominance of LAM might be at least in part attributed to estrogen stimulation of PKM2-mediated cellular metabolic alterations. Targeting metabolic regulators of PKM2 might have therapeutic benefits for women with LAM and other female-specific neoplasms.
Collapse
Affiliation(s)
- Yiyang Lu
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
| | - Xiaolei Liu
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
| | - Erik Zhang
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
| | - Elizabeth J. Kopras
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
| | - Eric P. Smith
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
| | - Aristotelis Astreinidis
- Division of Pediatric Nephrology, Department of Pediatrics, College of Medicine, University of Tennessee Health Sciences Center and Tuberous Sclerosis Complex Center of Excellence, Le Bonheur Children’s Hospital, Memphis, TN, United States of America
| | - Chenggang Li
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Yuet-Kin Leung
- College of Medicine Department of Pharmacology and Toxicology, the University of Arkansas for Medical Science (UAMS), Little Rock, AR, United States of America
| | - Shuk-Mei Ho
- College of Medicine Department of Pharmacology and Toxicology, the University of Arkansas for Medical Science (UAMS), Little Rock, AR, United States of America
| | - Jane J. Yu
- University of Cincinnati College of Medicine, Department of Internal Medicine, Cincinnati, OH, United States of America
| |
Collapse
|
22
|
Krymskaya VP, Courtwright AM, Fleck V, Dorgan D, Kotloff R, McCormack FX, Kreider M. A phase II clinical trial of the Safety Of Simvastatin (SOS) in patients with pulmonary lymphangioleiomyomatosis and with tuberous sclerosis complex. Respir Med 2020; 163:105898. [PMID: 32125970 DOI: 10.1016/j.rmed.2020.105898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 01/20/2023]
Abstract
INRODUCTION The mechanistic target of rapamycin inhibitors (mTORi) sirolimus and everolimus stabilize lung function in patients with pulmonary lymphangioleiomyomatosis (LAM) but do not induce remission. Pre-clinical studies suggest that simvastatin in combination with sirolimus induces LAM cell death. The objective of this study was to assess the safety of simvastatin with either sirolimus or everolimus in LAM patients. METHODS This was a phase II single arm trial evaluating the safety of escalating daily simvastatin (20-40 mg) in LAM patients already treated with sirolimus or everolimus. Adverse events and changes in lipid panel profile, pulmonary function tests, and VEGF-D were assessed. RESULTS Ten LAM patients on a stable dose of mTORi for >3 months were treated with 20 mg simvastatin for two months followed by 40 mg for two months. The most common adverse events were peripheral edema (30%), cough (30%), and diarrhea (30%). No patients withdrew or had a reduction in simvastatin dose because of adverse events. Two patients required sirolumus dose reduction for supratherapeutic trough levels following simvastatin initiation. Total cholesterol and low density lipoproteins declined over the study period (-46.0 mg/dL±20.8, p = 0.008; -41.9 mg/dL±22.0, p = 0.01, respectively). There was also a decline in FEV1 (-82.0 mL±86.4, p = 0.02) but no significant change in FVC, DLCO, or VEGF-D. CONCLUSIONS The combination of simvastatin with mTORi in LAM patients is safe and well-tolerated from an adverse events perspective. The addition of simvastatin, however, was associated with decline in FEV1 and the efficacy of this combination should be explored in larger trials.
Collapse
Affiliation(s)
- Vera P Krymskaya
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrew M Courtwright
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, Philadelphia, PA, USA
| | - Victoria Fleck
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Dorgan
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, Philadelphia, PA, USA
| | - Robert Kotloff
- Department of Pulmonary Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Maryl Kreider
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Inhaled rapamycin solid lipid nano particles for the treatment of Lymphangioleiomyomatosis. Eur J Pharm Sci 2020; 142:105098. [DOI: 10.1016/j.ejps.2019.105098] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 01/03/2023]
|
24
|
Liu J, Zhao W, Ou X, Zhao Z, Hu C, Sun M, Liu F, Deng J, Gu W, An J, Zhang Q, Zhang X, Xie J, Li S, Chen R, Yu S, Zhong N. Mutation spectrums of TSC1 and TSC2 in Chinese women with lymphangioleiomyomatosis (LAM). PLoS One 2019; 14:e0226400. [PMID: 31856217 PMCID: PMC6922431 DOI: 10.1371/journal.pone.0226400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/26/2019] [Indexed: 11/19/2022] Open
Abstract
The aim of our study was to elucidate the landscapes of genetic alterations of TSC1 and TSC2 as well as other possible non-TSC1/2 in Lymphangioleiomyomatosis (LAM) patients. Sixty-one Chinese LAM patients’ clinical information was collected. Tumor biopsies and matched leukocytes from these patients were retrospectively analyzed by next generation sequencing (NGS), chromosomal microarray analysis (CMA), and multiplex ligation-dependent probe amplification (MLPA). Eighty-six TSC1/2 variants were identified in 46 of the 61 LAM patients (75.4%) in which TSC2 and TSC1 variants were 88.37% and 11.63% respectively. The 86 variants are composed of (i) 52 single nucleotide variants (SNVs) (including 30 novel variants), (ii) 23 indels (including 21deletions, and 2 insertions), (iii) a germline duplication of exon 31–42 of TSC2, (iv) a 2.68 Mb somatic duplication containing TSC2, and (v) 9 regions with copy-neutral loss of heterogeneity (CN-LOHs) present only in the LAM patients with single TSC1/2 mutations. Sixty-one non-TSC1/2 variants in 31 genes were identified in 37 LAM patients. Combined applications of different techniques are necessary to achieve maximal detection rate of TSC1/2 variants in LAM patients. Thirty novel TSC1/2 variants expands the spectrum of TSC1/2 in LAM patients. Identification of 61 non-TSC1/2 variants suggests that alternative genes might have contributed to the initiation and progression of LAM.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Weiwei Zhao
- Guangzhou KingMed Diagnostics Group Co., Ltd, Guangzhou, Guangdong, China
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
- KingMed College of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou KingMed Translational Medicine Institute Co., Ltd, Guangzhou, Guangdong, China
| | - Xiaohua Ou
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
- Guangzhou KingMed Translational Medicine Institute Co., Ltd, Guangzhou, Guangdong, China
| | - Zhen Zhao
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
- Guangzhou KingMed Translational Medicine Institute Co., Ltd, Guangzhou, Guangdong, China
| | - Changming Hu
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
| | - Mingming Sun
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
- Guangzhou KingMed Translational Medicine Institute Co., Ltd, Guangzhou, Guangdong, China
| | - Feifei Liu
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
- Guangzhou KingMed Translational Medicine Institute Co., Ltd, Guangzhou, Guangdong, China
| | - Junhao Deng
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
| | - Weili Gu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
| | - Jiaying An
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
| | - Qingling Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiaoxian Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
| | - Jiaxing Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Shiyue Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Shihui Yu
- Guangzhou KingMed Diagnostics Group Co., Ltd, Guangzhou, Guangdong, China
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong, China
- KingMed College of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou KingMed Translational Medicine Institute Co., Ltd, Guangzhou, Guangdong, China
- KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, Guangdong, China
- * E-mail: (SY); (NZ)
| | - Nanshan Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Institute for Respiratory Health, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- * E-mail: (SY); (NZ)
| |
Collapse
|
25
|
Koul PA, Mehfooz N. Sirolimus in lymphangioleiomyomatosis: A case in point for research in 'orphan' diseases. Lung India 2019; 36:353-355. [PMID: 31290424 PMCID: PMC6625251 DOI: 10.4103/lungindia.lungindia_280_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Parvaiz A Koul
- Department of Internal and Pulmonary Medicine, Sheri Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Nazia Mehfooz
- Department of Pulmonary Medicine, Sheri Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
26
|
Harari S, Spagnolo P, Cocconcelli E, Luisi F, Cottin V. Recent advances in the pathobiology and clinical management of lymphangioleiomyomatosis. Curr Opin Pulm Med 2019; 24:469-476. [PMID: 29927757 DOI: 10.1097/mcp.0000000000000502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Lymphangioleiomyomatosis (LAM) is a rare systemic disease that occurs almost exclusively in women. In the last few years, our understanding of disease pathobiology has improved substantially; in addition, a guideline document has recently been developed that provides recommendations for the diagnosis and clinical management of patients with LAM. Yet, significant gaps in knowledge remain. RECENT FINDINGS Groundbreaking insights into the cellular biochemistry of LAM have led to the reclassification of the disease as a low-grade, destructive, metastasizing neoplasm. In addition, recent data confirm the potential of next-generation sequencing to detect low-prevalence mutations in tuberous sclerosis (TSC) genes in sporadic LAM. A randomized, double-blind, multicentre trial has confirmed the efficacy of sirolimus in stabilizing lung function, improving functional performance and quality of life, and reducing lymphatic manifestations in patients with LAM. Accordingly, recent guidelines issued by the American Thoracic Society and the Japanese Respiratory Society recommend sirolimus treatment for patients with LAM and reduced lung function. Uncertainty remains, however, with regard to patient selection, and timing of initiation, duration and dosing of treatment. SUMMARY Significant advances have been made in the diagnosis and clinical management of patients with LAM. However, additional studies are needed to assess long-term safety and efficacy of sirolimus therapy, and to identify predictors of disease behaviour and response to treatment.
Collapse
Affiliation(s)
- Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, IRCCS MultiMedica, Milan
| | - Paolo Spagnolo
- Section of Respiratory Diseases, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Elisabetta Cocconcelli
- Section of Respiratory Diseases, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Francesca Luisi
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, IRCCS MultiMedica, Milan
| | - Vincent Cottin
- Hospices Civils de Lyon, Louis Pradel Hospital, National Reference Center for Rare Pulmonary Diseases, Department of Respiratory Diseases, Claude Bernard University Lyon 1, UMR754, Lyon, France
| |
Collapse
|
27
|
Rostamzadeh D, Yousefi M, Haghshenas MR, Ahmadi M, Dolati S, Babaloo Z. mTOR Signaling pathway as a master regulator of memory CD8 + T-cells, Th17, and NK cells development and their functional properties. J Cell Physiol 2019; 234:12353-12368. [PMID: 30710341 DOI: 10.1002/jcp.28042] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/03/2018] [Indexed: 12/27/2022]
Abstract
The mammalian target of rapamycin (mTOR) is a member of the evolutionary phosphatidylinositol kinase-related kinases (PIKKs). mTOR plays a pivotal role in the regulation of diverse aspects of cellular physiology such as body metabolism, cell growth, protein synthesis, cell size, autophagy, and cell differentiation. Immunologically, mTOR has a fundamental part in controlling and shaping diverse functions of innate and adaptive immune cells, in particular, T-cell subsets differentiation, survival, and metabolic reprogramming to ultimately regulate the fate of diverse immune cell types. Researchers report that rapamycin, a selective mTOR inhibitor, and immunosuppressive agent, has surprising immunostimulatory effects on inducing both quantitative and qualitative aspects of virus-specific memory CD8+ T-cells differentiation and homeostasis in a T-cell-intrinsic manner. The mTOR signaling pathway also plays a critical role in dictating the outcome of regulatory T cells (Treg), T helper 17 (Th17) cells, and natural killer (NK) cells proliferation and maturation, as well as the effector functions and cytotoxic properties of NK cells. Manipulation of mTOR activity is a critical therapeutic approach for pharmacological agents that seek to inhibit mTOR. This approach should enhance specific memory CD8 + T-cells responses and induce fully functional effector properties of NK cells to provoke their antitumor and antiviral activities.
Collapse
Affiliation(s)
- Davood Rostamzadeh
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Yousefi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Ahmadi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Unit, Drug Applied Research Center, Tabriz University of Medical Sciences.,Head of Immunology Department, Medicine Faculty, Tabriz University of Medical Science
| |
Collapse
|
28
|
Zhao DD, Yuan J, Cheng Q, Qi YL, Lu K, Lai SS, Sun Q, Zhao Y, Fang L, Jin ML, Yu DC, Qiu YD, Li CJ, Chen J, Xue B. Evidence for a role of geranylgeranylation in renal angiomyolipoma and renal epithelioid angiomyolipoma. Oncol Lett 2019; 17:1523-1530. [PMID: 30675208 PMCID: PMC6341897 DOI: 10.3892/ol.2018.9808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 10/19/2018] [Indexed: 01/17/2023] Open
Abstract
Research on mevalonate kinase deficiency has revealed that it may lead to the development of renal angiomyolipomas (RAMLs). Thus, it was suspected that geranylgeranyl pyrophosphate synthase (GGPPS), a key enzyme in the mevalonate pathway, may be involved in the development of RAMLs. In the present study, the expression of GGPPS in RAMLs and renal epithelioid angiomyolipomas (REAs) was assessed, and paraffin embedded specimens from 60 patients, including 9 cases with REA and 51 cases with RAML, were examined. Immunoreactivity was evaluated semi-quantitatively according to the intensity of staining and the percentage of positively stained cells. The results indicated that GGPPS was predominantly present in the cytoplasm, and REA tissues exhibited higher expression of GGPPS in the cytoplasm compared with RAML tissues. It was also identified that GGPPS was upregulated in TSC2-null cells, and inhibition of GGPPS could induce apoptosis of TSC2-null cells by autophagy. In conclusion, the increased expression of GGPPS in RAMLs and REAs indicated that mevalonate pathways may be involved in disease progression. GGPPS may serve as a potential therapeutic target and the current results may provide a novel therapeutic strategy for RAML and lymphangioleiomyomatosis.
Collapse
Affiliation(s)
- Dan-Dan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Jun Yuan
- Biochemical and Environmental Engineering School of Xiaozhuang College, Nanjing 211171, P.R. China
| | - Qi Cheng
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Ya-Ling Qi
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Ke Lu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Shan-Shan Lai
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Qian Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Yue Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Liver Disease Collaborative Research Platform of Medical School of Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Liver Disease Collaborative Research Platform of Medical School of Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Mei-Ling Jin
- Pulmonary Department, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - De-Cai Yu
- Liver Disease Collaborative Research Platform of Medical School of Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Yu-Dong Qiu
- Liver Disease Collaborative Research Platform of Medical School of Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Chao-Jun Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Jun Chen
- Liver Disease Collaborative Research Platform of Medical School of Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Department of Pathology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Bin Xue
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,Liver Disease Collaborative Research Platform of Medical School of Nanjing University, Nanjing, Jiangsu 210093, P.R. China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
29
|
Kim SW, Kim HI, Thapa B, Nuwormegbe S, Lee K. Critical Role of mTORC2-Akt Signaling in TGF-β1-Induced Myofibroblast Differentiation of Human Pterygium Fibroblasts. ACTA ACUST UNITED AC 2019; 60:82-92. [DOI: 10.1167/iovs.18-25376] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Sun Woong Kim
- Department of Ophthalmology, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Hye-In Kim
- Department of Biomedical Science, Hallym University, Chuncheon, Korea
| | - Bikash Thapa
- Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Selikem Nuwormegbe
- Department of Ophthalmology, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Keunwook Lee
- Department of Biomedical Science, Hallym University, Chuncheon, Korea
- Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| |
Collapse
|
30
|
Jones AT, Narov K, Yang J, Sampson JR, Shen MH. Efficacy of Dual Inhibition of Glycolysis and Glutaminolysis for Therapy of Renal Lesions in Tsc2 +/- Mice. Neoplasia 2019; 21:230-238. [PMID: 30622053 PMCID: PMC6324218 DOI: 10.1016/j.neo.2018.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022] Open
Abstract
Tuberous sclerosis is caused by mutations in the TSC1 or TSC2 gene and characterized by development of tumors in multiple organs including the kidneys. TSC-associated tumors exhibit somatic loss of the second allele of the TSC genes, leading to aberrant activation of the mechanistic target of rapamycin (mTOR) signaling pathway. Activation of mTOR complex 1 (mTORC1) causes addiction to glucose and glutamine in Tsc1−/−or Tsc2−/− mouse embryonic fibroblasts (MEFs). Blocking of glutamine anaplerosis in combination with glycolytic inhibition causes significant cell death in Tsc2−/− but not Tsc2+/+ MEFs. In this study, we tested efficacy of dual inhibition of glycolysis with 3-BrPA and glutaminolysis with CB-839 for renal tumors in Tsc2+/− mice. Following 2 months of treatment of Tsc2+/− mice from the age of 12 months, combination of 3-BrPA and CB-839 significantly reduced overall size and cellular areas of all renal lesions (cystic/papillary adenomas and solid carcinomas), but neither alone did. Combination of 3-BrPA and CB-839 inhibited mTORC1 and the proliferation of tumor cells but did not increase apoptosis. However, combination of 3-BrPA and CB-839 was not as efficacious as rapamycin alone or rapamycin in combination with either 3-BrPA or CB-839 for renal lesions of Tsc2+/− mice. Consistently, rapamycin alone or rapamycin in combination with either 3-BrPA or CB-839 had stronger inhibitory effects on mTORC1 and proliferation of tumor cells than combination of 3-BrPA and CB-839. We conclude that combination of 3-BRPA and CB-839 may not offer a better therapeutic strategy than rapamycin for TSC-associated tumors.
Collapse
Affiliation(s)
- Ashley T Jones
- Institute of Medical Genetics, Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Kalin Narov
- Institute of Medical Genetics, Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Jian Yang
- Institute of Medical Genetics, Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Julian R Sampson
- Institute of Medical Genetics, Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Ming Hong Shen
- Institute of Medical Genetics, Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
31
|
Tang H, Wu K, Wang J, Vinjamuri S, Gu Y, Song S, Wang Z, Zhang Q, Balistrieri A, Ayon RJ, Rischard F, Vanderpool R, Chen J, Zhou G, Desai AA, Black SM, Garcia JGN, Yuan JXJ, Makino A. Pathogenic Role of mTORC1 and mTORC2 in Pulmonary Hypertension. JACC Basic Transl Sci 2018; 3:744-762. [PMID: 30623134 PMCID: PMC6314964 DOI: 10.1016/j.jacbts.2018.08.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/23/2018] [Accepted: 08/16/2018] [Indexed: 01/07/2023]
Abstract
G protein-coupled receptors and tyrosine kinase receptors signal through the phosphoinositide 3-kinase/Akt/mTOR pathway to induce cell proliferation, survival, and growth. mTOR is a kinase present in 2 functionally distinct complexes, mTORC1 and mTORC2. Functional disruption of mTORC1 by knockout of Raptor (regulatory associated protein of mammalian target of rapamycin) in smooth muscle cells ameliorated the development of experimental PH. Functional disruption of mTORC2 by knockout of Rictor (rapamycin insensitive companion of mammalian target of rapamycin) caused spontaneous PH by up-regulating platelet-derived growth factor receptors. Use of mTOR inhibitors (e.g., rapamycin) to treat PH should be accompanied by inhibitors of platelet-derived growth factor receptors (e.g., imatinib).
Concentric lung vascular wall thickening due to enhanced proliferation of pulmonary arterial smooth muscle cells is an important pathological cause for the elevated pulmonary vascular resistance reported in patients with pulmonary arterial hypertension. We identified a differential role of mammalian target of rapamycin (mTOR) complex 1 and complex 2, two functionally distinct mTOR complexes, in the development of pulmonary hypertension (PH). Inhibition of mTOR complex 1 attenuated the development of PH; however, inhibition of mTOR complex 2 caused spontaneous PH, potentially due to up-regulation of platelet-derived growth factor receptors in pulmonary arterial smooth muscle cells, and compromised the therapeutic effect of the mTOR inhibitors on PH. In addition, we describe a promising therapeutic strategy using combination treatment with the mTOR inhibitors and the platelet-derived growth factor receptor inhibitors on PH and right ventricular hypertrophy. The data from this study provide an important mechanism-based perspective for developing novel therapies for patients with pulmonary arterial hypertension and right heart failure.
Collapse
Key Words
- EC, endothelial cell
- FOXO3a, Forkhead box O3a
- GPCR, G protein-coupled receptor
- HPH, hypoxia-induced pulmonary hypertension
- PA, pulmonary artery
- PAEC, pulmonary arterial endothelial cell
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary arterial smooth muscle cell
- PDGF, platelet-derived growth factor
- PDGFR, platelet-derived growth factor receptor
- PH, pulmonary hypertension
- PI3K, phosphoinositide 3-kinase
- PTEN, phosphatase and tensin homolog
- PVR, pulmonary vascular resistance
- RVH, right ventricular hypertrophy
- RVSP, right ventricular systolic pressure
- Raptor
- Raptor, regulatory associated protein of mammalian target of rapamycin
- Rictor
- Rictor, rapamycin insensitive companion of mammalian target of rapamycin
- SM, smooth muscle
- TKR, tyrosine kinase receptor
- WT, wild-type
- mTOR
- mTORC1, mammalian target of rapamycin complex 1
- mTORC2, mammalian target of rapamycin complex 2
- pAKT, phosphorylated AKT
- pulmonary hypertension
- right ventricle
Collapse
Affiliation(s)
- Haiyang Tang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kang Wu
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sujana Vinjamuri
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Yali Gu
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Shanshan Song
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ziyi Wang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Zhang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Angela Balistrieri
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Franz Rischard
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Rebecca Vanderpool
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jiwang Chen
- Department of Pediatrics, University of Illinois College of Medicine, Chicago, Illinois
| | - Guofei Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pediatrics, University of Illinois College of Medicine, Chicago, Illinois
| | - Ankit A Desai
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Division of Cardiology, Department of Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Joe G N Garcia
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona.,Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ayako Makino
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
32
|
Yoon HY, Hwang JJ, Kim DS, Song JW. Efficacy and safety of low-dose Sirolimus in Lymphangioleiomyomatosis. Orphanet J Rare Dis 2018; 13:204. [PMID: 30428897 PMCID: PMC6236936 DOI: 10.1186/s13023-018-0946-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/30/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Lymphangioleiomyomatosis is a rare disease caused by unregulated activation of mammalian target of rapamycin (mTOR) signalling pathway. Sirolimus showed efficacy in a phase 3 trial of patients with lymphangioleiomyomatosis, but the optimal dose remains unclear. METHODS We investigated the efficacy and safety of low-dose compared with conventional-dose sirolimus. Clinical data of 39 patients with lymphangioleiomyomatosis (mean age, 34.8 years; median treatment period, 29.6 months) who received sirolimus were retrospectively reviewed. Low-dose sirolimus was defined as any dose that maintained mean blood trough levels lower than those maintained with conventional doses (5-15 ng/mL). RESULTS Fifty-one percent of patients received low-dose therapy. The rate of decline in lung function decreased after treatment in the whole group (forced expiratory volume in 1 s [FEV1], - 0.12 ± 0.47 [before] vs. 0.24 ± 0.48% predicted/month [after], p = 0.027; diffusing capacity for carbon monoxide [DLco], - 0.33 ± 0.61 vs. 0.03 ± 0.26% predicted/month, p = 0.006) compared with before treatment. In the low-dose group, the rate of decline in FEV1 (- 0.08 ± 0.38 [before] vs. 0.19 ± 0.51% predicted/month [after], p = 0.264) and DLco (-0.13 ± 0.62 vs. 0.02 ± 0.28% predicted/month, p = 0.679) showed a numeric trend towards improvement after treatment; however, the conventional-dose group showed significant improvement in FEV1 (- 0.26 ± 0.54 [before] vs. 0.22 ± 0.38 [after] % predicted/month, p = 0.024) and DLco (- 0.55 ± 0.58 vs. 0.04 ± 0.25% predicted/month, p = 0.002) after treatment. Adverse events (AEs) occurred in 89.7% of patients and the most common AEs was hypercholesterolaemia (43.6%), followed by stomatitis (35.9%). The occurrences of AE were similar between the low- and conventional-dose groups (85.0% vs. 94.7%, p = 0.605). CONCLUSIONS Low-dose sirolimus may stabilise lung function decline in lymphangioleiomyomatosis patients, but its efficacy appears to be inferior to that of conventional-dose sirolimus.
Collapse
Affiliation(s)
- Hee-Young Yoon
- Departments of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jung Jin Hwang
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Dong Soon Kim
- Departments of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jin Woo Song
- Departments of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
33
|
Identification of MΦ specific POTEE expression: Its role in mTORC2 activation via protein-protein interaction in TAMs. Cell Immunol 2018; 335:30-40. [PMID: 30420269 DOI: 10.1016/j.cellimm.2018.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 01/28/2023]
Abstract
POTE is known as cancer antigen, expressed in many cancers, along with very few normal tissues like prostate, ovary, testes and embryo. Till date, POTEE identified as majorly expressed POTE paralog. Functionally, POTEF regulates TLR signaling which play important role in innate immunity provided clue about expression of POTE in immune cells. We have chosen three Thp1monocytes, Jurkat T1 and MΦ cells as a model. Here, first time we report expression of POTEE in immune cells specifically only in MΦ but not in monocytes or T-cells. In addition, expression level remains unaltered in MΦ subtypes M1 and M2 and MΦ subjected to various stresses, except MΦs treated with Hyp-CM where MΦs acquires properties of TAMs. In TAMs, POTEE was involved differential protein-protein interaction with mTOR, RICTOR, and Rad51 indicating its biological role in cell invasion through mTORC2 activation. siRNA mediated knockdown of POTEE suggests its importance in cell survival of MΦs as well as TAMs.
Collapse
|
34
|
Vekariya U, Saxena R, Singh P, Rawat K, Kumar B, Kumari S, Agnihotri SK, Kaur S, Sachan R, Nazir A, Bhadauria S, Sachdev M, Tripathi RK. HIV-1 Nef-POTEE; A novel interaction modulates macrophage dissemination via mTORC2 signaling pathway. Life Sci 2018; 214:158-166. [PMID: 30391463 DOI: 10.1016/j.lfs.2018.10.068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/29/2018] [Indexed: 12/16/2022]
Abstract
AIMS Human immunodeficiency virus -1 [HIV-1] Nef, localizes in different cellular compartments and modulates several cellular pathways. Nef promotes virus pathogenicity through alteration in cell surface receptor expression, apoptosis, protein trafficking etc. Nef regulates viral pathogenesis through interaction with different host proteins. Thus, molecular mechanisms of pathogenesis could be deciphered by identifying novel Nef interacting proteins. MAIN METHODS HIV-1 Nef interacting proteins were identified by pull down assay and MALDI-TOF analysis. The interaction was further validated through mammalian two hybrid assay. Functional role of this interaction was identified by immunoprecipitation assay, cell invasion and cell migration studies. Fold Change in mRNA levels of CD163, CD206, CCL17 and CCL18 was analyzed using qPCR. KEY FINDINGS In current study, C. elegans protein ACT4C and its human homolog POTEE was identified to be interacting with Nef. This interaction activates mTORC2 complex, which in-turn activates AKT and PKC-α. The activation of mTORC2 complex was found to be initiated by the interaction of Nef, mTORC2, Rictor to POTEE. The cellular phenotype and functions affected by Nef-POTEE interaction resulted in significant increase in cell invasion and migration of macrophages (MΦ). SIGNIFICANCE MΦ is primary target of HIV-1 infection where HIV-1 replicates and polarizes immunosuppressive M2 phenotype. Combine effect of M2 phenotype and Viral-host protein interactions compromise the MΦ associated physiological functions. Infected MΦ dissemination into other system also leads to HIV-1 induced malignancies. Therefore, targeting POTEE-Nef interaction can lead to formulating better therapeutic strategy against HIV-1.
Collapse
Affiliation(s)
- Umeshkumar Vekariya
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Reshu Saxena
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Poonam Singh
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Kavita Rawat
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Balawant Kumar
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Sushila Kumari
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | | | - Supinder Kaur
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Rekha Sachan
- Department of Obstetrics & Gynecology, King George Medical University, Lucknow, UP, India
| | - Aamir Nazir
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Smrati Bhadauria
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Monika Sachdev
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Raj Kamal Tripathi
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, UP, India.
| |
Collapse
|
35
|
Malik N, Sansom OJ, Michie AM. The role of mTOR-mediated signals during haemopoiesis and lineage commitment. Biochem Soc Trans 2018; 46:1313-1324. [PMID: 30154096 PMCID: PMC6195642 DOI: 10.1042/bst20180141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
The serine/threonine protein kinase mechanistic target of rapamycin (mTOR) has been implicated in the regulation of an array of cellular functions including protein and lipid synthesis, proliferation, cell size and survival. Here, we describe the role of mTOR during haemopoiesis within the context of mTORC1 and mTORC2, the distinct complexes in which it functions. The use of conditional transgenic mouse models specifically targeting individual mTOR signalling components, together with selective inhibitors, have generated a significant body of research emphasising the critical roles played by mTOR, and individual mTOR complexes, in haemopoietic lineage commitment and development. This review will describe the profound role of mTOR in embryogenesis and haemopoiesis, underscoring the importance of mTORC1 at the early stages of haemopoietic cell development, through modulation of stem cell potentiation and self-renewal, and erythroid and B cell lineage commitment. Furthermore, the relatively discrete role of mTORC2 in haemopoiesis will be explored during T cell development and B cell maturation. Collectively, this review aims to highlight the functional diversity of mTOR signalling and underline the importance of this pathway in haemopoiesis.
Collapse
Affiliation(s)
- Natasha Malik
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Owen J Sansom
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, U.K
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K.
| |
Collapse
|
36
|
Lam HC, Siroky BJ, Henske EP. Renal disease in tuberous sclerosis complex: pathogenesis and therapy. Nat Rev Nephrol 2018; 14:704-716. [DOI: 10.1038/s41581-018-0059-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
37
|
Gui Y, Li J, Lu Q, Feng Y, Wang M, He W, Yang J, Dai C. Yap/Taz mediates mTORC2-stimulated fibroblast activation and kidney fibrosis. J Biol Chem 2018; 293:16364-16375. [PMID: 30154246 DOI: 10.1074/jbc.ra118.004073] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/07/2018] [Indexed: 11/06/2022] Open
Abstract
Our previously published study demonstrated that mammalian target of rapamycin complex 2 (mTORC2) signaling mediates TGFβ1-induced fibroblast activation. However, the underlying mechanisms for mTORC2 in stimulating fibroblast activation remain poorly understood. Here, we found that TGFβ1 could stimulate mTORC2 and Yap/Taz activation in NRK-49F cells. Blocking either mTORC2 or Yap/Taz signaling diminished TGFβ1-induced fibroblast activation. In addition, blockade of mTORC2 could down-regulate the expression of Yap/Taz, connective tissue growth factor (CTGF), and ankyrin repeat domain 1 (ANKRD1). Overexpression of constitutively active Taz (Taz-S89A) could restore fibroblast activation suppressed by PP242, an mTOR kinase inhibitor in NRK-49F cells. In mouse kidneys with unilateral ureter obstructive (UUO) nephropathy, both mTORC2 and Yap/Taz were activated in the interstitial myofibroblasts. Ablation of Rictor in fibroblasts/pericytes or blockade of mTOR signaling with PP242 attenuated Yap/Taz activation and UUO nephropathy in mice. Together, this study uncovers that targeting mTORC2 retards fibroblast activation and kidney fibrosis through suppressing Yap/Taz activation.
Collapse
Affiliation(s)
- Yuan Gui
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Jianzhong Li
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Qingmiao Lu
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Ye Feng
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Mingjie Wang
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Weichun He
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Junwei Yang
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| | - Chunsun Dai
- From the Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu 210003, China
| |
Collapse
|
38
|
Biological Aspects of mTOR in Leukemia. Int J Mol Sci 2018; 19:ijms19082396. [PMID: 30110936 PMCID: PMC6121663 DOI: 10.3390/ijms19082396] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a central processor of intra- and extracellular signals, regulating many fundamental cellular processes such as metabolism, growth, proliferation, and survival. Strong evidences have indicated that mTOR dysregulation is deeply implicated in leukemogenesis. This has led to growing interest in the development of modulators of its activity for leukemia treatment. This review intends to provide an outline of the principal biological and molecular functions of mTOR. We summarize the current understanding of how mTOR interacts with microRNAs, with components of cell metabolism, and with controllers of apoptotic machinery. Lastly, from a clinical/translational perspective, we recapitulate the therapeutic results in leukemia, obtained by using mTOR inhibitors as single agents and in combination with other compounds.
Collapse
|
39
|
Zhao T, Li R, Tan X, Zhang J, Fan C, Zhao Q, Deng Y, Xu A, Lukong KE, Genth H, Xiang J. Simulated Microgravity Reduces Focal Adhesions and Alters Cytoskeleton and Nuclear Positioning Leading to Enhanced Apoptosis via Suppressing FAK/RhoA-Mediated mTORC1/NF-κB and ERK1/2 Pathways. Int J Mol Sci 2018; 19:ijms19071994. [PMID: 29986550 PMCID: PMC6073227 DOI: 10.3390/ijms19071994] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 12/16/2022] Open
Abstract
Simulated-microgravity (SMG) promotes cell-apoptosis. We demonstrated that SMG inhibited cell proliferation/metastasis via FAK/RhoA-regulated mTORC1 pathway. Since mTORC1, NF-κB, and ERK1/2 signaling are important in cell apoptosis, we examined whether SMG-enhanced apoptosis is regulated via these signals controlled by FAK/RhoA in BL6-10 melanoma cells under clinostat-modelled SMG-condition. We show that SMG promotes cell-apoptosis, alters cytoskeleton, reduces focal adhesions (FAs), and suppresses FAK/RhoA signaling. SMG down-regulates expression of mTORC1-related Raptor, pS6K, pEIF4E, pNF-κB, and pNF-κB-regulated Bcl2, and induces relocalization of pNF-κB from the nucleus to the cytoplasm. In addition, SMG also inhibits expression of nuclear envelope proteins (NEPs) lamin-A, emerin, sun1, and nesprin-3, which control nuclear positioning, and suppresses nuclear positioning-regulated pERK1/2 signaling. Moreover, rapamycin, the mTORC1 inhibitor, also enhances apoptosis in cells under 1 g condition via suppressing the mTORC1/NF-κB pathway. Furthermore, the FAK/RhoA activator, toxin cytotoxic necrotizing factor-1 (CNF1), reduces cell apoptosis, restores the cytoskeleton, FAs, NEPs, and nuclear positioning, and converts all of the above SMG-induced changes in molecular signaling in cells under SMG. Therefore, our data demonstrate that SMG reduces FAs and alters the cytoskeleton and nuclear positioning, leading to enhanced cell apoptosis via suppressing the FAK/RhoA-regulated mTORC1/NF-κB and ERK1/2 pathways. The FAK/RhoA regulatory network may, thus, become a new target for the development of novel therapeutics for humans under spaceflight conditions with stressed physiological challenges, and for other human diseases.
Collapse
Affiliation(s)
- Tuo Zhao
- School of Life Sciences, Beijing Institute of Technology, Beijing 10081, China.
| | - Rong Li
- Cancer Research, Saskatchewan Cancer Agency, Saskatoon, SK S7N 4H4, Canada.
- Department of Oncology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Xin Tan
- School of Life Sciences, Beijing Institute of Technology, Beijing 10081, China.
| | - Jun Zhang
- School of Life Sciences, Beijing Institute of Technology, Beijing 10081, China.
| | - Cuihong Fan
- School of Life Sciences, Beijing Institute of Technology, Beijing 10081, China.
| | - Qin Zhao
- School of Life Sciences, Beijing Institute of Technology, Beijing 10081, China.
| | - Yulin Deng
- School of Life Sciences, Beijing Institute of Technology, Beijing 10081, China.
| | - Aizhang Xu
- Cancer Research, Saskatchewan Cancer Agency, Saskatoon, SK S7N 4H4, Canada.
- Department of Oncology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Kiven Erique Lukong
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Harald Genth
- Institute of Toxicology, Hannover Medical School, D-30625 Hannover, Germany.
| | - Jim Xiang
- Cancer Research, Saskatchewan Cancer Agency, Saskatoon, SK S7N 4H4, Canada.
- Department of Oncology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
40
|
Himes BE, Obraztsova K, Lian L, Shumyatcher M, Rue R, Atochina-Vasserman EN, Hur SK, Bartolomei MS, Evans JF, Krymskaya VP. Rapamycin-independent IGF2 expression in Tsc2-null mouse embryo fibroblasts and human lymphangioleiomyomatosis cells. PLoS One 2018; 13:e0197105. [PMID: 29758070 PMCID: PMC5951544 DOI: 10.1371/journal.pone.0197105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/26/2018] [Indexed: 11/19/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, almost exclusively female lung disease linked to inactivating mutations in tuberous sclerosis complex 2 (TSC2), a tumor suppressor gene that controls cell metabolic state and growth via regulation of the mechanistic target of rapamycin (mTORC1) signaling. mTORC1 is frequently activated in human cancers and, although the mTORC1 inhibitor rapamycin has a cytostatic effect, it is, in general, unable to elicit a robust curative effect or tumor regression. Using RNA-Seq, we identified (1) Insulin-like Growth Factor (IGF2) as one of the genes with the highest fold-change difference between human TSC2-null and TSC2-expressing angiomyolipoma cells from a patient with LAM, and (2) the mouse IGF2 homolog Igf2, as a top-ranking gene according to fold change between Tsc2-/- and Tsc2+/+ mouse embryo fibroblasts (MEFs). We extended transcript-level findings to protein level, observing increased Igf2 protein expression and Igf2 secretion by Tsc2-/- MEFs. Increased Igf2 expression was not due to epigenetic imprinting, but was partially mediated through the Stat3 pathway and was completely insensitive to rapamycin treatment. An siRNA-mediated decrease of Igf2 resulted in decreased Stat3 phosphorylation, suggesting presence of an autocrine Igf2/Stat3 amplification cycle in Tsc2-/- MEFs. In human pulmonary LAM lesions and metastatic cell clusters, high levels of IGF2 were associated with mTORC1 activation. In addition, treatment of three primary IGF2-expressing LAM lung cell lines with rapamycin did not result in IGF2 level changes. Thus, targeting of IGF2 signaling may be of therapeutic value to LAM patients, particularly those who are unresponsive to rapamycin.
Collapse
Affiliation(s)
- Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lurong Lian
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ryan Rue
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elena N. Atochina-Vasserman
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Stella K. Hur
- Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marisa S. Bartolomei
- Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jilly F. Evans
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Vera P. Krymskaya
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
41
|
Lee HK, Kwon B, Lemere CA, de la Monte S, Itamura K, Ha AY, Querfurth HW. mTORC2 (Rictor) in Alzheimer's Disease and Reversal of Amyloid-β Expression-Induced Insulin Resistance and Toxicity in Rat Primary Cortical Neurons. J Alzheimers Dis 2018; 56:1015-1036. [PMID: 28035937 DOI: 10.3233/jad-161029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1), a nutrient sensor and central controller of cell growth and proliferation, is altered in various models of Alzheimer's disease (AD). Even less studied or understood in AD is mammalian target of rapamycin complex 2 (mTORC2) that influences cellular metabolism, in part through the regulations of Akt/PKB and SGK. Dysregulation of insulin/PI3K/Akt signaling is another important feature of AD pathogenesis. We found that both total mTORC1 and C2 protein levels and individual C1 and C2 enzymatic activities were decreased in human AD brain samples. In two rodent AD models, mTORC1 and C2 activities were also decreased. In a neuronal culture model of AD characterized by accumulation of cellular amyloid-β (Aβ)42, mTORC1 activity was reduced. Autophagic vesicles and markers were correspondingly increased and new protein synthesis was inhibited, consistent with mTORC1 hypofunction. Interestingly, mTORC2 activity in neural culture seemed resistant to the effects of intracellular amyloid. In various cell lines, Aβ expression provoked insulin resistance, characterized by inhibition of stimulated Akt phosphorylation, and an increase in negative mTORC1 regular, p-AMPK, itself a nutrient sensor. Rapamycin decreased phospho-mTOR and to lesser degree p-Rictor. This further suppression of mTORC1 activity protected cells from Aβ-induced toxicity and insulin resistance. More striking, Rictor over-expression fully reversed the Aβ-effects on primary neuronal cultures. Finally, using in vitro assay, Rictor protein addition completely overcame oligomeric Aβ-induced inhibition of the PDK-Akt activation step. We conclude that striking a new balance by restoring mTORC2 abundance and/or inhibition of mTORC1 has therapeutic potential in AD.
Collapse
Affiliation(s)
- Han-Kyu Lee
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Bumsup Kwon
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suzanne de la Monte
- Department of Pathology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Kyohei Itamura
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Austin Y Ha
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Henry W Querfurth
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
42
|
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, low-grade, metastasizing neoplasm that arises from an unknown source, spreads via the lymphatics, and targets the lungs. All pulmonary structures become infiltrated with benign-appearing spindle and epithelioid cells (LAM cells) that express smooth-muscle and melanocyte-lineage markers, harbor mTOR-activating mutations in tuberous sclerosis complex (TSC) genes, and recruit abundant stromal cells. Elaboration of lymphangiogenic growth factors and matrix remodeling enzymes by LAM cells enables their access to lymphatic channels and likely drives the cystic lung remodeling that often culminates in respiratory failure. Dysregulated cellular signaling results in a shift from oxidative phosphorylation to glycolysis as the preferred mode of energy generation, to allow for the accumulation of biomass required for cell growth and tolerance of nutrient-poor, anaerobic environments. Symptomatic LAM occurs almost exclusively in females after menarche, highlighting the central but as yet poorly understood role for sex-restricted anatomical structures and/or hormones in disease pathogenesis. LAM is an elegant model of malignancy because biallelic mutations at a single genetic locus confer all features that define cancer upon the LAM cell-metabolic reprogramming and proliferative signals that drive uncontrolled growth and inappropriate migration and invasion, the capacity to exploit the lymphatic circulation as a vehicle for metastasis and access to the lungs, and destruction of remote tissues. The direct benefit of the study of this rare disease has been the rapid identification of an effective FDA-approved therapy, and the collateral benefits have included elucidation of the pivotal roles of mTOR signaling in the regulation of cellular metabolism and the pathogenesis of cancer.
Collapse
Affiliation(s)
- Vera P Krymskaya
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Francis X McCormack
- Department of Internal Medicine, University of Cincinnati School of Medicine, Cincinnati, Ohio 45267;
| |
Collapse
|
43
|
Stepanova V, Dergilev KV, Holman KR, Parfyonova YV, Tsokolaeva ZI, Teter M, Atochina-Vasserman EN, Volgina A, Zaitsev SV, Lewis SP, Zabozlaev FG, Obraztsova K, Krymskaya VP, Cines DB. Urokinase-type plasminogen activator (uPA) is critical for progression of tuberous sclerosis complex 2 (TSC2)-deficient tumors. J Biol Chem 2017; 292:20528-20543. [PMID: 28972182 DOI: 10.1074/jbc.m117.799593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a fatal lung disease associated with germline or somatic inactivating mutations in tuberous sclerosis complex genes (TSC1 or TSC2). LAM is characterized by neoplastic growth of smooth muscle-α-actin-positive cells that destroy lung parenchyma and by the formation of benign renal neoplasms called angiolipomas. The mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin slows progression of these diseases but is not curative and associated with notable toxicity at clinically effective doses, highlighting the need for better understanding LAM's molecular etiology. We report here that LAM lesions and angiomyolipomas overexpress urokinase-type plasminogen activator (uPA). Tsc1-/- and Tsc2-/- mouse embryonic fibroblasts expressed higher uPA levels than their WT counterparts, resulting from the TSC inactivation. Inhibition of uPA expression in Tsc2-null cells reduced the growth and invasiveness and increased susceptibility to apoptosis. However, rapamycin further increased uPA expression in TSC2-null tumor cells and immortalized TSC2-null angiomyolipoma cells, but not in cells with intact TSC. Induction of glucocorticoid receptor signaling or forkhead box (FOXO) 1/3 inhibition abolished the rapamycin-induced uPA expression in TSC-compromised cells. Moreover, rapamycin-enhanced migration of TSC2-null cells was inhibited by the uPA inhibitor UK122, dexamethasone, and a FOXO inhibitor. uPA-knock-out mice developed fewer and smaller TSC2-null lung tumors, and introduction of uPA shRNA in tumor cells or amiloride-induced uPA inhibition reduced tumorigenesis in vivo These findings suggest that interference with the uPA-dependent pathway, when used along with rapamycin, might attenuate LAM progression and potentially other TSC-related disorders.
Collapse
Affiliation(s)
| | - Konstantin V Dergilev
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Kelci R Holman
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Yelena V Parfyonova
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Zoya I Tsokolaeva
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Mimi Teter
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Elena N Atochina-Vasserman
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Alla Volgina
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | | | - Shane P Lewis
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Fedor G Zabozlaev
- the Department of Pathology, Federal Research Clinical Center Federal Medical and Biological Agency of Russia, Moscow 115682, Russia
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Vera P Krymskaya
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Douglas B Cines
- From the Department of Pathology and Laboratory Medicine and
| |
Collapse
|
44
|
Torre O, Elia D, Caminati A, Harari S. New insights in lymphangioleiomyomatosis and pulmonary Langerhans cell histiocytosis. Eur Respir Rev 2017; 26:26/145/170042. [PMID: 28954765 PMCID: PMC9488980 DOI: 10.1183/16000617.0042-2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) and pulmonary Langerhans cell histiocytosis (PLCH) are rare diseases that lead to progressive cystic destruction of the lungs. Despite their distinctive characteristics, these diseases share several features. Patients affected by LAM or PLCH have similar radiological cystic patterns, a similar age of onset, and the possibility of extrapulmonary involvement. In this review, the recent advances in the understanding of the molecular pathogenesis, as well as the current and most promising biomarkers and therapeutic approaches, are described. Understanding of LAM/PLCH pathogenesis has improved over the past years, leading to new therapeutic approacheshttp://ow.ly/7wjR30erSJY
Collapse
Affiliation(s)
- Olga Torre
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica IRCCS, Milan, Italy
| | - Davide Elia
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica IRCCS, Milan, Italy
| | - Antonella Caminati
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica IRCCS, Milan, Italy
| | - Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica IRCCS, Milan, Italy
| |
Collapse
|
45
|
Abstract
Lymphangioleiomyomatosis is a rare multisystem disease predominantly affecting women that can occur sporadically or in association with tuberous sclerosis. Lung cysts progressively replace the lung parenchyma, which leads to dyspnea, recurrent pneumothorax, and in some cases respiratory failure. Patients may also have lymphatic disease in the thorax, abdomen, and pelvis, and renal angiomyolipomas. Treatment includes supportive care, bronchodilators, and for those with progressive disease, mammalian target of rapamycin (mTOR) inhibitors.
Collapse
|
46
|
Shenkar R, Shi C, Austin C, Moore T, Lightle R, Cao Y, Zhang L, Wu M, Zeineddine HA, Girard R, McDonald DA, Rorrer A, Gallione C, Pytel P, Liao JK, Marchuk DA, Awad IA. RhoA Kinase Inhibition With Fasudil Versus Simvastatin in Murine Models of Cerebral Cavernous Malformations. Stroke 2016; 48:187-194. [PMID: 27879448 DOI: 10.1161/strokeaha.116.015013] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE We sought to compare the effect of chronic treatment with commonly tolerated doses of Fasudil, a specific RhoA kinase (ROCK) inhibitor, and simvastatin (with pleiotropic effects including ROCK inhibition) on cerebral cavernous malformation (CCM) genesis and maturation in 2 models that recapitulate the human disease. METHODS Two heterozygous murine models, Ccm1+/-Msh2-/- and Ccm2+/-Trp53-/-, were treated from weaning to 4 to 5 months of age with Fasudil (100 mg/kg per day), simvastatin (40 mg/kg per day) or with placebo. Mouse brains were blindly assessed for CCM lesion burden, nonheme iron deposition (as a quantitative measure of chronic lesional hemorrhage), and ROCK activity. RESULTS Fasudil, but not simvastatin, significantly decreased mature CCM lesion burden in Ccm1+/-Msh2-/- mice, and in meta-analysis of both models combined, when compared with mice receiving placebo. Fasudil and simvastatin both significantly decreased the integrated iron density per mature lesion area in Ccm1+/-Msh2-/- mice, and in both models combined, compared with mice given placebo. ROCK activity in mature lesions of Ccm1+/-Msh2-/- mice was similar with both treatments. Fasudil, but not simvastatin, improved survival in Ccm1+/-Msh2-/- mice. Fasudil and simvastatin treatment did not affect survival or lesion development significantly in Ccm2+/-Trp53-/- mice alone, and Fasudil benefit seemed limited to males. CONCLUSIONS ROCK inhibitor Fasudil was more efficacious than simvastatin in improving survival and blunting the development of mature CCM lesions. Both drugs significantly decreased chronic hemorrhage in CCM lesions. These findings justify the development of ROCK inhibitors and the clinical testing of commonly used statin agents in CCM.
Collapse
Affiliation(s)
- Robert Shenkar
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Changbin Shi
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Cecilia Austin
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Thomas Moore
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Rhonda Lightle
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Ying Cao
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Lingjiao Zhang
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Meijing Wu
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Hussein A Zeineddine
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Romuald Girard
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - David A McDonald
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Autumn Rorrer
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Carol Gallione
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Peter Pytel
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - James K Liao
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Douglas A Marchuk
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Issam A Awad
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk).
| |
Collapse
|
47
|
Kudryashova TV, Goncharov DA, Pena A, Kelly N, Vanderpool R, Baust J, Kobir A, Shufesky W, Mora AL, Morelli AE, Zhao J, Ihida-Stansbury K, Chang B, DeLisser H, Tuder RM, Kawut SM, Silljé HHW, Shapiro S, Zhao Y, Goncharova EA. HIPPO-Integrin-linked Kinase Cross-Talk Controls Self-Sustaining Proliferation and Survival in Pulmonary Hypertension. Am J Respir Crit Care Med 2016; 194:866-877. [PMID: 27119551 PMCID: PMC5074651 DOI: 10.1164/rccm.201510-2003oc] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/29/2016] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Enhanced proliferation and impaired apoptosis of pulmonary arterial vascular smooth muscle cells (PAVSMCs) are key pathophysiologic components of pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). OBJECTIVES To determine the role and therapeutic relevance of HIPPO signaling in PAVSMC proliferation/apoptosis imbalance in PAH. METHODS Primary distal PAVSMCs, lung tissue sections from unused donor (control) and idiopathic PAH lungs, and rat and mouse models of SU5416/hypoxia-induced pulmonary hypertension (PH) were used. Immunohistochemical, immunocytochemical, and immunoblot analyses and transfection, infection, DNA synthesis, apoptosis, migration, cell count, and protein activity assays were performed in this study. MEASUREMENTS AND MAIN RESULTS Immunohistochemical and immunoblot analyses demonstrated that the HIPPO central component large tumor suppressor 1 (LATS1) is inactivated in small remodeled pulmonary arteries (PAs) and distal PAVSMCs in idiopathic PAH. Molecular- and pharmacology-based analyses revealed that LATS1 inactivation and consequent up-regulation of its reciprocal effector Yes-associated protein (Yap) were required for activation of mammalian target of rapamycin (mTOR)-Akt, accumulation of HIF1α, Notch3 intracellular domain and β-catenin, deficiency of proapoptotic Bim, increased proliferation, and survival of human PAH PAVSMCs. LATS1 inactivation and up-regulation of Yap increased production and secretion of fibronectin that up-regulated integrin-linked kinase 1 (ILK1). ILK1 supported LATS1 inactivation, and its inhibition reactivated LATS1, down-regulated Yap, suppressed proliferation, and promoted apoptosis in PAH, but not control PAVSMCs. PAVSM in small remodeled PAs from rats and mice with SU5416/hypoxia-induced PH showed down-regulation of LATS1 and overexpression of ILK1. Treatment of mice with selective ILK inhibitor Cpd22 at Days 22-35 of SU5416/hypoxia exposure restored LATS1 signaling and reduced established pulmonary vascular remodeling and PH. CONCLUSIONS These data report inactivation of HIPPO/LATS1, self-supported via Yap-fibronectin-ILK1 signaling loop, as a novel mechanism of self-sustaining proliferation and apoptosis resistance of PAVSMCs in PAH and suggest a new potential target for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Andressa Pena
- 1 Heart, Lung, Blood and Vascular Medicine Institute
| | | | | | - Jeff Baust
- 1 Heart, Lung, Blood and Vascular Medicine Institute
| | | | - William Shufesky
- 3 Department of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ana L Mora
- 1 Heart, Lung, Blood and Vascular Medicine Institute
- 2 Department of Medicine, and
| | - Adrian E Morelli
- 3 Department of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Kaori Ihida-Stansbury
- 4 Department of Pathology and Laboratory Medicine
- 5 Pulmonary Vascular Disease Program
| | - Baojun Chang
- 1 Heart, Lung, Blood and Vascular Medicine Institute
- 2 Department of Medicine, and
| | - Horace DeLisser
- 5 Pulmonary Vascular Disease Program
- 6 Department of Medicine, and
| | - Rubin M Tuder
- 7 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado; and
| | - Steven M Kawut
- 5 Pulmonary Vascular Disease Program
- 8 Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Herman H W Silljé
- 9 Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Yutong Zhao
- 1 Heart, Lung, Blood and Vascular Medicine Institute
- 2 Department of Medicine, and
| | - Elena A Goncharova
- 1 Heart, Lung, Blood and Vascular Medicine Institute
- 2 Department of Medicine, and
| |
Collapse
|
48
|
Laugier F, Finet-Benyair A, André J, Rachakonda PS, Kumar R, Bensussan A, Dumaz N. RICTOR involvement in the PI3K/AKT pathway regulation in melanocytes and melanoma. Oncotarget 2016; 6:28120-31. [PMID: 26356562 PMCID: PMC4695048 DOI: 10.18632/oncotarget.4866] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 08/17/2015] [Indexed: 01/16/2023] Open
Abstract
Several studies have highlighted the importance of the PI3K pathway in melanocytes and its frequent over-activation in melanoma. However, little is known about regulation of the PI3K pathway in melanocytic cells. We showed that normal human melanocytes are less sensitive to selective PI3K or mTOR inhibitors than to dual PI3K/mTOR inhibitors. The resistance to PI3K inhibitor was due to a rapid AKT reactivation limiting the inhibitor effect on proliferation. Reactivation of AKT was linked to a feedback mechanism involving the mTORC2 complex and in particular its scaffold protein RICTOR. RICTOR overexpression in melanocytes disrupted the negative feedback, activated the AKT pathway and stimulated clonogenicity highlighting the importance of this feedback to restrict melanocyte proliferation. We found that the RICTOR locus is frequently amplified and overexpressed in melanoma and that RICTOR over-expression in NRAS-transformed melanocytes stimulates their clonogenicity, demonstrating that RICTOR amplification can cooperate with NRAS mutation to stimulate melanoma proliferation. These results show that RICTOR plays a central role in PI3K pathway negative feedback in melanocytes and that its deregulation could be involved in melanoma development.
Collapse
Affiliation(s)
- Florence Laugier
- INSERM, U976, Centre de Recherche sur la Peau, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | - Adeline Finet-Benyair
- INSERM, U976, Centre de Recherche sur la Peau, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | - Jocelyne André
- INSERM, U976, Centre de Recherche sur la Peau, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | | | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Armand Bensussan
- INSERM, U976, Centre de Recherche sur la Peau, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | - Nicolas Dumaz
- INSERM, U976, Centre de Recherche sur la Peau, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| |
Collapse
|
49
|
Tulbah AS, Ong HX, Colombo P, Young PM, Traini D. Could simvastatin be considered as a potential therapy for chronic lung diseases? A debate on the pros and cons. Expert Opin Drug Deliv 2016; 13:1407-20. [PMID: 27212150 DOI: 10.1080/17425247.2016.1193150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Simvastatin (SV) is a drug from the statin class, currently used orally as an anti-cholesterolemic drug. It inhibits the 3-hydroxy-3-methyl-glutaryl-Coenzyme A (HMG-CoA) reductase to reduce cholesterol synthesis. Recently, it has been found that SV also has several other protective pharmacological actions unrelated to its anti-cholesterol effects that might be beneficial in the treatment of chronic airway diseases. AREAS COVERED This review summarizes the evidence relating to SV as a potential anti-inflammatory, anti-oxidant and muco-inhibitory agent, administered both orally and via pulmonary inhalation, and discusses its pro and cons. Evidence could potentially be used to support the delivery of SV as inhaled formulation for the treatment of chronic respiratory diseases. EXPERT OPINION The use of SV as anti-inflammatory, anti-oxidant and muco-inhibitory agent for drug delivery to the lung is promising. Inhaled SV formulations could allow the delivery profile to be customized and optimized to take advantage of the rapid onset of action, low systemic side effect and improved physico-chemical stability. This treatment could potentially to be used clinically for the localized treatment of lung diseases where inflammation and oxidative stress production is present.
Collapse
Affiliation(s)
- Alaa S Tulbah
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia.,b Faculty of Pharmacy , Umm Al Qura University , Makkah , Saudi Arabia
| | - Hui Xin Ong
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia
| | - Paolo Colombo
- c Department of Pharmacy , University of Parma , Parma , Italy
| | - Paul M Young
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia
| | - Daniela Traini
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia
| |
Collapse
|
50
|
Li J, Shin S, Sun Y, Yoon SO, Li C, Zhang E, Yu J, Zhang J, Blenis J. mTORC1-Driven Tumor Cells Are Highly Sensitive to Therapeutic Targeting by Antagonists of Oxidative Stress. Cancer Res 2016; 76:4816-27. [PMID: 27197195 DOI: 10.1158/0008-5472.can-15-2629] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 05/05/2016] [Indexed: 01/13/2023]
Abstract
mTORC1 is a central signaling node in controlling cell growth, proliferation, and metabolism that is aberrantly activated in cancers and certain cancer-associated genetic disorders, such as tuberous sclerosis complex (TSC) and sporadic lymphangioleiomyomatosis. However, while mTORC1-inhibitory compounds (rapamycin and rapalogs) attracted interest as candidate therapeutics, clinical trials have not replicated the promising findings in preclinical models, perhaps because these compounds tend to limit cell proliferation without inducing cell death. In seeking to address this issue, we performed a high-throughput screen for small molecules that could heighten the cytotoxicity of mTORC1 inhibitors. Here we report the discovery that combining inhibitors of mTORC1 and glutamate cysteine ligase (GCLC) can selectively and efficiently trigger apoptosis in Tsc2-deficient cells but not wild-type cells. Mechanistic investigations revealed that coinhibition of mTORC1 and GCLC decreased the level of the intracellular thiol antioxidant glutathione (GSH), thereby increasing levels of reactive oxygen species, which we determined to mediate cell death in Tsc2-deficient cells. Our findings offer preclinical proof of concept for a strategy to selectively increase the cytotoxicity of mTORC1 inhibitors as a therapy to eradicate tumor cells marked by high mTORC1 signaling, based on cotargeting a GSH-controlled oxidative stress pathway. Cancer Res; 76(16); 4816-27. ©2016 AACR.
Collapse
Affiliation(s)
- Jing Li
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Sejeong Shin
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Yang Sun
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Department of Dermatology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Sang-Oh Yoon
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Chenggang Li
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Erik Zhang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jane Yu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jianming Zhang
- Department of Dermatology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - John Blenis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts. Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York.
| |
Collapse
|