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
|
Hartung J, Müller C, Calkhoven CF. The dual role of the TSC complex in cancer. Trends Mol Med 2024:S1471-4914(24)00276-4. [PMID: 39488444 DOI: 10.1016/j.molmed.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
The tuberous sclerosis complex (TSC1/TSC2/TBC1D7) primarily functions to inhibit the mechanistic target of rapamycin complex 1 (mTORC1), a crucial regulator of cell growth. Mutations in TSC1 or TSC2 cause tuberous sclerosis complex (TSC), a rare autosomal dominant genetic disorder marked by benign tumors in multiple organs that rarely progress to malignancy. Traditionally, TSC proteins are considered tumor suppressive due to their inhibition of mTORC1 and other mechanisms. However, more recent studies have shown that TSC proteins can also promote tumorigenesis in certain cancer types. In this review, we explore the composition and function of the TSC protein complex, the roles of its individual components in cancer biology, and potential future therapeutic targeting strategies.
Collapse
Affiliation(s)
- Josephine Hartung
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands.
| |
Collapse
|
3
|
Amosu MM, Jankowski AM, McCright JC, Yang BE, de Oro Fernandez JG, Moore KA, Gadde HS, Donthi M, Kaluzienski ML, Sriram V, Maisel K. Plasmacytoid dendritic cells mediate CpG-ODN induced increase in survival in a mouse model of lymphangioleiomyomatosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.06.527331. [PMID: 36798234 PMCID: PMC9934559 DOI: 10.1101/2023.02.06.527331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
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 re-activate immunity and the TLR9 agonist, CpG-ODN, has been effective in treating lung cancer in animal models. Here we investigate the use of TLR9 agonist CpG-ODN as LAM immunotherapy in combination with checkpoint inhibitor, anti-PD1, standard of care rapamycin and determine 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 due to fewer local side effects but increased LAM nodule count and size compared to the higher dose. CpG-ODN treatment also reduced regulatory T cells and increased the number of Th17 helper T cells as well as cytotoxic T cells. These effects appear to be mediated in part by plasmacytoid dendritic cells (pDCs), as depletion of pDCs reduces survival and abrogates Th17 T 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, MD 20742
| | - Ashleigh M Jankowski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Jacob C McCright
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Bennett E Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | | | - Kaitlyn A Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Havish S Gadde
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Mehul Donthi
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Michele L Kaluzienski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Vedanth Sriram
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| |
Collapse
|
4
|
Olatoke T, Wagner A, Astrinidis A, Zhang EY, Guo M, Zhang AG, Mattam U, Kopras EJ, Gupta N, Smith EP, Karbowniczek M, Markiewski MM, Wikenheiser-Brokamp KA, Whitsett JA, McCormack FX, Xu Y, Yu JJ. Single-cell multiomic analysis identifies a HOX-PBX gene network regulating the survival of lymphangioleiomyomatosis cells. SCIENCE ADVANCES 2023; 9:eadf8549. [PMID: 37163604 PMCID: PMC10171823 DOI: 10.1126/sciadv.adf8549] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/07/2023] [Indexed: 05/12/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, progressive lung disease that predominantly affects women. LAM cells carry TSC1/TSC2 mutations, causing mTORC1 hyperactivation and uncontrolled cell growth. mTORC1 inhibitors stabilize lung function; however, sustained efficacy requires long-term administration, and some patients fail to tolerate or respond to therapy. Although the genetic basis of LAM is known, mechanisms underlying LAM pathogenesis remain elusive. We integrated single-cell RNA sequencing and single-nuclei ATAC-seq of LAM lungs to construct a gene regulatory network controlling the transcriptional program of LAM cells. We identified activation of uterine-specific HOX-PBX transcriptional programs in pulmonary LAMCORE cells as regulators of cell survival depending upon HOXD11-PBX1 dimerization. Accordingly, blockage of HOXD11-PBX1 dimerization by HXR9 suppressed LAM cell survival in vitro and in vivo. PBX1 regulated STAT1/3, increased the expression of antiapoptotic genes, and promoted LAM cell survival in vitro. The HOX-PBX gene network provides promising targets for treatment of LAM/TSC mTORC1-hyperactive cancers.
Collapse
Affiliation(s)
- Tasnim Olatoke
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Andrew Wagner
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Aristotelis Astrinidis
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Erik Y. Zhang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Minzhe Guo
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Alan G. Zhang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ushodaya Mattam
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Elizabeth J. Kopras
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Nishant Gupta
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Eric P. Smith
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Magdalena Karbowniczek
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Maciej M. Markiewski
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Kathryn A. Wikenheiser-Brokamp
- Division of Pathology and Laboratory Medicine, Perinatal Institute, Division of Pulmonary Biology, Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jeffrey A. Whitsett
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Francis X. McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yan Xu
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jane J. Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
5
|
Kundu N, Holz MK. Lymphangioleiomyomatosis: a metastatic lung disease. Am J Physiol Cell Physiol 2023; 324:C320-C326. [PMID: 36571446 PMCID: PMC9886342 DOI: 10.1152/ajpcell.00202.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare disease affecting women, caused by somatic mutations in the TSC1 or TSC2 genes, and driven by estrogen. Similar to many cancers, it is metastatic, primarily to the lung. Despite its monogenetic nature, like many cancers, LAM is a heterogeneous disease. The cellular constituents of LAM are very diverse, including mesenchymal, epithelial, endothelial, and immune cells. LAM is characterized by dysregulation of many cell signaling pathways, distinct populations of LAM cells, and a rich microenvironment, in which the immune system appears to play an important role. This review delineates the heterogeneity of LAM and focuses on the metastatic features of LAM, the deregulated signaling mechanisms and the tumor microenvironment. Understanding the tumor-host interaction in LAM may provide insights into the development of new therapeutic strategies, which could be combinatorial or superlative to Sirolimus, the current U.S. Food and Drug Administration-approved treatment.
Collapse
Affiliation(s)
- Nandini Kundu
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, New York
| | - Marina K Holz
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, New York
- Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, New York
| |
Collapse
|
6
|
Huang J, Xu W, Liu P, Liu Y, Shen C, Liu S, Wang Y, Wang J, Zhang T, He Y, Cheng C, Yang L, Zhang W, Tian X, Xu KF. Gene mutations in sporadic lymphangioleiomyomatosis and genotype–phenotype correlation analysis. BMC Pulm Med 2022; 22:354. [PMID: 36117164 PMCID: PMC9482747 DOI: 10.1186/s12890-022-02154-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background Sporadic lymphangioleiomyomatosis (S-LAM) is a rare neoplasm with heterogeneous clinical features that is conventionally considered to be related to TSC2. This study serves to elucidate the mutation landscape and potential correlation between S-LAM genomic profiles and clinical phenotypes. Methods Genomic profiles of 22 S-LAM patients were obtained by sequencing genomic DNA and cell-free DNA from various specimens using an NGS (next-generation sequencing)-based tumor-driver gene panel. Detected mutations were summarized. Symptoms, serum vascular endothelial growth factor D (VEGF-D) values, pulmonary function, and six-minute walk distance (6MWD) were compared among groups with different TSC2 status and genotypes to analyze genotype–phenotype correlations. Results 67 Variants in 43 genes were detected, with a TSC2 mutation detection rate of 68.2%. The TSC2 detection rate was similar in specimens obtained either through transbronchial lung biopsy (TBLB) or surgical lung biopsy (70.0% vs. 69.2%, p > 0.05). A novel mutation in VEZF1 (c.A659G) was detected in four participants and may represent a mild disease state. TSC2 mutation was significantly related to a shorter 6MWD (p < 0.05), and a higher percentage of VEGF-D over 800 pg/mL (p < 0.05); stop-gain mutation was significantly related to a higher prevalence of pneumothorax. Conclusions Tumor-driver mutations in genes other than TSC2 may have a role in S-LAM, and TBLB specimens are practical alternatives for genomic analysis. TSC2 mutation detectability and types are related to the disease severity and phenotypes of S-LAM. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-02154-0.
Collapse
|
7
|
Liu X, Xu Y, Wu X, Liu Y, Wu Q, Wu J, Zhang H, Zhou M, Qu J. Soluble Immune-Related Proteins as New Candidate Serum Biomarkers for the Diagnosis and Progression of Lymphangioleiomyomatosis. Front Immunol 2022; 13:844914. [PMID: 35300340 PMCID: PMC8923288 DOI: 10.3389/fimmu.2022.844914] [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: 12/29/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background The goal of this study was to analyze serum from lymphangioleiomyomatosis (LAM) patients and healthy controls to identify novel biomarkers that could shed light on disease diagnosis and pathogenesis. Methods From April 2017 to October 2019, qualified serum samples were obtained to explore differences in 59 immune proteins between 67 LAM patients and 49 healthy controls by the Luminex method. Results We characterized 22 serum immune proteins that were differentially expressed in LAM patients compared with healthy people. Fifty-nine proteins were then classified into eight categories according to their biological function, and the results showed that LAM patients displayed significantly higher levels of growth factors (p = 0.006) and lower levels of costimulatory molecules (p = 0.008). LAG-3 was not only likely to have better predictive value than VEGF-D but also showed a significant difference between patients without elevated VEGF-D and healthy people. IL-18 was positively correlated with lung function and six-minute walk test (6MWT) distance and negatively correlated with St. George’s Respiratory Questionnaire (SGRQ) score and pulmonary artery systolic pressure (PASP), which suggested that IL-18 was related to disease severity. PD-1 was significantly different between patients with pneumothorax and/or chylothorax and those without complications. Conclusion We performed a large-scale serum immune factor analysis of LAM. Our study provides evidence that LAG-3 may be a novel candidate serum biomarker for the diagnosis of LAM. Future independent validation in prospective studies is warranted.
Collapse
Affiliation(s)
- Xuefei Liu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanping Xu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueying Wu
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Ninth School of Clinical Medicine, Peking University, Beijing, China.,School of Oncology, Capital Medical University, Beijing, China
| | - Yanpu Liu
- Department of Respiration, Xiangshan Traditional Chinese Medicine Hospital, Shanghai, China
| | - Qiang Wu
- Department of Respiration, Xiangshan Traditional Chinese Medicine Hospital, Shanghai, China
| | - Jialiang Wu
- Department of Respiration, Xiangshan Traditional Chinese Medicine Hospital, Shanghai, China
| | - Henghui Zhang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Ninth School of Clinical Medicine, Peking University, Beijing, China.,School of Oncology, Capital Medical University, Beijing, China
| | - Min Zhou
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Espín R, Baiges A, Blommaert E, Herranz C, Roman A, Saez B, Ancochea J, Valenzuela C, Ussetti P, Laporta R, Rodríguez-Portal JA, van Moorsel CHM, van der Vis JJ, Quanjel MJR, Villar-Piqué A, Diaz-Lucena D, Llorens F, Casanova Á, Molina-Molina M, Plass M, Mateo F, Moss J, Pujana MA. Heterogeneity and Cancer-Related Features in Lymphangioleiomyomatosis Cells and Tissue. Mol Cancer Res 2021; 19:1840-1853. [PMID: 34312290 PMCID: PMC8568632 DOI: 10.1158/1541-7786.mcr-21-0220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/23/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, low-grade metastasizing disease characterized by cystic lung destruction. LAM can exhibit extensive heterogeneity at the molecular, cellular, and tissue levels. However, the molecular similarities and differences among LAM cells and tissue, and their connection to cancer features are not fully understood. By integrating complementary gene and protein LAM signatures, and single-cell and bulk tissue transcriptome profiles, we show sources of disease heterogeneity, and how they correspond to cancer molecular portraits. Subsets of LAM diseased cells differ with respect to gene expression profiles related to hormones, metabolism, proliferation, and stemness. Phenotypic diseased cell differences are identified by evaluating lumican (LUM) proteoglycan and YB1 transcription factor expression in LAM lung lesions. The RUNX1 and IRF1 transcription factors are predicted to regulate LAM cell signatures, and both regulators are expressed in LAM lung lesions, with differences between spindle-like and epithelioid LAM cells. The cancer single-cell transcriptome profiles most similar to those of LAM cells include a breast cancer mesenchymal cell model and lines derived from pleural mesotheliomas. Heterogeneity is also found in LAM lung tissue, where it is mainly determined by immune system factors. Variable expression of the multifunctional innate immunity protein LCN2 is linked to disease heterogeneity. This protein is found to be more abundant in blood plasma from LAM patients than from healthy women. IMPLICATIONS: This study identifies LAM molecular and cellular features, master regulators, cancer similarities, and potential causes of disease heterogeneity.
Collapse
Affiliation(s)
- Roderic Espín
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Alexandra Baiges
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Eline Blommaert
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Carmen Herranz
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Antonio Roman
- Lung Transplant Unit, Pneumology Service, Lymphangioleiomyomatosis Clinic, Vall d'Hebron University Hospital, Barcelona, Catalonia, Spain
| | - Berta Saez
- Lung Transplant Unit, Pneumology Service, Lymphangioleiomyomatosis Clinic, Vall d'Hebron University Hospital, Barcelona, Catalonia, Spain
| | - Julio Ancochea
- Pneumology Service, University Hospital La Princesa, La Princesa Research Institute (IIS-IP), Madrid, Spain
| | - Claudia Valenzuela
- Pneumology Service, University Hospital La Princesa, La Princesa Research Institute (IIS-IP), Madrid, Spain
| | - Piedad Ussetti
- Pneumology Service, University Hospital Clínica Puerta del Hierro, Majadahonda, Madrid, Spain
| | - Rosalía Laporta
- Pneumology Service, University Hospital Clínica Puerta del Hierro, Majadahonda, Madrid, Spain
| | - José A Rodríguez-Portal
- Medical-Surgical Unit of Respiratory Diseases, University Hospital Virgen del Rocío, Institute of Biomedicine of Seville (IBiS), Seville, Spain
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Coline H M van Moorsel
- Interstitial Lung Disease (ILD) Center of Excellence, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Joanne J van der Vis
- Interstitial Lung Disease (ILD) Center of Excellence, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Marian J R Quanjel
- Interstitial Lung Disease (ILD) Center of Excellence, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Anna Villar-Piqué
- Neuroscience Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
- Biomedical Research Network Centre in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniela Diaz-Lucena
- Neuroscience Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
- Biomedical Research Network Centre in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Franc Llorens
- Neuroscience Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
- Biomedical Research Network Centre in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Göttingen, Germany
| | - Álvaro Casanova
- Pneumology Service, University Hospital of Henares, University Francisco de Vitoria, Coslada, Madrid, Spain
| | - María Molina-Molina
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Interstitial Lung Disease Unit, Department of Respiratory Medicine, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Mireya Plass
- Program for Advancing Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
- Gene Regulation of Cell Identity, Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
- Biomedical Research Network Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francesca Mateo
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Miquel Angel Pujana
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain.
| |
Collapse
|
9
|
Herranz C, Mateo F, Baiges A, Ruiz de Garibay G, Junza A, Johnson SR, Miller S, García N, Capellades J, Gómez A, Vidal A, Palomero L, Espín R, Extremera AI, Blommaert E, Revilla‐López E, Saez B, Gómez‐Ollés S, Ancochea J, Valenzuela C, Alonso T, Ussetti P, Laporta R, Xaubet A, Rodríguez‐Portal JA, Montes‐Worboys A, Machahua C, Bordas J, Menendez JA, Cruzado JM, Guiteras R, Bontoux C, La Motta C, Noguera‐Castells A, Mancino M, Lastra E, Rigo‐Bonnin R, Perales JC, Viñals F, Lahiguera A, Zhang X, Cuadras D, van Moorsel CHM, van der Vis JJ, Quanjel MJR, Filippakis H, Hakem R, Gorrini C, Ferrer M, Ugun‐Klusek A, Billett E, Radzikowska E, Casanova Á, Molina‐Molina M, Roman A, Yanes O, Pujana MA. Histamine signaling and metabolism identify potential biomarkers and therapies for lymphangioleiomyomatosis. EMBO Mol Med 2021; 13:e13929. [PMID: 34378323 PMCID: PMC8422079 DOI: 10.15252/emmm.202113929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/12/2022] Open
Abstract
Inhibition of mTOR is the standard of care for lymphangioleiomyomatosis (LAM). However, this therapy has variable tolerability and some patients show progressive decline of lung function despite treatment. LAM diagnosis and monitoring can also be challenging due to the heterogeneity of symptoms and insufficiency of non-invasive tests. Here, we propose monoamine-derived biomarkers that provide preclinical evidence for novel therapeutic approaches. The major histamine-derived metabolite methylimidazoleacetic acid (MIAA) is relatively more abundant in LAM plasma, and MIAA values are independent of VEGF-D. Higher levels of histamine are associated with poorer lung function and greater disease burden. Molecular and cellular analyses, and metabolic profiling confirmed active histamine signaling and metabolism. LAM tumorigenesis is reduced using approved drugs targeting monoamine oxidases A/B (clorgyline and rasagiline) or histamine H1 receptor (loratadine), and loratadine synergizes with rapamycin. Depletion of Maoa or Hrh1 expression, and administration of an L-histidine analog, or a low L-histidine diet, also reduce LAM tumorigenesis. These findings extend our knowledge of LAM biology and suggest possible ways of improving disease management.
Collapse
Affiliation(s)
- Carmen Herranz
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Francesca Mateo
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Alexandra Baiges
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Gorka Ruiz de Garibay
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Alexandra Junza
- Department of Electronic EngineeringInstitute of Health Research Pere Virgili (IIPSV)University Rovira i VirgiliTarragonaSpain
- Biomedical Research Network Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)Instituto de Salud Carlos IIIMadridSpain
| | - Simon R Johnson
- National Centre for LymphangioleiomyomatosisNottingham University Hospitals NHS Trust, NottinghamshireDivision of Respiratory MedicineUniversity of NottinghamNottinghamUK
| | - Suzanne Miller
- National Centre for LymphangioleiomyomatosisNottingham University Hospitals NHS Trust, NottinghamshireDivision of Respiratory MedicineUniversity of NottinghamNottinghamUK
| | - Nadia García
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Jordi Capellades
- Department of Electronic EngineeringInstitute of Health Research Pere Virgili (IIPSV)University Rovira i VirgiliTarragonaSpain
- Biomedical Research Network Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)Instituto de Salud Carlos IIIMadridSpain
| | - Antonio Gómez
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
- Present address:
Rheumatology Department and Rheumatology Research GroupVall d'Hebron Hospital Research Institute (VHIR)BarcelonaSpain
| | - August Vidal
- Department of PathologyUniversity Hospital of BellvitgeOncobellIDIBELL, L’Hospitalet del LlobregatBarcelonaSpain
- CIBER on Cancer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| | - Luis Palomero
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Roderic Espín
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Ana I Extremera
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Eline Blommaert
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Eva Revilla‐López
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Berta Saez
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Susana Gómez‐Ollés
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Julio Ancochea
- Pneumology ServiceLa Princesa Research InstituteUniversity Hospital La PrincesaMadridSpain
| | - Claudia Valenzuela
- Pneumology ServiceLa Princesa Research InstituteUniversity Hospital La PrincesaMadridSpain
| | - Tamara Alonso
- Pneumology ServiceLa Princesa Research InstituteUniversity Hospital La PrincesaMadridSpain
| | - Piedad Ussetti
- Pneumology ServiceUniversity Hospital Clínica Puerta del Hierro, MajadahondaMadridSpain
| | - Rosalía Laporta
- Pneumology ServiceUniversity Hospital Clínica Puerta del Hierro, MajadahondaMadridSpain
| | - Antoni Xaubet
- Pneumology ServiceHospital Clínic de BarcelonaBarcelonaSpain
| | - José A Rodríguez‐Portal
- Medical‐Surgical Unit of Respiratory DiseasesInstitute of Biomedicine of Seville (IBiS)University Hospital Virgen del RocíoSevilleSpain
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
| | - Ana Montes‐Worboys
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Carlos Machahua
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Jaume Bordas
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Javier A Menendez
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Josep M Cruzado
- Experimental NephrologyDepartment of Clinical SciencesUniversity of BarcelonaBarcelonaSpain
- Department of NephrologyUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Roser Guiteras
- Experimental NephrologyDepartment of Clinical SciencesUniversity of BarcelonaBarcelonaSpain
- Department of NephrologyUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Christophe Bontoux
- Department of PathologyUniversity Hospital Pitié‐SalpêtrièreFaculty of MedicineUniversity of SorbonneParisFrance
| | | | - Aleix Noguera‐Castells
- Biomedical Research Institute “August Pi i Sunyer” (IDIBAPS)Department of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Mario Mancino
- Biomedical Research Institute “August Pi i Sunyer” (IDIBAPS)Department of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Enrique Lastra
- Genetic Counseling UnitDepartment of Medical OncologyUniversity Hospital of BurgosBurgosSpain
| | - Raúl Rigo‐Bonnin
- Clinical LaboratoryUniversity Hospital of BellvitgeIDIBELLL'Hospitalet de LlobregatBarcelonaSpain
| | - Jose C Perales
- Department of Physiological Science IIUniversity of BarcelonaBarcelonaSpain
| | - Francesc Viñals
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
- Department of Physiological Science IIUniversity of BarcelonaBarcelonaSpain
| | - Alvaro Lahiguera
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Xiaohu Zhang
- National Center for Advancing Translational Sciences (NCATS)National Institute of Health (NIH)BethesdaMDUSA
| | - Daniel Cuadras
- Statistics DepartmentFoundation Sant Joan de DéuEspluguesSpain
| | - Coline H M van Moorsel
- Interstitial Lung Disease (ILD) Center of ExcellenceSt. Antonius HospitalNieuwegeinThe Netherlands
| | - Joanne J van der Vis
- Interstitial Lung Disease (ILD) Center of ExcellenceSt. Antonius HospitalNieuwegeinThe Netherlands
| | - Marian J R Quanjel
- Interstitial Lung Disease (ILD) Center of ExcellenceSt. Antonius HospitalNieuwegeinThe Netherlands
| | - Harilaos Filippakis
- Pulmonary and Critical Care MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Razq Hakem
- Princess Margaret Cancer CentreUniversity Health NetworkDepartment of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
| | - Chiara Gorrini
- Princess Margaret HospitalThe Campbell Family Institute for Breast Cancer ResearchOntario Cancer InstituteUniversity Health NetworkTorontoONCanada
| | - Marc Ferrer
- National Center for Advancing Translational Sciences (NCATS)National Institute of Health (NIH)BethesdaMDUSA
| | - Aslihan Ugun‐Klusek
- Centre for Health, Ageing and Understanding Disease (CHAUD)School of Science and TechnologyNottingham Trent UniversityNottinghamUK
| | - Ellen Billett
- Centre for Health, Ageing and Understanding Disease (CHAUD)School of Science and TechnologyNottingham Trent UniversityNottinghamUK
| | - Elżbieta Radzikowska
- Department of Lung Diseases IIINational Tuberculosis and Lung Disease Research InstituteWarsawPoland
| | - Álvaro Casanova
- Pneumology ServiceUniversity Hospital of HenaresUniversity Francisco de Vitoria, CosladaMadridSpain
| | - María Molina‐Molina
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Antonio Roman
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Oscar Yanes
- Department of Electronic EngineeringInstitute of Health Research Pere Virgili (IIPSV)University Rovira i VirgiliTarragonaSpain
- Biomedical Research Network Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)Instituto de Salud Carlos IIIMadridSpain
| | - Miquel A Pujana
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| |
Collapse
|
10
|
Obraztsova K, Basil MC, Rue R, Sivakumar A, Lin SM, Mukhitov AR, Gritsiuta AI, Evans JF, Kopp M, Katzen J, Robichaud A, Atochina-Vasserman EN, Li S, Carl J, Babu A, Morley MP, Cantu E, Beers MF, Frank DB, Morrisey EE, Krymskaya VP. mTORC1 activation in lung mesenchyme drives sex- and age-dependent pulmonary structure and function decline. Nat Commun 2020; 11:5640. [PMID: 33159078 PMCID: PMC7648630 DOI: 10.1038/s41467-020-18979-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare fatal cystic lung disease due to bi-allelic inactivating mutations in tuberous sclerosis complex (TSC1/TSC2) genes coding for suppressors of the mechanistic target of rapamycin complex 1 (mTORC1). The origin of LAM cells is still unknown. Here, we profile a LAM lung compared to an age- and sex-matched healthy control lung as a hypothesis-generating approach to identify cell subtypes that are specific to LAM. Our single-cell RNA sequencing (scRNA-seq) analysis reveals novel mesenchymal and transitional alveolar epithelial states unique to LAM lung. This analysis identifies a mesenchymal cell hub coordinating the LAM disease phenotype. Mesenchymal-restricted deletion of Tsc2 in the mouse lung produces a mTORC1-driven pulmonary phenotype, with a progressive disruption of alveolar structure, a decline in pulmonary function, increase of rapamycin-sensitive expression of WNT ligands, and profound female-specific changes in mesenchymal and epithelial lung cell gene expression. Genetic inactivation of WNT signaling reverses age-dependent changes of mTORC1-driven lung phenotype, but WNT activation alone in lung mesenchyme is not sufficient for the development of mouse LAM-like phenotype. The alterations in gene expression are driven by distinctive crosstalk between mesenchymal and epithelial subsets of cells observed in mesenchymal Tsc2-deficient lungs. This study identifies sex- and age-specific gene changes in the mTORC1-activated lung mesenchyme and establishes the importance of the WNT signaling pathway in the mTORC1-driven lung phenotype.
Collapse
Affiliation(s)
- Kseniya Obraztsova
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria C Basil
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Rue
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Susan M Lin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander R Mukhitov
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrei I Gritsiuta
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jilly F Evans
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meghan Kopp
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeremy Katzen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elena N Atochina-Vasserman
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shanru Li
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Justine Carl
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Apoorva Babu
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Morley
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael F Beers
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - David B Frank
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Children Hospital of Philadelphia, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward E Morrisey
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Vera P Krymskaya
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Immunotherapy for Lymphangioleiomyomatosis and Tuberous Sclerosis: Progress and Future Directions. Chest 2019; 156:1062-1067. [PMID: 31437431 DOI: 10.1016/j.chest.2019.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/24/2019] [Accepted: 08/04/2019] [Indexed: 01/19/2023] Open
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is a rare genetic multisystem disease characterized by the nodular proliferation of smooth muscle-like LAM cells, progressive cystic changes of the lung, lymphatic abnormalities, and renal angiomyolipomas (AMLs). LAM can arise sporadically or in women with the autosomal dominant disorder, tuberous sclerosis complex (TSC), in which hamartomatous tumors of brain, heart, skin, kidney, and lung are found. LAM and TSC are caused by mutations in the TSC1 or TSC2 tumor suppressor genes leading to elevated mechanistic/mammalian target of rapamycin complex activity. Recent data indicate that T cells within LAM nodules and renal AMLs exhibit features of T-cell exhaustion, with coinhibitory receptor programmed cell death protein 1 (PD-1) expression on tumor-infiltrating T cells. Treatment of animal models of TSC and LAM with anti-PD-1 antibodies or with the combination of anti-PD-1 and anti-CTLA4 antibodies has led to remarkable results, suppressing TSC2-null tumor growth and inducing tumor rejection. Here we review our current knowledge about the potential for immunotherapy for the treatment of LAM and TSC and highlight critical unknowns and key next steps.
Collapse
|
12
|
Circulating Lymphangioleiomyomatosis Tumor Cells With Loss of Heterozygosity in the TSC2 Gene Show Increased Aldehyde Dehydrogenase Activity. Chest 2019; 156:298-307. [PMID: 31034819 DOI: 10.1016/j.chest.2019.03.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/27/2019] [Accepted: 03/22/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Lymphangioleiomyomatosis (LAM) is a destructive metastasizing neoplasm of the lung characterized by proliferation of LAM cells in specialized lung nodules. LAM cells are characterized by expression of the prometastatic and cancer-initiating hyaluronan receptor CD44v6, and loss of heterozygosity (LOH) of TSC1 and TSC2. The circulating neoplastic LAM cells are thought to be involved in metastasis. Because LAM cells display properties of neoplastic, metastatic, and stem cell-like cancer cells, we hypothesized that elevated aldehyde dehydrogenase (ALDH) activity, characteristic of cancer and stem cells, is a property of LAM cells. METHODS We performed an in silico search of ALDH genes in microdissected LAM lung nodules. To identify circulating LAM cells, we osmotically removed red blood cells from whole blood to obtain peripheral blood mononuclear cells, which were then sorted by fluorescence-activated cell sorting based on their level of ALDH activity. RESULTS Microdissected LAM lung nodules possess a distinctive ALDH gene profile. The cell subpopulation with high ALDH activity, isolated from circulating cells, possessed TSC2 LOH in 8 of 14 patients with LAM. Approximately 60% of the circulating cells with high ALDH activity expressed CD44v6. Cells with TSC2 LOH from patients with LAM and LAM/TSC exhibited different properties in different body locations, but all cell types showed high ALDH activity. CONCLUSIONS This new procedure allows for isolation of circulating LAM cells from cultured cells, blood, and chylous effusions and shows that circulating LAM cells are heterogeneous with neoplastic, metastatic, and cancer-stem cell-like properties.
Collapse
|
13
|
Steagall WK, Stylianou M, Pacheco-Rodriguez G, Moss J. Angiotensin-converting enzyme inhibitors may affect pulmonary function in lymphangioleiomyomatosis. JCI Insight 2019; 4:126703. [PMID: 30843885 DOI: 10.1172/jci.insight.126703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/25/2019] [Indexed: 01/21/2023] Open
Abstract
INTRODUCTION A local renin-angiotensin system exists in the pulmonary nodules of lymphangioleiomyomatosis patients. Sirolimus, the standard treatment for lymphangioleiomyomatosis, stabilizes lung function, but all patients do not respond to or tolerate sirolimus. As renin-angiotensin systems may affect tumor growth and metastasis, we questioned if angiotensin-converting enzyme inhibitors affected lymphangioleiomyomatosis disease progression. METHODS Retrospective study of 426 patients was performed, examining angiotensin-converting enzyme levels, pulmonary function data, and angiotensin-converting enzyme inhibitor treatment. RESULTS Serum angiotensin-converting enzyme levels were elevated in approximately 33% of patients, increased with duration of disease, and were inversely correlated with pulmonary function. Levels decreased significantly over time with sirolimus treatment. Treatment with angiotensin-converting enzyme inhibitors was reported by approximately 15% of patients and was significantly associated with a slower rate of decline in percentage predicted forced expiratory volume (FEV1) and diffusing capacity of the lungs for carbon monoxide (DLCO) in patients not treated with sirolimus. No significant differences in rates of decline of FEV1 or DLCO were seen in patients treated with both inhibitors and sirolimus versus sirolimus alone. CONCLUSIONS Angiotensin-converting enzyme inhibitors may slow decline of pulmonary function in patients with lymphangioleiomyomatosis not treated with sirolimus. These inhibitors may be an option or adjunct in the treatment of lymphangioleiomyomatosis. A clinical trial may be warranted to examine this possibility. FUNDING NIH.
Collapse
Affiliation(s)
| | - Mario Stylianou
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | | | | |
Collapse
|
14
|
Steagall WK, Pacheco-Rodriguez G, Darling TN, Torre O, Harari S, Moss J. The Lymphangioleiomyomatosis Lung Cell and Its Human Cell Models. Am J Respir Cell Mol Biol 2018; 58:678-683. [PMID: 29406787 PMCID: PMC6002654 DOI: 10.1165/rcmb.2017-0403tr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/05/2018] [Indexed: 01/11/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a multisystem disease of women, affecting lungs, kidneys, and lymphatics. It is caused by the proliferation of abnormal smooth muscle-like LAM cells, with mutations and loss of heterozygosity in the TSC1 or, more frequently, TSC2 genes. Isolated pulmonary LAM cells have been difficult to maintain in culture, and most studies of LAM lung cells involve mixtures of TSC2 wild-type and TSC2-null cells. A clonal population of LAM lung cells has not been established, making analysis of the cells challenging. Cell lines have been established from angiomyolipomas, a common manifestation of LAM, and from tumors from patients with TSC. Circulating LAM cells have also been isolated from blood and other body fluids. LAM cells may also be identified in clusters apparently derived from lymphatic vessels. Genetics, patterns of antigen expression, and signaling pathways have been studied in LAM lung tissue and in LAM cell models, although rarely all in the same study. We show here that LAM cells manifest differences in these characteristics, depending on the source investigated, suggesting further studies.
Collapse
Affiliation(s)
- Wendy K. Steagall
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Gustavo Pacheco-Rodriguez
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Thomas N. Darling
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, Maryland; and
| | - Olga Torre
- Unità Operativa di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Sergio Harari
- Unità Operativa di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
15
|
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
|
16
|
Abstract
INTRODUCTION Lymphangioleiomyomatosis (LAM) is a destructive lung disease affecting primarily women. LAM is caused by inactivating mutations in the tuberous sclerosis complex (TSC) genes, resulting in hyperactivation of mechanistic/mammalian target of rapamycin complex 1 (mTORC1). Over the past five years, there have been remarkable advances in the diagnosis and therapy of LAM, including the identification of vascular endothelial growth factor D (VEGF-D) as a diagnostic biomarker and the US Food and Drug Administration approval of sirolimus as therapy for LAM. In appropriate clinical situations VEGF-D testing can make lung biopsy unnecessary to diagnose LAM. However, there remains an urgent unmet need for additional biomarkers of disease activity and/or response to therapy. Areas covered: This work reviews VEGF-D, an established LAM biomarker, and discusses emerging biomarkers, including circulating LAM cells, imaging, lipid, and metabolite biomarkers, focusing on those with the highest potential impact for LAM patients. Expert commentary: Ongoing research priorities include the development of validated biomarkers to 1) noninvasively diagnose LAM in women whose VEGF-D levels are not diagnostic, 2) accurately predict the likelihood of disease progression and 3) quantitatively measure disease activity and LAM cell burden. These biomarkers would enable personalized, precision clinical care and fast-track clinical trial implementation, with high clinical impact.
Collapse
Affiliation(s)
- Julie Nijmeh
- a Pulmonary and Critical Care Medicine, Department of Medicine , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| | - Souheil El-Chemaly
- a Pulmonary and Critical Care Medicine, Department of Medicine , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| | - Elizabeth P Henske
- a Pulmonary and Critical Care Medicine, Department of Medicine , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| |
Collapse
|
17
|
Taveira-DaSilva AM, Moss J. Addressing the challenges of lymphangioleiomyomatosis assessment in the clinic. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1400966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Angelo M. Taveira-DaSilva
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Li C, Liu X, Liu Y, Zhang E, Medepalli K, Masuda K, Li N, Wikenheiser-Brokamp KA, Osterburg A, Borchers MT, Kopras EJ, Plas DR, Sun J, Franz DN, Capal JK, Mays M, Sun Y, Kwiatkowski DJ, Alayev A, Holz MK, Krueger DA, Siroky BJ, Yu JJ. Tuberin Regulates Prostaglandin Receptor-Mediated Viability, via Rheb, in mTORC1-Hyperactive Cells. Mol Cancer Res 2017; 15:1318-1330. [PMID: 28710231 DOI: 10.1158/1541-7786.mcr-17-0077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/09/2017] [Accepted: 07/10/2017] [Indexed: 11/16/2022]
Abstract
Tuberous sclerosis complex (TSC) is a tumor-suppressor syndrome affecting multiple organs, including the brain, skin, kidneys, heart, and lungs. TSC is associated with mutations in TSC1 or TSC2, resulting in hyperactivation of mTOR complex 1 (mTORC1). Clinical trials demonstrate that mTORC1 inhibitors decrease tumor volume and stabilize lung function in TSC patients; however, mTOR inhibitors are cytostatic not cytocidal, and long-term benefits and toxicities are uncertain. Previously, we identified rapamycin-insensitive upregulation of cyclooxygenase 2 (PTGS2/COX2) and prostaglandin E2 (PGE2) production in TSC2-deficient cells and postulated that the action of excess PGE2 and its cognate receptors (EP) contributes to cell survival. In this study, we identify upregulation of EP3 (PTGER3) expression in TSC2-deficient cells, TSC renal angiomyolipomas, lymphangioleiomyomatosis lung nodules, and epileptic brain tubers. TSC2 negatively regulated EP3 expression via Rheb in a rapamycin-insensitive manner. The EP3 antagonist, L-798106, selectively suppressed the viability of TSC2-deficient cells in vitro and decreased the lung colonization of TSC2-deficient cells. Collectively, these data reveal a novel function of TSC2 and Rheb in the regulation of EP3 expression and cell viability.Implications: Therapeutic targeting of an aberrant PGE2-EP3 signaling axis may have therapeutic benefit for TSC patients and for other mTOR-hyperactive neoplasms. Mol Cancer Res; 15(10); 1318-30. ©2017 AACR.
Collapse
Affiliation(s)
- Chenggang Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xiaolei Liu
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yang Liu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Scientific Research Center for Translational Medicine, Dalian, China
| | - Erik Zhang
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kantha Medepalli
- Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Kouhei Masuda
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Na Li
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kathryn A Wikenheiser-Brokamp
- Pathology and Laboratory Medicine and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Andrew Osterburg
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael T Borchers
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Elizabeth J Kopras
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David R Plas
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julia Sun
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David N Franz
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jamie K Capal
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maxwell Mays
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yang Sun
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | | | - Anya Alayev
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, New York, New York
| | - Marina K Holz
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, New York, New York
| | - Darcy A Krueger
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Brian J Siroky
- Division of Nephrology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jane J Yu
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio. .,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Cui Y, Steagall WK, Lamattina AM, Pacheco-Rodriguez G, Stylianou M, Kidambi P, Stump B, Golzarri F, Rosas IO, Priolo C, Henske EP, Moss J, El-Chemaly S. Aberrant SYK Kinase Signaling Is Essential for Tumorigenesis Induced by TSC2 Inactivation. Cancer Res 2017; 77:1492-1502. [PMID: 28202529 DOI: 10.1158/0008-5472.can-16-2755] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 01/27/2023]
Abstract
Somatic or germline mutations in the tuberous sclerosis complex (TSC) tumor suppressor genes are associated closely with the pathogenesis of lymphangioleiomyomatosis, a rare and progressive neoplastic disease that predominantly affects women in their childbearing years. Serum levels of the lymphangiogenic growth factor VEGF-D are elevated significantly in lymphangioleiomyomatosis. However, there are gaps in knowledge regarding VEGF-D dysregulation and its cellular origin in lymphangioleiomyomatosis. Here, we show that increased expression and activation of the tyrosine kinase Syk in TSC2-deficient cells and pulmonary nodules from lymphangioleiomyomatosis patients contributes to tumor growth. Syk kinase inhibitors blocked Syk signaling and exhibited potent antiproliferative activities in TSC2-deficient cells and an immunodeficient mouse xenograft model of lymphangioleiomyomatosis. In TSC2-deficient cells, Syk signaling increased the expression of monocyte chemoattractant protein MCP-1, which in peripheral blood mononuclear cells (PBMC) stimulated the production of VEGF-D. In clinical isolates of PBMCs from lymphangioleiomyomatosis patients, VEGF-D expression was elevated. Furthermore, levels of VEGF-D and MCP-1 in patient sera correlated positively with each other. Our results illuminate the basis for lymphangioleiomyomatosis growth and demonstrate the therapeutic potential of targeting Syk in this and other settings driven by TSC genetic mutation. Cancer Res; 77(6); 1492-502. ©2017 AACR.
Collapse
Affiliation(s)
- Ye Cui
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Wendy K Steagall
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Anthony M Lamattina
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gustavo Pacheco-Rodriguez
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Mario Stylianou
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Pranav Kidambi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin Stump
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Fernanda Golzarri
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Carmen Priolo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
20
|
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
|
21
|
Courtwright AM, Goldberg HJ, Henske EP, El-Chemaly S. The effect of mTOR inhibitors on respiratory infections in lymphangioleiomyomatosis. Eur Respir Rev 2017; 26:26/143/160004. [PMID: 28096282 DOI: 10.1183/16000617.0004-2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/25/2016] [Indexed: 11/05/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a destructive cystic lung disease. Mammalian target of rapamycin (mTOR) inhibitors are the primary treatment for LAM but it is unknown whether these immunosuppressing medications increase the risk for or the severity of respiratory infections in LAM patients.We searched multiple databases for original articles that reported the rate of respiratory infections in LAM patients treated with mTOR inhibitors or placebo. We calculated incidence rates for respiratory infections in these groups and incidence rate ratios for respiratory infections and severe respiratory infections in mTOR inhibitors treated versus placebo treated patients.11 studies were included. There were 294 patients in the treatment groups and 93 patients in the placebo groups. Among subjects in placebo arms, the incidence rate of respiratory infections was 58.8 per 100 patient-years (95% CI 35.3-82.3 per 100 patient-years). The incidence-rate ratio (IRR) for respiratory infection among treated subjects was 0.71 (95% CI 0.50-1.02; p=0.06 compared to placebo subjects). The IRR for severe respiratory infections among treated subjects was 1.56 (95% CI 0.43-8.55; p=0.52).We found that respiratory infections are common in patients with LAM. Importantly, treatment with mTOR inhibitors does not increase the incidence of these infections and may be protective.
Collapse
Affiliation(s)
- Andrew M Courtwright
- Division of Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Hilary J Goldberg
- Division of Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth Petri Henske
- Division of Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
22
|
Abstract
INTRODUCTION Lymphangioleiomyomatosis (LAM) is a disease of women characterized by cystic lung destruction, lymphatic involvement, and renal angiomyolipomas. AREAS COVERED LAM is caused by proliferation of abnormal smooth muscle-like LAM cells containing mutations and perhaps epigenetic modifications of the TSC1 or TSC2 genes, which encode, respectively, hamartin and tuberin, two proteins controlling the mechanistic target of rapamycin (mTOR) signaling pathway. LAM occurs sporadically or in association with tuberous sclerosis complex. LAM may present with dyspnea, recurrent pneumothorax or chylothorax. Pulmonary function tests show reduced flow rates and lung diffusion capacity. Exercise testing may reveal hypoxemia and ventilatory limitation. The severity and progression of disease may be assessed by computer tomography, and pulmonary function and exercise testing. mTOR inhibitors, (e.g., sirolimus) are effective in stabilizing lung function, and reducing the size of chylous effusions, lymphangioleiomyomas, and angiomyolipomas. EXPERT OPINION Different clinical phenotypes including variable rates of disease progression and variable responses to therapy are seen in LAM patients. No one test is available that predicts the course of disease at the time of diagnosis. Further research regarding the molecular biology of LAM clinical phenotypes is warranted. Recent advances in the characterization of the pathogenesis of LAM are leading to the development of new therapies.
Collapse
Affiliation(s)
- Angelo M. Taveira-DaSilva
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room 6D03, MSC 1590, Bethesda, Maryland 20892-1590, USA
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room 6D03, MSC 1590, Bethesda, Maryland 20892-1590, USA
| |
Collapse
|
23
|
Atochina-Vasserman EN, Guo CJ, Abramova E, Golden TN, Sims M, James ML, Beers MF, Gow AJ, Krymskaya VP. Surfactant dysfunction and lung inflammation in the female mouse model of lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2015; 53:96-104. [PMID: 25474372 DOI: 10.1165/rcmb.2014-0224oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is a rare lung disease caused by mutations of the tumor suppressor genes, tuberous sclerosis complex (TSC) 1 or TSC2. LAM affects women almost exclusively, and it is characterized by neoplastic growth of atypical smooth muscle-like TSC2-null LAM cells in the pulmonary interstitium, cystic destruction of lung parenchyma, and progressive decline in lung function. In this study, we hypothesized that TSC2-null lesions promote a proinflammatory environment, which contributes to lung parenchyma destruction. Using a TSC2-null female murine LAM model, we demonstrate that TSC2-null lesions promote alveolar macrophage accumulation, recruitment of immature multinucleated cells, an increased induction of proinflammatory genes, nitric oxide (NO) synthase 2, IL-6, chemokine (C-C motif) ligand 2 (CCL2)/monocyte chemotactic protein 1 (MCP1), chemokine (C-X-C motif) ligand 1 (CXCL1)/keratinocyte chemoattractant (KC), and up-regulation of IL-6, KC, MCP-1, and transforming growth factor-β1 levels in bronchoalveolar lavage fluid. Bronchoalveolar lavage fluid also contained an increased level of surfactant protein (SP)-D, but not SP-A, significant reduction of SP-B levels, and a resultant increase in alveolar surface tension. Consistent with the growth of TSC2-null lesions, NO levels were also increased and, in turn, modified SP-D through S-nitrosylation, forming S-nitrosylated SP-D, a known consequence of lung inflammation. Progressive growth of TSC2-null lesions was accompanied by elevated levels of matrix metalloproteinase-3 and -9. This report demonstrates a link between growth of TSC2-null lesions and inflammation-induced surfactant dysfunction that might contribute to lung destruction in LAM.
Collapse
Affiliation(s)
- Elena N Atochina-Vasserman
- 1 Airway Biology Initiative and.,2 Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; and
| | - Chang-Jiang Guo
- 3 Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey
| | - Elena Abramova
- 3 Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey
| | - Thea N Golden
- 3 Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey
| | - Michael Sims
- 2 Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; and
| | - Melane L James
- 1 Airway Biology Initiative and.,2 Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; and
| | - Michael F Beers
- 2 Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; and
| | - Andrew J Gow
- 3 Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey
| | - Vera P Krymskaya
- 1 Airway Biology Initiative and.,2 Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; and
| |
Collapse
|
24
|
Steagall WK, Zhang L, Cai X, Pacheco-Rodriguez G, Moss J. Genetic heterogeneity of circulating cells from patients with lymphangioleiomyomatosis with and without lung transplantation. Am J Respir Crit Care Med 2015; 191:854-6. [PMID: 25830522 DOI: 10.1164/rccm.201412-2170le] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
25
|
Sun Y, Gallacchi D, Zhang EY, Reynolds SB, Robinson L, Malinowska IA, Chiou TT, Pereira AM, Li C, Kwiatkowski DJ, Lee PS, Yu JJ. Rapamycin-resistant poly (ADP-ribose) polymerase-1 overexpression is a potential therapeutic target in lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2015; 51:738-49. [PMID: 24874429 DOI: 10.1165/rcmb.2014-0033oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a female-predominant cystic lung disease that can lead to respiratory failure. LAM cells typically have inactivating tuberous sclerosis complex 2 (TSC2) mutations and mammalian target of rapamycin (mTOR) complex (mTORC) 1 activation. Clinical response to the mTORC1 inhibitors has been limited, prompting a search for additional therapy for LAM. In this study, we investigated the impact of TSC2 on the expression of poly (ADP-ribose) polymerase (PARP)-1 that initiates the DNA repair pathway, and tested the efficacy of PARP1 inhibitors in the survival of TSC2-deficient (TSC2(-)) cells. We analyzed publicly available expression arrays of TSC2(-) cells and validated the findings using real-time RT-PCR, immunoblotting, and immunohistochemistry. We examined the impact of rapamycin and Torin 1 on PARP1 expression. We also tested the effect of PARP1 inhibitors, 8-hydroxy-2-methylquinazoline-4-one and 3,4-dihydro-5[4-(1-piperindinyl)butoxy]-1(2H)-isoquinoline, on the survival of TSC2(-) cells. We identified the up-regulation of PARP1 in TSC2(-) cells relative to cells in which wild-type TSC2 has been reintroduced (TSC2-addback [TSC2(+)] cells). The transcript levels of PARP1 in TSC2(-) cells were not affected by rapamycin. PARP1 levels were increased in TSC2(-) cells, xenograft tumors of rat-derived TSC2(-) cells, renal cystadenomas from Tsc2(+/-) mice, and human LAM nodules. RNA interference of mTOR failed to reduce PARP1 levels. Proliferation and survival of TSC2(-) cells was reduced in response to PARP1 inhibitor treatment, more so than TSC2(+) cells. TSC2(-) cells exhibit higher levels of PARP1 relative to TSC2(+) cells in an mTOR-insensitive manner. PARP1 inhibitors selectively suppress the growth and induce apoptosis of TSC2(-) cells from patients with LAM. Targeting PARP1 may be beneficial in the treatment of LAM and other neoplasm with mTORC1 activation.
Collapse
Affiliation(s)
- Yang Sun
- Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yao J, Taveira-DaSilva AM, Jones AM, Julien-Williams P, Stylianou M, Moss J. Sustained effects of sirolimus on lung function and cystic lung lesions in lymphangioleiomyomatosis. Am J Respir Crit Care Med 2015; 190:1273-82. [PMID: 25329516 DOI: 10.1164/rccm.201405-0918oc] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RATIONALE Sirolimus therapy stabilizes lung function and reduces the size of chylous effusions and lymphangioleiomyomas in patients with lymphangioleiomyomatosis. OBJECTIVES To determine whether sirolimus has beneficial effects on lung function, cystic areas, and adjacent lung parenchyma; whether these effects are sustained; and whether sirolimus is well tolerated by patients. METHODS Lung function decline over time, lung volume occupied by cysts (cyst score), and lung tissue texture in the vicinity of the cysts were quantified with a computer-aided diagnosis system in 38 patients. Then we compared cyst scores from the last study on sirolimus with studies done on sirolimus therapy. In 12 patients, we evaluated rates of change in lung function and cyst scores off and on sirolimus. MEASUREMENTS AND MAIN RESULTS Sirolimus reduced yearly declines in FEV1 (-2.3 ± 0.1 vs. 1.0 ± 0.3% predicted; P < 0.001) and diffusing capacity of carbon monoxide (-2.6 ± 0.1 vs. 0.9 ± 0.2% predicted; P < 0.001). Cyst scores 1.2 ± 0.8 years (30.5 ± 11.9%) and 2.5 ± 2 years (29.7 ± 12.1%) after initiating sirolimus were not significantly different from pretreatment values (28.4 ± 12.5%). In 12 patients followed for 5 years, a significant reduction in rates of yearly decline in FEV1 (-1.4 ± 0.2 vs. 0.3 ± 0.4% predicted; P = 0.025) was observed. Analyses of 104 computed tomography scans showed a nonsignificant (P = 0.23) reduction in yearly rates of change of cyst scores (1.8 ± 0.2 vs. 0.3 ± 0.3%; P = 0.23) and lung texture features. Despite adverse events, most patients were able to continue sirolimus therapy. CONCLUSIONS Sirolimus therapy slowed down lung function decline and increase in cystic lesions. Most patients were able to tolerate sirolimus therapy.
Collapse
|
27
|
Li C, Zhang E, Sun Y, Lee PS, Zhan Y, Guo Y, Osorio JC, Rosas IO, Xu KF, Kwiatkowski DJ, Yu JJ. Rapamycin-insensitive up-regulation of adipocyte phospholipase A2 in tuberous sclerosis and lymphangioleiomyomatosis. PLoS One 2014; 9:e104809. [PMID: 25347447 PMCID: PMC4210122 DOI: 10.1371/journal.pone.0104809] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/14/2014] [Indexed: 11/18/2022] Open
Abstract
Tuberous sclerosis syndrome (TSC) is an autosomal dominant tumor suppressor gene syndrome affecting multiple organs, including renal angiomyolipomas and pulmonary lymphangioleiomyomatosis (LAM). LAM is a female-predominant interstitial lung disease characterized by the progressive cyst formation and respiratory failure, which is also seen in sporadic patients without TSC. Mutations in TSC1 or TSC2 cause TSC, result in hyperactivation of mammalian target of rapamycin (mTOR), and are also seen in LAM cells in sporadic LAM. We recently reported that prostaglandin biosynthesis and cyclooxygenase-2 were deregulated in TSC and LAM. Phospholipase A2 (PLA2) is the rate-limiting enzyme that catalyzes the conversion of plasma membrane phospholipids into prostaglandins. In this study, we identified upregulation of adipocyte AdPLA2 (PLA2G16) in LAM nodule cells using publicly available expression data. We showed that the levels of AdPLA2 transcript and protein were higher in LAM lungs compared with control lungs. We then showed that TSC2 negatively regulates the expression of AdPLA2, and loss of TSC2 is associated with elevated production of prostaglandin E2 (PGE2) and prostacyclin (PGI2) in cell culture models. Mouse model studies also showed increased expression of AdPLA2 in xenograft tumors, estrogen-induced lung metastatic lesions of Tsc2 null leiomyoma-derived cells, and spontaneous renal cystadenomas from Tsc2+/- mice. Importantly, rapamycin treatment did not affect the expression of AdPLA2 and the production of PGE2 by TSC2-deficient mouse embryonic fibroblast (Tsc2-/-MEFs), rat uterine leiomyoma-derived ELT3 cells, and LAM patient-associated renal angiomyolipoma-derived "mesenchymal" cells. Furthermore, methyl arachidonyl fluorophosphate (MAFP), a potent irreversible PLA2 inhibitor, selectively suppressed the growth and induced apoptosis of TSC2-deficient LAM patient-derived cells relative to TSC2-addback cells. Our findings suggest that AdPLA2 plays an important role in promoting tumorigenesis and disease progression by modulating the production of prostaglandins and may serve as a potential therapeutic target in TSC and LAM.
Collapse
Affiliation(s)
- Chenggang Li
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Erik Zhang
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yang Sun
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Po-Shun Lee
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Yanan Guo
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Juan C. Osorio
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ivan O. Rosas
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kai-Feng Xu
- Peking Union Medical College, Beijing, China
| | - David J. Kwiatkowski
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jane J. Yu
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
28
|
Weckmann M, Moir LM, Heckman CA, Black JL, Oliver BG, Burgess JK. Lamstatin--a novel inhibitor of lymphangiogenesis derived from collagen IV. J Cell Mol Med 2014; 16:3062-73. [PMID: 22998238 PMCID: PMC4393734 DOI: 10.1111/j.1582-4934.2012.01648.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 09/12/2012] [Indexed: 12/31/2022] Open
Abstract
The lymphatic system is essential for the maintenance of tissue homeostasis and immunity. Its dysfunction in disease (such as lymphangioleiomyomatosis) can lead to chylous effusions, oedema or dissemination of malignant cells. Collagen IV has six α chains, of which some of the non-collagenous-1 domains have endogenous anti-angiogenic properties, however, little is known about specific endogenous anti-lymphangiogenic characteristics. In this study we sought to investigate the expression levels of collagen IV non-collagenous-1 domains in lung tissue of patients with and without lymphangioleiomyomatosis to explore the hypothesis that a member of the collagen IV family, specifically the non-collagenous domain-1 of α5, which we named lamstatin, has anti-lymphangiogenic properties. Levels of lamstatin detected by immunohistochemistry were decreased in lungs of lymphangioleiomyomatosis patients. We produced recombinant lamstatin in an E.coli expression system and synthesized a 17-amino acid peptide from a theoretically identified, active region (CP17) and tested their effects in vitro and in vivo. Recombinant lamstatin and CP17 inhibited proliferation, migration and cord formation of human microvascular lung lymphatic endothelial cells, in vitro. Furthermore, lamstatin and CP17 decreased complexity and dysplasia of the tumour-associated lymphatic network in a lung adenocarcinoma xenograft mouse model. In this study we identified a novel, direct inhibitor of lymphangiogenesis, derived from collagen IV. This may prove useful for exploring new avenues of treatment for lymphangioleiomyomatosis and metastasis via the lymphatic system in general.
Collapse
Affiliation(s)
- Markus Weckmann
- Woolcock Institute of Medical Research, Glebe, NSW, Australia
| | | | | | | | | | | |
Collapse
|
29
|
Osteoprotegerin contributes to the metastatic potential of cells with a dysfunctional TSC2 tumor-suppressor gene. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:938-50. [PMID: 23867796 DOI: 10.1016/j.ajpath.2013.05.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 04/29/2013] [Accepted: 05/28/2013] [Indexed: 01/15/2023]
Abstract
In addition to its effects on bone metabolism, osteoprotegerin (OPG), a soluble member of the tumor necrosis factor family of receptors, promotes smooth muscle cell proliferation and migration and may act as a survival factor for tumor cells. We hypothesized that these cellular mechanisms of OPG may be involved in the growth and proliferation of lymphangioleiomyomatosis (LAM) cells, abnormal smooth muscle-like cells with mutations in one of the tuberous sclerosis complex tumor-suppressor genes (TSC1/TSC2) that cause LAM, a multisystem disease characterized by cystic lung destruction, lymphatic infiltration, and abdominal tumors. Herein, we show that OPG stimulated proliferation of cells cultured from explanted LAM lungs, and selectively induced migration of LAM cells identified by the loss of heterozygosity for TSC2. Consistent with these observations, cells with TSC2 loss of heterozygosity expressed the OPG receptors, receptor activator of NF-κB ligand, syndecan-1, and syndecan-2. LAM lung nodules showed reactivities to antibodies to tumor necrosis factor-related apoptosis-inducing ligand, receptor activator of NF-κB ligand, syndecan-1, and syndecan-2. LAM lung nodules also produced OPG, as shown by expression of OPG mRNA and colocalization of reactivities to anti-OPG and anti-gp100 (HMB45) antibodies in LAM lung nodules. Serum OPG was significantly higher in LAM patients than in normal volunteers. Based on these data, it appears that OPG may have tumor-promoting roles in the pathogenesis of lymphangioleiomyomatosis, perhaps acting as both autocrine and paracrine factors.
Collapse
|
30
|
Trindade AJ, Medvetz DA, Neuman NA, Myachina F, Yu J, Priolo C, Henske EP. MicroRNA-21 is induced by rapamycin in a model of tuberous sclerosis (TSC) and lymphangioleiomyomatosis (LAM). PLoS One 2013; 8:e60014. [PMID: 23555865 PMCID: PMC3612076 DOI: 10.1371/journal.pone.0060014] [Citation(s) in RCA: 28] [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: 12/27/2012] [Accepted: 02/20/2013] [Indexed: 01/04/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM), a multisystem disease of women, is manifest by the proliferation of smooth muscle-like cells in the lung resulting in cystic lung destruction. Women with LAM can also develop renal angiomyolipomas. LAM is caused by mutations in the tuberous sclerosis complex genes (TSC1 or TSC2), resulting in hyperactive mammalian Target of Rapamycin (mTOR) signaling. The mTOR inhibitor, Rapamycin, stabilizes lung function in LAM and decreases the volume of renal angiomyolipomas, but lung function declines and angiomyolipomas regrow when treatment is discontinued, suggesting that factors induced by mTORC1 inhibition may promote the survival of TSC2-deficient cells. Whether microRNA (miRNA, miR) signaling is involved in the response of LAM to mTORC1 inhibition is unknown. We identified Rapamycin-dependent miRNA in LAM patient angiomyolipoma-derived cells using two separate screens. First, we assayed 132 miRNA of known significance to tumor biology. Using a cut-off of >1.5-fold change, 48 microRNA were Rapamycin-induced, while 4 miRs were downregulated. In a second screen encompassing 946 miRNA, 18 miRs were upregulated by Rapamycin, while eight were downregulated. Dysregulation of miRs 29b, 21, 24, 221, 106a and 199a were common to both platforms and were classified as candidate “RapamiRs.” Validation by qRT-PCR confirmed that these microRNA were increased. miR-21, a pro-survival miR, was the most significantly increased by mTOR-inhibition (p<0.01). The regulation of miR-21 by Rapamycin is cell type independent. mTOR inhibition promotes the processing of the miR-21 transcript (pri-miR-21) to a premature form (pre-miR-21). In conclusion, our findings demonstrate that Rapamycin upregulates multiple miRs, including pro-survival miRs, in TSC2-deficient patient-derived cells. The induction of miRs may contribute to the response of LAM and TSC patients to Rapamycin therapy.
Collapse
Affiliation(s)
- Anil J. Trindade
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Douglas A. Medvetz
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nicole A. Neuman
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Faina Myachina
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jane Yu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carmen Priolo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elizabeth P. Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Goncharova EA, Goncharov DA, Fehrenbach M, Khavin I, Ducka B, Hino O, Colby TV, Merrilees MJ, Haczku A, Albelda SM, Krymskaya VP. Prevention of alveolar destruction and airspace enlargement in a mouse model of pulmonary lymphangioleiomyomatosis (LAM). Sci Transl Med 2013; 4:154ra134. [PMID: 23035046 DOI: 10.1126/scitranslmed.3003840] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is a rare genetic disease characterized by neoplastic growth of atypical smooth muscle-like LAM cells, destruction of lung parenchyma, obstruction of lymphatics, and formation of lung cysts, leading to spontaneous pneumothoraces (lung rupture and collapse) and progressive loss of pulmonary function. The disease is caused by mutational inactivation of the tumor suppressor gene tuberous sclerosis complex 1 (TSC1) or TSC2. By injecting TSC2-null cells into nude mice, we have developed a mouse model of LAM that is characterized by multiple random TSC2-null lung lesions, vascular endothelial growth factor-D expression, lymphangiogenesis, destruction of lung parenchyma, and decreased survival, similar to human LAM. The mice show enlargement of alveolar airspaces that is associated with progressive growth of TSC2-null lesions in the lung, up-regulation of proinflammatory cytokines and matrix metalloproteinases (MMPs) that degrade extracellular matrix, and destruction of elastic fibers. TSC2-null lesions and alveolar destruction were differentially inhibited by the macrolide antibiotic rapamycin (which inhibits TSC2-null lesion growth by a cytostatic mechanism) and a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, simvastatin (which inhibits growth of TSC2-null lesions by a predominantly proapoptotic mechanism). Treatment with simvastatin markedly inhibited MMP-2, MMP-3, and MMP-9 levels in lung and prevented alveolar destruction. The combination of rapamycin and simvastatin prevented both growth of TSC2-null lesions and lung destruction by inhibiting MMP-2, MMP-3, and MMP-9. Our findings demonstrate a mechanistic link between loss of TSC2 and alveolar destruction and suggest that treatment with rapamycin and simvastatin together could benefit patients with LAM by targeting cells with TSC2 dysfunction and preventing airspace enlargement.
Collapse
Affiliation(s)
- Elena A Goncharova
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Glasgow CG, El-Chemaly S, Moss J. Lymphatics in lymphangioleiomyomatosis and idiopathic pulmonary fibrosis. Eur Respir Rev 2013; 21:196-206. [PMID: 22941884 DOI: 10.1183/09059180.00009311] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The primary function of the lymphatic system is absorbing and transporting macromolecules and immune cells to the general circulation, thereby regulating fluid, nutrient absorption and immune cell trafficking. Lymphangiogenesis plays an important role in tissue inflammation and tumour cell dissemination. Lymphatic involvement is seen in lymphangioleiomyomatosis (LAM) and idiopathic pulmonary fibrosis (IPF). LAM, a disease primarily affecting females, involves the lung (cystic destruction), kidney (angiomyolipoma) and axial lymphatics (adenopathy and lymphangioleiomyoma). LAM occurs sporadically or in association with tuberous sclerosis complex (TSC). Cystic lung destruction results from proliferation of LAM cells, which are abnormal smooth muscle-like cells with mutations in the TSC1 or TSC2 gene. Lymphatic abnormalities arise from infiltration of LAM cells into the lymphatic wall, leading to damage or obstruction of lymphatic vessels. Benign appearing LAM cells possess metastatic properties and are found in the blood and other body fluids. IPF is a progressive lung disease resulting from fibroblast proliferation and collagen deposition. Lymphangiogenesis is associated with pulmonary destruction and disease severity. A macrophage subset isolated from IPF bronchoalveolar lavage fluid (BALF) express lymphatic endothelial cell markers in vitro, in contrast to the same macrophage subset from normal BALF. Herein, we review lymphatic involvement in LAM and IPF.
Collapse
Affiliation(s)
- Connie G Glasgow
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1590, USA
| | | | | |
Collapse
|
33
|
Breast tumor-associated osteoblast-derived CXCL5 increases cancer progression by ERK/MSK1/Elk-1/snail signaling pathway. Oncogene 2012; 32:4436-47. [PMID: 23045282 DOI: 10.1038/onc.2012.444] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 08/13/2012] [Accepted: 08/16/2012] [Indexed: 12/17/2022]
Abstract
The skeleton is the most common metastatic site for breast cancer, with bone metastasis causing pain as well as risk of pathological fractures. Interaction between tumors and the bone microenvironment creates a vicious cycle that accelerates both bone destruction and cancer progression. This study is the first to analyze the soluble factors secreted by breast tumor-associated osteoblasts (TAOBs), which are responsible for promoting cancer progression. The addition of CXCL5 (chemokine (C-X-C motif) ligand 5), present in large amounts in TAOB-condition medium (TAOB-CM), mimicked the inductive effect of TAOB-CM on breast cancer epithelial-mesenchymal transition, migration and invasion. In contrast, inhibition of CXCL5 in OBs decreased TAOB-mediated cancer progression. Inducement of MCF-7 and MDA-MB-231 cancer progression by TAOB-derived CXCL5 is associated with increased Raf/MEK/ERK activation, and mitogen- and stress-activated protein kinase 1 (MSK1) and Elk-1 phosphorylation, as well as Snail upregulation. Activation of Elk-1 facilitates recruitment of phosphorylated MSK1, which in turn enhances histone H3 acetylation and phosphorylation (serine 10) of Snail promoter, resulting in Snail enhancement and E-cadherin downregulation. Moreover, mice treated with anti-CXCL5 antibodies showed decreased metastasis of 4T1 breast cancer cells. Our study suggests that inhibition of CXCL5-mediated ERK/Snail signaling is an attractive therapeutic target for treating metastases in breast cancer patients.
Collapse
|
34
|
Abstract
Chemokines are best known for their classic leukocyte chemotactic activity, which is critical for directing the immune response to sites of infection and injury. However, recent studies have suggested that at least some chemokines may also interfere with infectious agents directly. Antimicrobial chemokines tend to contain amphipathic alpha helical secondary structure, and broad-spectrum activity against both Gram-positive and Gram negative bacteria, as well as fungi. Conversely, several bacteria have been identified that possess mechanisms for specifically blocking the antimicrobial activities of chemokines. Although the precise mechanisms by which chemokines and microbes disarm one another in vitro remain unknown, there is now emerging evidence in vivo that such interactions may be biologically significant. More research will be needed to determine whether chemokines with direct antimicrobial activity may be translated into a novel class of antibiotics.
Collapse
Affiliation(s)
- Sunny C. Yung
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesda, MD, USA
| | - Philip M. Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesda, MD, USA
| |
Collapse
|
35
|
Abstract
Lymphangioleiomyomatosis (LAM), a multisystem disease predominantly affecting premenopausal women, is associated with cystic lung destruction and lymphatic and kidney tumors. LAM results from the proliferation of a neoplastic cell that has mutations in the tuberous sclerosis complex 1 or 2 genes, leading to activation of a critical regulatory protein, mammalian target of rapamycin. In this report, we discuss the molecular mechanisms regulating LAM cell growth and report the results of therapeutic trials employing new targeted agents. At present, inhibitors of mammalian target of rapamycin such as sirolimus appear to be the most promising therapeutic agents, although drug toxicity and development of resistance are potential problems. As the pathogenesis of LAM is being further recognized, other therapeutic agents such as matrix metalloproteinase inhibitors, statins, interferon, VEGF inhibitors, chloroquine analogs and cyclin-dependent kinase inhibitors, along with sirolimus or a combination of several of these agents, may offer the best hope for effective therapy.
Collapse
Affiliation(s)
- Angelo M Taveira-DaSilva
- Cardiovascular and Pulmonary Branch, Bldg. 10, Rm. 6D05, MSC 1590, NHLBI, NIH, Bethesda, MD 20892-1590, USA
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, Bldg. 10, Rm. 6D05, MSC 1590, NHLBI, NIH, Bethesda, MD 20892-1590, USA
| |
Collapse
|
36
|
Schulte-Merker S, Sabine A, Petrova TV. Lymphatic vascular morphogenesis in development, physiology, and disease. ACTA ACUST UNITED AC 2011; 193:607-18. [PMID: 21576390 PMCID: PMC3166860 DOI: 10.1083/jcb.201012094] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lymphatic vasculature constitutes a highly specialized part of the vascular system that is essential for the maintenance of interstitial fluid balance, uptake of dietary fat, and immune response. Recently, there has been an increased awareness of the importance of lymphatic vessels in many common pathological conditions, such as tumor cell dissemination and chronic inflammation. Studies of embryonic development and genetically engineered animal models coupled with the discovery of mutations underlying human lymphedema syndromes have contributed to our understanding of mechanisms regulating normal and pathological lymphatic morphogenesis. It is now crucial to use this knowledge for the development of novel therapies for human diseases.
Collapse
Affiliation(s)
- Stefan Schulte-Merker
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, and University Medical Centre, 3584 CT Utrecht, Netherlands
| | | | | |
Collapse
|
37
|
Zengel P, Nguyen-Hoang A, Schildhammer C, Zantl R, Kahl V, Horn E. μ-Slide Chemotaxis: a new chamber for long-term chemotaxis studies. BMC Cell Biol 2011; 12:21. [PMID: 21592329 PMCID: PMC3118187 DOI: 10.1186/1471-2121-12-21] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 05/18/2011] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Effective tools for measurement of chemotaxis are desirable since cell migration towards given stimuli plays a crucial role in tumour metastasis, angiogenesis, inflammation, and wound healing. As for now, the Boyden chamber assay is the longstanding "gold-standard" for in vitro chemotaxis measurements. However, support for live cell microscopy is weak, concentration gradients are rather steep and poorly defined, and chemotaxis cannot be distinguished from migration in a single experiment. RESULTS Here, we describe a novel all-in-one chamber system for long-term analysis of chemotaxis in vitro that improves upon many of the shortcomings of the Boyden chamber assay. This chemotaxis chamber was developed to provide high quality microscopy, linear concentration gradients, support for long-term assays, and observation of slowly migrating cells via video microscopy. AlexaFluor 488 dye was used to demonstrate the establishment, shape and time development of linear chemical gradients. Human fibrosarcoma cell line HT1080 and freshly isolated human umbilical vein endothelial cells (HUVEC) were used to assess chemotaxis towards 10% fetal calf serum (FCS) and FaDu cells' supernatant. Time-lapse video microscopy was conducted for 48 hours, and cell tracking and analysis was performed using ImageJ plugins. The results disclosed a linear steady-state gradient that was reached after approximately 8 hours and remained stable for at least 48 hours. Both cell types were chemotactically active and cell movement as well as cell-to-cell interaction was assessable. CONCLUSIONS Compared to the Boyden chamber assay, this innovative system allows for the generation of a stable gradient for a much longer time period as well as for the tracking of cell locomotion along this gradient and over long distances. Finally, random migration can be distinguished from primed and directed migration along chemotactic gradients in the same experiment, a feature, which can be qualified via cell morphology imaging.
Collapse
Affiliation(s)
- Pamela Zengel
- Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Centre, Ludwig-Maximilians-University of Munich, Munich, Germany.
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
LAM is a rare lung disease, found primarily in women of childbearing age, characterized by cystic lung destruction and abdominal tumors (e.g., renal angiomyolipoma, lymphangioleiomyoma). The disease results from proliferation of a neoplastic cell, termed the LAM cell, which has mutations in either of the tuberous sclerosis complex (TSC) 1 or TSC2 genes. Molecular phenotyping of LAM patients resulted in the identification of therapeutic targets for drug trials. Loss of TSC gene function leads to activation of mammalian target of rapamycin (mTOR), and thereby, effects on cell size and number. The involvement of mTOR in LAM pathogenesis is the basis for initiation of therapeutic trials of mTOR inhibitors (e.g., sirolimus). Occurrence of LAM essentially entirely in women is consistent with the hypothesis that anti-estrogen agents might prevent disease progression (e.g., gonadotropin-releasing hormone analogues). Levels of urinary matrix metalloproteinases (MMPs) were elevated in LAM patients, and MMPs were found in LAM lung nodules. In part because of these observations, effects of doxycycline, an anti-MMP, and anti-angiogenic agent, are under investigation. The metastatic properties of LAM cells offer additional potential for targets. Thus, insights into the molecular and biological properties of LAM cells and molecular phenotyping of patients with LAM have led to clinical trials of targeted therapies. Funded by the Intramural Research Program, NIH/NHLBI.
Collapse
|
39
|
Pacheco-Rodriguez G, Moss J. The role of chemokines in migration of metastatic-like lymphangioleiomyomatosis cells. Crit Rev Immunol 2010; 30:387-94. [PMID: 20666708 DOI: 10.1615/critrevimmunol.v30.i4.40] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Lymphangioleiomyomatosis (LAM), a rare cystic lung disease with multi-organ involvement, occurs primarily in women of childbearing age. LAM can present sporadically or in association with tuberous sclerosis complex (TSC). Loss of lung function in patients with LAM can be attributed to the dysregulated growth of LAM cells, with dysfunctional TSC1 or TSC2 genes, which encode hamartin and tuberin, respectively, leading to hyperactivation of the mammalian target of rapamycin (mTOR). LAM cells are smooth muscle-like cells that express melanoma antigens such as gp100, a splice variant of the Pmel17 gene. Tuberin and hamartin form heterodimers that act as negative regulators of mTOR. Lack of TSC2 function, as occurs in LAM cells, leads to the production of the chemokine CCL2/monocyte chemotactic protein 1 (MCP-1), which increases LAM cell mobility. Although many chemokines and their receptors could influence LAM cell mobilization, we propose that a positive-feedback loop is generated when dysfunctional TSC2 is present in LAM cells. We identified a group of chemokine receptors that is expressed in LAM cells and differs from those on smooth muscle and melanoma cells (Malme-3M). Chemokines have been implicated in tumor metastasis, and our data suggest a role for chemokines in LAM cell mobilization and thereby in the pathogenesis of LAM.
Collapse
Affiliation(s)
- Gustavo Pacheco-Rodriguez
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1590, USA
| | | |
Collapse
|
40
|
Clements D, Markwick LJ, Puri N, Johnson SR. Role of the CXCR4/CXCL12 Axis in Lymphangioleiomyomatosis and Angiomyolipoma. THE JOURNAL OF IMMUNOLOGY 2010; 185:1812-21. [DOI: 10.4049/jimmunol.0902149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
41
|
Yu J, Henske EP. mTOR activation, lymphangiogenesis, and estrogen-mediated cell survival: the "perfect storm" of pro-metastatic factors in LAM pathogenesis. Lymphat Res Biol 2010; 8:43-9. [PMID: 20235886 DOI: 10.1089/lrb.2009.0020] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Research interest in lymphangioleiomyomatosis (LAM) has grown dramatically in the past decade, particularly among cancer biologists. There are at least two reasons for this: first, the discovery in the year 2000 that LAM cells carry TSC2 gene mutations, linking LAM with cellular pathways including the PI3K/Akt/mTOR axis, and allowing the Tuberous Sclerosis Complex (TSC)-regulated pathways that are believed to underlie LAM pathogenesis to be studied in cells, yeast, Drosophila, and mice. A second reason for the rising interest in LAM is the discovery that LAM cells can travel to the lung, including repopulating a donor lung after lung transplantation, despite the fact that LAM cells are histologically benign. This "benign metastasis" underpinning suggests that elucidating LAM pathogenesis will unlock a set of fundamental mechanisms that underlie metastatic potential in the context of a cell that has not yet undergone malignant transformation. Here, we will outline the data supporting the metastatic model of LAM, consider the biochemical and cellular mechanisms that may enable LAM cells to metastasize, including both cell autonomous and non-cell autonomous factors, and highlight a mouse model in which estrogen promotes the metastasis and survival of TSC2-deficient cells in a MEK-dependent manner. We propose a multistep model of LAM cell metastasis that highlights multiple opportunities for therapeutic intervention. Taken together, the metastatic behavior of LAM cells and the involvement of tumor-related signaling pathways lead to optimism that cancer-related paradigms for diagnosis, staging, and therapy will lead to therapeutic breakthroughs for women living with LAM.
Collapse
Affiliation(s)
- Jane Yu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
42
|
Taveira-DaSilva AM, Pacheco-Rodriguez G, Moss J. The natural history of lymphangioleiomyomatosis: markers of severity, rate of progression and prognosis. Lymphat Res Biol 2010; 8:9-19. [PMID: 20235883 DOI: 10.1089/lrb.2009.0024] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a multisystem disease of women, characterized by proliferation of abnormal smooth muscle-like cells (LAM cells) that can metastasize, leading to the formation of lung cysts, fluid-filled cystic structures in the axial lymphatics (e.g., lymphangioleiomyomas), and angiomyolipomas, benign tumors usually involving the kidneys, comprising LAM cells and adipocytes, intermixed with incompletely developed vascular structures. LAM occurs sporadically or in association with tuberous sclerosis complex, an autosomal dominant syndrome characterized by hamartoma-like tumor growths. LAM may present with progressive dyspnea, recurrent pneumothorax, chylothorax, or abdominal hemorrhage. Computed tomography scans show thin-walled cysts scattered throughout the lungs, abdominal angiomyolipomas, and lymphangioleiomyomas. Pulmonary function tests show reduced flow rates (FEV(1)) and diffusion capacity (DL(CO)). Exercise testing may reveal gas exchange abnormalities, ventilatory limitation, and hypoxemia, which can occur with near-normal lung function. Methods used to grade the severity of disease are the LAM histology score, semiquantitative and quantitative computer tomography, pulmonary function testing, and cardiopulmonary exercise testing. Currently, progression of disease is best assessed by serial measurements of FEV(1), DL(CO), and exercise performance. New quantitative radiographic techniques that may offer advantages over physiologic testing are now available. Several potential biomarkers, such as LAM cells in peripheral blood, urine, and chyle and chemokines, vascular endothelial growth factors, and matrix metalloproteinases, may be useful as diagnostic tools or markers of organ involvement, disease severity, and progression.
Collapse
Affiliation(s)
- Angelo M Taveira-DaSilva
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1590, USA.
| | | | | |
Collapse
|
43
|
Darling TN, Pacheco-Rodriguez G, Gorio A, Lesma E, Walker C, Moss J. Lymphangioleiomyomatosis and TSC2-/- cells. Lymphat Res Biol 2010; 8:59-69. [PMID: 20235888 DOI: 10.1089/lrb.2009.0031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The cells comprising pulmonary lymphangioleiomyomatosis (LAM) and renal angiomyolipomas (AMLs) are heterogeneous, with variable mixtures of cells exhibiting differentiation towards smooth muscle, fat, and vessels. Cells grown from LAM and AMLs have likewise tended to be heterogeneous. The discovery that LAM and AMLs contain cells with mutations in the TSC1 or TSC2 genes is allowing investigators to discriminate between "two-hit" cells and neighboring cells, providing insights into disease pathogenesis. In rare cases, it has been possible to derive cells from human tumors, including AMLs and TSC skin tumors that are highly enriched for TSC2(-/-) cells. Cells derived from an Eker rat uterine leiomyoma (ELT3 cells) are Tsc2-null and these have been used in a rodent cell models for LAM. Further improvements in the ability to reliably grow well-characterized TSC2(-/-) cells from human tumors are critical to developing in vitro and in vivo model systems for studies of LAM pathogenesis and treatment.
Collapse
Affiliation(s)
- Thomas N Darling
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Glasgow CG, Taveira-DaSilva A, Pacheco-Rodriguez G, Steagall WK, Tsukada K, Cai X, El-Chemaly S, Moss J. Involvement of lymphatics in lymphangioleiomyomatosis. Lymphat Res Biol 2010; 7:221-8. [PMID: 20143921 DOI: 10.1089/lrb.2009.0017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM), a rare multisystem disease, occurs primarily in women, with cystic destruction of the lungs, abdominal tumors, and involvement of the axial lymphatics in the thorax and abdomen. To understand the pathogenesis of LAM, we initiated a longitudinal study of patients with LAM; over 500 patients have been enrolled. LAM results from the proliferation of a neoplastic cell (LAM cell), which has mutations in the tuberous sclerosis complex (TSC) genes, TSC1 or TSC2. Consistent with their metastatic behavior, LAM cells were isolated from blood, urine, and chylous effusions. Surface proteins on LAM cells include those found on metastatic cells and those involved in cell migration. In the lung, LAM cells are found clustered in nodules, which appear in the walls of the cysts, and in the interstitium. LAM lung nodules are traversed by slit-like vascular structures, with lining cells showing reactivity with antibodies against components of lymphatic endothelial cells. The axial lymphatics appear to be infiltrated by LAM cells, which may result in obstruction and formation of chyle-filled lymphangioleiomyomas. LAM cell clusters have been isolated from chylous pleural effusions, and it is hypothesized that these clusters may be responsible for metastatic spread of LAM cells via lymphatic vessels. Consistent with a lymphangiogenic process, levels of VEGF-D, a lymphangiogenic factor, were higher in sera of patients with LAM and lymphatic involvement (i.e., lymphangioleiomyoma, adenopathy) than in healthy volunteers or LAM patients with cystic disease limited to the lung. These findings are consistent with an important function for lymphangiogenesis in LAM.
Collapse
Affiliation(s)
- Connie G Glasgow
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1590, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Odajima N, Betsuyaku T, Yoshida T, Hosokawa T, Nishimura M. High levels of extracellular matrix metalloproteinase inducer are expressed in lymphangioleiomyomatosis. Hum Pathol 2010; 41:935-43. [PMID: 20236683 DOI: 10.1016/j.humpath.2009.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 12/01/2009] [Accepted: 12/02/2009] [Indexed: 10/19/2022]
Abstract
Pulmonary lymphangioleiomyomatosis is pathologically characterized by the proliferation of abnormal smooth muscle-like cells (lymphangioleiomyomatosis cells) that synthesize excess matrix metalloproteinases. Extracellular matrix metalloproteinase inducer is minimally expressed in the healthy lung, but is up-regulated in various lung injuries that are apparently associated with matrix metalloproteinases. We therefore immunohistochemically stained extracellular matrix metalloproteinase inducer in lymphangioleiomyomatosis cells and measured extracellular matrix metalloproteinase inducer in bronchoalveolar lavage fluid using an enzyme-linked immunosorbent assay. We also quantified extracellular matrix metalloproteinase inducer messenger RNA expression in the lung using quantitative reverse transcriptase-polymerase chain reaction. Intense staining for extracellular matrix metalloproteinase inducer in lymphangioleiomyomatosis cells indicated high immunoreactivity. Dual immunofluorescence studies revealed diffuse colocalization between extracellular matrix metalloproteinase inducer and alpha smooth muscle actin, matrix metalloproteinase-2, or matrix metalloproteinase-9. Although levels of extracellular matrix metalloproteinase inducer messenger RNA did not differ in whole lung homogenates from patients with lymphangioleiomyomatosis and healthy controls (1.7 +/- 0.1 versus 1.5 +/- 0.2 SE, not significant; both n = 4), lymphangioleiomyomatosis nodules retrieved by laser capture microdissection expressed 5.1 (+/-1.0 SE)-fold higher levels of extracellular matrix metalloproteinase inducer messenger RNA than lung homogenates (P = .0077, n = 4). Levels of extracellular matrix metalloproteinase inducer were significantly elevated in bronchoalveolar lavage fluid from lymphangioleiomyomatosis patients compared with controls (82.7 +/- 19.5 versus 38.6 +/- 9.0 SE pg/mL, P = .0497; n = 8 and 9, respectively). Increased levels of extracellular matrix metalloproteinase inducer colocalized with increased matrix metalloproteinases in lymphangioleiomyomatosis cells indicate that it potentially functions in pulmonary lymphangioleiomyomatosis.
Collapse
Affiliation(s)
- Nao Odajima
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo 060-8638, Japan
| | | | | | | | | |
Collapse
|