Abstract

EDITORIAL FOCUSThe LAM cell: what is it, where does it come from, and why does it grow?G. FinlayG. FinlayTupper Research Institute, Tufts-New England Medical Center, Boston, Massachusetts 02111Published Online:01 Apr 2004https://doi.org/10.1152/ajplung.00311.2003MoreSectionsPDF (32 KB)Download PDF ToolsExport citationAdd to favoritesGet permissionsTrack citations ShareShare onFacebookTwitterLinkedInEmailWeChat lymphangioleiomyomatosis (LAM) is a rare disease that affects females of reproductive age (38). Pathologically, it is characterized by the abnormal proliferation of immature smooth muscle-like cells that grow aberrantly in the lung. The relentless growth of LAM cells in the pulmonary airway, parenchyma, lymphatics, and blood vessels leads to respiratory failure and death. Recent progress has been made in understanding more about the pathogenesis of LAM. Studies such as those published by Yu et al., one of the current articles in focus (Ref. 42, see page in this issue), deepen our understanding of the disease and facilitate the identification of potential therapeutic targets aimed at abrogating the aberrant cellular proliferation characteristic of LAM.The LAM Cell: What Is It?To understand the pathogenesis of LAM, one needs to have a keen understanding of the nature of the atypical cell so characteristic of LAM. In 1966, Cornog and Enterline (8) described the first significant collection of patients with LAM. These insightful observers postulated that the cell present in LAM lung was smooth muscle like, lacked the features of a malignancy, and perhaps shared a common genetic origin. Indeed, the cells found in LAM are not malignant and stain positive for smooth muscle markers, α-smooth muscle actin, desmin, and vimentin (9). What is unusual about LAM is that it is not due to the proliferation of one monoclonal cell type only but instead due to the infiltration and proliferation of a phenotypically heterogeneous group of smooth muscle cells, many of which have myofibroblast- and epithelioid-like features (4). Interestingly, LAM cells are unusually reactive to mouse monoclonal antibody HMB45 (20a), which reacts with a glycoprotein (gp100) found in immature melanocytes. However, considerable variability of HMB45 positivity is frequently observed and is not absolutely necessary for the diagnosis of LAM (20a, 30). Although the relevance of HMB45 positivity and its antigen gp100 is as yet unstudied, an inverse relationship has been described in LAM smooth muscle cells between immunostaining for HMB45 and proliferating cell nuclear antigen (PCNA), which is a marker of cell proliferation (30). Thin, spindle-shaped smooth muscle cells are less likely to be immunoreactive for HMB45 and more likely to be PCNA positive. In contrast, larger epithelioid-like smooth muscle cells are more likely to exhibit positive immunostaining for HMB45 with poor or no reactivity for PCNA. These observations have led to the hypothesis that LAM cells are a population of smooth muscle cells that at any one point in time may be in a state of variable differentiation (22). Furthermore, it is reasonable to postulate that each phenotype is associated with a cell-specific function, at least in terms of its proliferative capacity, which may help to explain why many patients with LAM have variable responses to therapy and outcomes.Cornog and Enterline (8) also postulated in 1966 that the smooth muscle cells may share a common genetic origin. They noted that pulmonary LAM is histologically indistinct from the pulmonary lesions observed in tuberous sclerosis complex (TSC), and recent reports indicate that LAM has a prevalence as high as 39% in TSC (10, 31, 32). TSC, which is characterized by hamartomatous proliferations in the brain, kidney, skin, heart, and lung, is caused by germline mutations of either the TSC1 or TSC2 gene located on chromosomes 9q34 and 16p13, respectively (18). It is now clear that both of these genes are tumor suppressor genes encoding hamartin (TSC1) and tuberin (TSC2) and that the tumors associated with TSC have undergone loss of heterozygosity (LOH), indicating that tumor development in TSC is associated with the “two-hit” hypothesis of Knudson (5). In 1998, the discovery of LOH of the TSC2 gene in the angiomyolipomas (AMLs) and lymph nodes of women who suffer from sporadic LAM heralded a new wave of investigation of the role of the TSC genes and, in particular, the TSC2 gene in sporadic LAM (36). Subsequently, mutational analysis of the 41 coding exons of the TSC2 gene from LAM patients revealed that germline mutations of the TSC2 or TSC1 genes are extremely uncommon (1). Additional analysis of microdissected smooth muscle cells from renal AMLs and pulmonary LAM lesions revealed that mutations of the TSC2 gene in renal and pulmonary smooth muscle cells are not found in surrounding normal kidney or lung (6). Subsequently, single-strand conformation polymorphism (SSCP) analysis revealed that a spectrum of TSC2 gene mutations exists in microdissected smooth muscle cells from sporadic pulmonary LAM (37). These studies support the hypothesis that LAM is caused by a somatic mutation in the TSC2 gene. However, mutations of the TSC2 gene were not detected in every patient. This may be due to limitations of SSCP in the detection of important mutations, contamination of the microdissected specimen with other pulmonary cells that may not express mutations of TSC2, or perhaps inconsistencies of mutations of the TSC2 gene in LAM cells and LAM patients. Nonetheless, despite these limitations, the available evidence is convincing for a strong association between somatic TSC2 gene mutations and sporadic LAM.The LAM Cell: Where Does It Come From?Studies supporting a causative role for somatic mutations of the TSC2 gene in sporadic LAM (6, 37) suggest that a normal resident pulmonary cell undergoes a spontaneous mutation resulting in aberrant proliferation locally. The question is which smooth muscle cell underwent such a mutation? In the lung, smooth muscle is predominantly found in the airway and vasculature with a small amount in lung interstitium. Based on the frequent observation of microscopic hemorrhage in LAM lesions, and the frequent involvement of blood vessels, it has been hypothesized that LAM cells may be derived from vascular smooth muscle. It has been shown that four distinct phenotypes of smooth muscle exist in the media of bovine aorta and that each phenotype is associated with a specific gene expression profile (40). Interestingly, in culture, each smooth muscle cell phenotype has different proliferative capacities (15). Similarly, in LAM, distinct smooth muscle cell phenotypes also occur that express markers indicating phenotype differences in growth potential. These parallel observations between vascular and LAM smooth muscle cell biology have led to the hypothesis that the LAM smooth muscle cell may be derived from vascular smooth muscle that differentiates into a unique migratory or proliferative phenotype by undergoing a spontaneous mutation within the vessel wall. However, no direct evidence supports this theory, leaving it yet to be proven.LAM is a multisystem disease that is not necessarily limited to lung involvement but can also involve lymph nodes, kidney, and colon. Recently, the observation that identical mutations are shared between the renal and pulmonary lesions in LAM have led some members of the LAM scientific community to hypothesize that the pulmonary LAM cell may have originally metastasized from a distant site (41). AMLs are highly vascular tumors that occur frequently in LAM (3, 24). They contain strong vascular and smooth muscle elements with histological appearances similar to pulmonary LAM and are also immunoreactive to HMB45 (7). AMLs that occur in sporadic pulmonary LAM share identical mutations of the TSC2 gene, indicating that they are genetically related. Karbowniczek et al. (23) have recently demonstrated five morphologically distinct vessel phenotypes in renal AML from patients with LAM, four of which were neoplastic in character, indicating a higher propensity to metastasize. In addition, smooth muscle cells are aberrantly found in the lung at autopsy in trauma patients (personal communication from L. S. Schuger), and on occasion, benign uterine leiomyomata can metastasize to the lung, resulting in a clinical syndrome similar to LAM (12). Thus it could be postulated that pulmonary LAM is a rare benign metastasizing disease.An alternative, but closely allied, explanation for the aberrant proliferation of immature smooth muscle cells in the lung is that LAM cells are donor cells from a nonhost source. Recent studies indicate that cell traffic occurs between the fetus and mother during pregnancy and that low numbers of cells or DNA persist in the maternal circulation for years thereafter (27). This process, known as microchimerism, is more common in diseases that have a female prevalence, such as autoimmune diseases, but can also be found in other conditions, such as in transplant or transfusion patients, where the source of nonhost cells is derived from the organ donor or blood (11). LAM has a predilection to the female sex, but the role of microchimerism is as yet unstudied in LAM, and the true source of the LAM cell remains elusive.The LAM Cell: How Does It Grow?Investigating LAM at a cellular level has been hampered by the lack of agreement on what exactly constitutes a LAM cell and the absence of an animal model of the disease. As previously discussed, the LAM cell is not a single phenotype, but many phenotypes, with cell-specific protein expression profiles and differential proliferative capacities. Thus applying the technique of microdissection to LAM tissue allows the limited evaluation of one cell type only. One group of microdissected cells may behave very differently from 1) microdissected cells from another LAM lesion within the same patient or 2) microdissected cells from another patient. Furthermore, contamination from nearby cells, influence of stage of disease, and hormonal therapy may also interfere with the genetic makeup and phenotype of microdissected pulmonary cells from the lung. This leads to a lack of standardization in the scientific approach to the cellular investigation of LAM.Nonetheless, one recent study clearly demonstrated activation of S6, a ribosomal protein intricately involved in regulating the cell cycle, in smooth muscle cells that were microdissected from LAM lung (19). These cells expressed an aberrant form of TSC2. Furthermore, reexpression of tuberin, the protein product of TSC2, in LAM-derived smooth muscle cells, led to reduced proliferation of LAM cells and reduced phosphorylation of S6. This is the first study to suggest that expression of mutant TSC2 in LAM smooth muscle cells leads to the aberrant cellular proliferation observed in LAM lung. Furthermore, the findings in this study parallel the observations of others investigating TSC2 biology who have noted that tuberin antagonizes the downstream pathways activated by the phosphorylation of tyrosine kinase receptors (16, 17, 33, 34). An explosion of research in the last five years has demonstrated that tuberin interacts with and binds to TOR (target of rapamycin), a cellular protein that is frequently activated in response to growth factor stimulation. Activation of mammalian TOR subsequently results in the downstream activation and phosphorylation of S6 (25). The study by Goncharova et al. (19) indicates that similar pathways are overactivated in LAM and suggests that studying the regulation of signaling pathways by tuberin provides useful insights into how LAM cells grow.Because of the paucity of LAM tissue and the absence of an ideal cell line in which to study signaling pathways in LAM, many investigators have relied on alternative cellular models. Such models include that described by Yu et al. (42). Renal AML smooth muscle cells bear remarkable morphological similarity to pulmonary LAM smooth muscle cells. Yu et al. describe the development of a cell line from an AML from a LAM patient that expressed inactivating mutations in both alleles of the TSC2 gene. Importantly, they demonstrate that similar to pulmonary LAM cells, the estrogen receptors (ER) α and ERβ are aberrantly expressed in LAM-derived renal AML cells. Furthermore, these cells grow in response to hormonal stimulation, and hormone-mediated growth of LAM-associated renal AML cell activates potent mediators of cell growth, i.e., ERK and c-myc. This study closely parallels findings by other groups that demonstrated a positive growth response to estrogen in smooth muscle cells that are null for TSC2 and TSC1 genes (14, 26). Yu et al. (42) add to our understanding of the proliferative effect of estrogen in cells that express dysfunctional TSC proteins that contrasts the inhibitory growth effects of estrogen frequently observed in vascular smooth cells that constitutively express tuberin (13, 14). Of even greater significance, Yu et al. also demonstrate that tamoxifen is equal to estrogen in its potency as an activator of growth of LAMderived AML cells. This observation supports the known fact that tamoxifen and, in some cases, progesterone, act as both estrogen agonists and antagonists and raises the question of whether anti-estrogen therapy is appropriate in LAM. Although estrogen has been implicated in the growth of LAM cells in vivo, it has not yet been demonstrated that pulmonary LAM-derived smooth muscle cells grow in culture in response to estrogen. However, if one extrapolates from the study of Yu et al. to the LAM patient, hormone-manipulating agents are potentially harmful and perhaps caution should be exercised when prescribing “anti-estrogen” therapy to patients who suffer from LAM.The aberrant expression of the ER and growth factors and their receptors in the lungs of LAM patients has received attention by researchers. Interestingly, immunohistochemical studies show that IGF binding proteins-2, -4, and -6 are associated with spindle-shaped LAM cells, whereas type 5 IGF binding protein is associated with epithelioid LAM cells (39). Similarly, FGF receptor-2 (FGFR-2; bek) is primarily expressed in LAM smooth muscle cells compared with FGFR-1 (Flg), which is expressed in epithelial cells adjacent to LAM smooth muscle cells (21). Matsui et al. (29) demonstrated that ER, localized in the nuclei of epithelioid (nonproliferating) LAM cells, is downregulated by hormonal therapy. In contrast, spindle-shaped (proliferating) LAM cells express little ER but highly express membrane type 1 matrix metalloproteinase (MT-1 MMP) (29). MT-1 MMP is a membrane-associated protein that activates MMP-2. MMP-2 is a potent matrix-degrading enzyme that is upregulated in LAM lung (20, 28). MMPs regulate matrix turnover, but it has also been shown that MMPs can induce cell growth by the activation of IGF binding proteins (35). These studies suggest that differential expression of growth factors and their receptors in LAM may result in cell-specific functions to exert fibrogenic, proliferative, and matrix-regulating effects in LAM. The immunohistochemical analysis of LAM tissue sheds new insights into the potential roles of ER, growth factors, and MMPs in LAM. However, it is limiting in terms of translating the role of such proteins into cellular function. The roles of ER, growth factors, their receptors, and MMPs in LAM-derived cells in culture has not been studied and would be extremely useful in clarifying the complex role of each individual growth factor in the pathogenesis of LAM.In summary, considerable progress has been made in our understanding of the pathogenesis of LAM. Although the jury is out on the true source of LAM cells, it is clear that they are a heterogeneous population of smooth muscle-like cells that variably exhibit immunoreactivity to HMB45. Evidence suggests that they possess specific functions regarding cell growth and matrix-degrading capacity and that the growth of at least a subpopulation of LAM cells is associated with the mutant expression of the TSC2 gene. Until a deeper understanding of the exact nature of LAM cells is achieved or until an animal model becomes available, scientists studying LAM will need to rely on immunohistochemical analysis of LAM tissue, the study of LAM-derived smooth muscle cells, renal AMLs, or other cells expressing aberrant forms of TSC2. However, it should be realized that limiting studies in LAM to specific genotypes may ultimately hamper the investigation of the pathogenesis of this disease. Studies that focus on TSC2 and TSC1 biology, other potential genetic mutations in LAM, the function of individual subpopulations of LAM cells, and factors that govern cell growth and matrix degradation all bear relevance to the pathogenesis of LAM. References 1 Astrinidis A, Khare L, Carsillo T, Smolarek T, Au KS, Northrup H, and Henske EP. Mutational analysis of the tuberous sclerosis gene TSC2 in patients with pulmonary lymphangioleiomyomatosis. J Med Genet 37: 55-57, 2000.Crossref | PubMed | ISI | Google Scholar3 Bernstein SM, Newell JD Jr, Adamczyk D, Mortenson RL, King TE Jr, and Lynch DA. How common are renal angiomyolipomas in patients with pulmonary lymphangiomyomatosis? Am J Respir Crit Care Med 152: 2138-2143, 1995.Crossref | PubMed | ISI | Google Scholar4 Bonetti F, Pea M, Martignoni G, Zamboni G, and Iuzzolino P. Cellular heterogeneity in lymphangiomyomatosis of the lung. Hum Pathol 22: 727-728, 1991.Crossref | PubMed | ISI | Google Scholar5 Carbonara C, Longa L, Grosso E, Borrone C, Garre MG, Brisigotti M, and Bigone M. 9q34 loss of heterozygosity in a tuberous sclerosis astrocytoma suggests a growth suppressor-like activity also for the TSC1 gene. Hum Mol Genet 3: 1829-1832, 1994.Crossref | PubMed | ISI | Google Scholar6 Carsillo T, Astrinidis A, and Henske EP. Mutations in the tuberous sclerosis complex gene TSC2 are a cause of sporadic pulmonary lymphangioleiomyomatosis. Proc Natl Acad Sci USA 97: 6085-6090, 2000.Crossref | PubMed | ISI | Google Scholar7 Chan JK, Tsang WY, Pau MY, Tang MC, Pang SW, and Fletcher CD. Lymphangiomyomatosis and angiomyolipoma: closely related entities characterized by hamartomatous proliferation of HMB-45-positive smooth muscle. Histopathology 22: 445-455, 1993.Crossref | PubMed | ISI | Google Scholar8 Cornog JL and Enterline HT. Lymphangiomyoma, a benign lesion of chyliferous lymphatics synonymous with lymphangiopericytoma. Cancer 19: 1909-1930, 1966.Crossref | PubMed | ISI | Google Scholar9 Corrin B, Liebow AA, and Friedman PJ. Pulmonary lymphangiomyomatosis. A review. Am J Pathol 79: 348-382, 1975.PubMed | ISI | Google Scholar10 Costello LC, Hartman TE, and Ryu JH. High frequency of pulmonary lymphangioleiomyomatosis in women with tuberous sclerosis complex. Mayo Clin Proc 75: 591-594, 2000.Crossref | PubMed | ISI | Google Scholar11 De Pauw L, Toungouz M, and Goldman M. Infusion of donor-derived hematopoietic stem cells in organ transplantation: clinical data. Transplantation 75: 46S-49S, 2003.Crossref | PubMed | ISI | Google Scholar12 Esteban JM, Allen WM, and Schaerf RH. Benign metastasizing leiomyoma of the uterus: histologic and immunohistochemical characterization of primary and metastatic lesions. Arch Pathol Lab Med 123: 960-962, 1999.PubMed | ISI | Google Scholar13 Farhat MY, Lavigne MC, and Ramwell PW. The vascular protective effects of estrogen. FASEB J 10: 615-624, 1996.Crossref | PubMed | ISI | Google Scholar14 Finlay GA, Hunter DS, Walker CL, Paulson KE, and Fanburg BL. Regulation of PDGF production and ERK activation by estrogen is associated with TSC2 gene expression. Am J Physiol Cell Physiol 285: C409-C418, 2003.Link | ISI | Google Scholar15 Frid M, Aldashev AA, Dempsey EC, and Stenmark KR. Smooth muscle cells isolated from discrete compartments of the mature vascular media exhibit unique phenotypes and distinct growth capabilities. Circ Res 81: 940-952, 1997.Crossref | PubMed | ISI | Google Scholar16 Gao X and Pan D. TSC1 and TSC2 tumor suppressors antagonize insulin signaling in cell growth. Genes Dev 15: 1383-1392, 2001.Crossref | PubMed | ISI | Google Scholar17 Gao X, Zhang Y, Arrazola P, Hino O, Kobayashi T, Yeung RS, Ru B, and Pan D. Tsc tumour suppressor proteins antagonize amino-acid-TOR signalling. Nat Cell Biol 4: 699-704, 2002.Crossref | PubMed | ISI | Google Scholar18 Gomez M, Sampson J, and Whittemore VH. Tuberous Sclerosis Complex. New York: Oxford Univ., 1999.Google Scholar19 Goncharova EA, Goncharov DA, Eszterhas A, Hunter DS, Glassberg MK, Yeung RS, Walker CL, Noonan D, Kwiatkowski DJ, Chou MM, Panettieri RA Jr, and Krymskaya VP. Tuberin regulates p70 S6 kinase activation and ribosomal protein S6 phosphorylation: a role for the TSC2 tumor suppressor gene in pulmonary lymphangioleiomyomatosis. J Biol Chem 3: 3, 2002.Google Scholar20 Hayashi T, Fleming MV, Stetler-Stevenson WG, Liotta LA, Moss J, Ferrans VJ, and Travis WD. Immunohistochemical study of matrix metalloproteinases and their tissue inhibitors in pulmonary lymphangioleiomyomatosis. Hum Pathol 28: 1071-1078, 1997.Crossref | PubMed | ISI | Google Scholar20a Hoon V, Thung SN, Kaneko M, and Unger PD. HMB reactivity in renal angiomyolipoma and lymphangioleiomyomatosis. Arch Path Lab Meth 118: 732-734, 1994.Google Scholar21 Inoue Y, King TE Jr, Barker E, Daniloff E, and Newman LS. Basic fibroblast growth factor and its receptors in idiopathic pulmonary fibrosis and lymphangioleiomyomatosis. Am J Respir Crit Care Med 166: 765-773, 2002.Crossref | PubMed | ISI | Google Scholar22 Kalassian KG, Doyle R, Kao P, Ruoss S, and Raffin TA. Lymphangioleiomyomatosis: new insights. Am J Respir Crit Care Med 155: 1183-1186, 1997.Crossref | PubMed | ISI | Google Scholar23 Karbowniczek M, Yu J, and Henske EP. Renal angiomyolipomas from patients with sporadic lymphangiomyomatosis contain both neoplastic and non-neoplastic vascular structures. Am J Pathol 162: 491-500, 2003.Crossref | PubMed | ISI | Google Scholar24 Kerr LA, Blute ML, Ryu JH, Swensen SJ, and Malek RS. Renal angiomyolipoma in association with pulmonary lymphangioleiomyomatosis: forme fruste of tuberous sclerosis? Urology 41: 440-444, 1993.Crossref | PubMed | ISI | Google Scholar25 Kwiatkowski DJ. Tuberous sclerosis: from tubers to mTOR. Ann Hum Genet 67: 87-96, 2003.Crossref | PubMed | ISI | Google Scholar26 Kwiatkowski DJ, Zhang H, Bandura JL, Heiberger KM, Glogauer M, el-Hashemite N, and Onda H. A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells. Hum Mol Genet 11: 525-534, 2002.Crossref | PubMed | ISI | Google Scholar27 Lambert N and Nelson JL. Microchimerism in autoimmune disease: more questions than answers? Autoimmun Rev 2: 133-139, 2003.Crossref | PubMed | ISI | Google Scholar28 Matsui K, Takeda K, Yu ZX, Travis WD, Moss J, and Ferrans VJ. Role for activation of matrix metalloproteinases in the pathogenesis of pulmonary lymphangioleiomyomatosis. Arch Pathol Lab Med 124: 267-275, 2000.PubMed | ISI | Google Scholar29 Matsui K, Takeda K, Yu ZX, Valencia J, Travis WD, Moss J, and Ferrans VJ. Downregulation of estrogen and progesterone receptors in the abnormal smooth muscle cells in pulmonary lymphangioleiomyomatosis following therapy. An immunohistochemical study. Am J Respir Crit Care Med 161: 1002-1009, 2000.Crossref | PubMed | ISI | Google Scholar30 Matsumoto Y, Horiba K, Usuki J, Chu SC, Ferrans VJ, and Moss J. Markers of cell proliferation and expression of melanosomal antigen in lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 21: 327-336, 1999.Crossref | PubMed | ISI | Google Scholar31 McCormack F, Brody A, Meyer C, Leonard J, Chuck G, Dabora S, Sethuraman G, Colby TV, Kwiatkowski DJ, and Franz DN. Pulmonary cysts consistent with lymphangioleiomyomatosis are common in women with tuberous sclerosis: genetic and radiographic analysis. Chest 121: 61S, 2002.ISI | Google Scholar32 Moss J, Avila NA, Barnes PM, Litzenberger RA, Bechtle J, Brooks PG, Hedin CJ, Hunsberger S, and Kristof AS. Prevalence and clinical characteristics of lymphangioleiomyomatosis in patients with tuberous sclerosis complex. Am J Respir Crit Care Med 164: 669-671, 2001.Crossref | PubMed | ISI | Google Scholar33 Potter CJ, Huang H, and Xu T. Drosophila Tsc1 functions with Tsc2 to antagonize insulin signaling in regulating cell growth, cell proliferation, and organ size. Cell 105: 357-368, 2001.Crossref | PubMed | ISI | Google Scholar34 Potter CJ, Pedraza LG, and Xu T. Akt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol 4: 658-665, 2002.Crossref | PubMed | ISI | Google Scholar35 Rajah R, Nunn SE, Herrick DJ, Grunstein MM, and Cohen P. Leukotriene D4 induces MMP-1, which functions as an IGFBP protease in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 271: L1014-L1022, 1996.Link | ISI | Google Scholar36 Smolarek TA, Wessner LL, McCormack FX, Mylet JC, Menon AG, and Henske EP. Evidence that lymphangiomyomatosis is caused by TSC2 mutations: chromosome 16p13 loss of heterozygosity in angiomyolipomas and lymph nodes from women with lymphangiomyomatosis. Am J Hum Genet 62: 810-815, 1998.Crossref | PubMed | ISI | Google Scholar37 Strizheva GD, Carsillo T, Kruger WD, Sullivan EJ, Ryu JH, and Henske EP. The spectrum of mutations in TSC1 and TSC2 in women with tuberous sclerosis and lymphangiomyomatosis. Am J Respir Crit Care Med 163: 253-258, 2001.Crossref | PubMed | ISI | Google Scholar38 Sullivan EJ. Lymphangioleiomyomatosis: a review. Chest 114: 1689-1703, 1998.Crossref | PubMed | ISI | Google Scholar39 Valencia JC, Matsui K, Bondy C, Zhou J, Rasmussen A, Cullen K, Yu ZX, Moss J, and Ferrans VJ. Distribution and mRNA expression of insulin-like growth factor system in pulmonary lymphangioleiomyomatosis. J Investig Med 49: 421-433, 2001.Crossref | PubMed | ISI | Google Scholar40 Weiser-Evans MC, Schwartz PE, Grieshaber NA, Quinn BE, Grieshaber SS, Belknap JK, Mourani PM, Majack RA, and Stenmark KR. Novel embryonic genes are preferentially expressed by autonomously replicating rat embryonic and neointimal smooth muscle cells. Circ Res 87: 608-615, 2000.Crossref | PubMed | ISI | Google Scholar41 Yu J, Astrinidis A, and Henske EP. Chromosome 16 loss of heterozygosity in tuberous sclerosis and sporadic lymphangiomyomatosis. Am J Respir Crit Care Med 164: 1537-1540, 2001.Crossref | PubMed | ISI | Google Scholar42 Yu J, Astrinidis A, Howard S, and Henske EP. Estradiol and tamoxifen stimulate LAM-associated angiomyolipoma cell growth and activate both genomic and nongenomic signaling pathways. Am J Physiol Lung Cell Mol Physiol 286: L694-L700, 2004.Link | ISI | Google ScholarAUTHOR NOTES Address for reprint requests and other correspondence: G. Finlay, Tufts-New England Medical Center, NEMC #257, 750 Washington St., Boston, MA 02111 (E-mail: [email protected]). Download PDF Previous Back to Top Next FiguresReferencesRelatedInformationCited BySporadic uterine Lymphangioleiomyomatosis (LAM): Report of a unique case arising in the lower uterine segment with short reviewGynecologic Oncology Reports, Vol. 37Mechanisms of Lung Cyst Formation29 January 2021Single-Cell Transcriptomic Analysis Identifies a Unique Pulmonary Lymphangioleiomyomatosis CellAmerican Journal of Respiratory and Critical Care Medicine, Vol. 202, No. 10Risk of breast cancer in patients with lymphangioleiomyomatosisCancer Epidemiology, Vol. 61Lymphangioleiomyomatosis9 June 2019Lung function response and side effects to rapamycin for lymphangioleiomyomatosis: a prospective national cohort study9 October 2017 | Thorax, Vol. 73, No. 4Lymphangioleiomyomatosis17 March 2018Human Pluripotent Stem Cell–Derived TSC2 -Haploinsufficient Smooth Muscle Cells Recapitulate Features of Lymphangioleiomyomatosis15 October 2017 | Cancer Research, Vol. 77, No. 20Metabolic Impact of Rapamycin (Sirolimus) and B-Estradiol Using Mouse Embryonic Fibroblasts as a Model for Lymphangioleiomyomatosis2 June 2017 | Lung, Vol. 195, No. 4Cathepsin K in LymphangioleiomyomatosisThe American Journal of Pathology, Vol. 187, No. 8Treatment of Lymphangioleiomyomatosis (LAM)2 February 2017Lymphangioléiomyomatose pulmonaire : de la physiopathologie à la prise en chargeRevue des Maladies Respiratoires, Vol. 33, No. 8Multiple cystic lung disease30 November 2015 | European Respiratory Review, Vol. 24, No. 138Malignant presentation of uterine lymphangioleiomyomatosisTaiwanese Journal of Obstetrics and Gynecology, Vol. 54, No. 5Lymphangioleiomyomatosis Biomarkers Linked to Lung Metastatic Potential and Cell Stemness13 July 2015 | PLOS ONE, Vol. 10, No. 7Wild Type Mesenchymal Cells Contribute to the Lung Pathology of Lymphangioleiomyomatosis15 May 2015 | PLOS ONE, Vol. 10, No. 5Analysis of gene expression array in TSC2-deficient AML cells reveals IRF7 as a pivotal factor in the Rheb/mTOR pathway4 December 2014 | Cell Death & Disease, Vol. 5, No. 12To Be or Not To Be a Neoplasm: What Is Lymphangioleiomyomatosis? Are We Calling It What It Really Is?American Journal of Respiratory and Critical Care Medicine, Vol. 188, No. 3MicroRNA-21 is Induced by Rapamycin in a Model of Tuberous Sclerosis (TSC) and Lymphangioleiomyomatosis (LAM)29 March 2013 | PLoS ONE, Vol. 8, No. 3Mesenchymal and miscellaneous neoplasmsLinfangioleiomiomatosis esporádica e hipertensión pulmonar. Estudio clínico-patológico en pacientes receptoras de un trasplante pulmonarMedicina Clínica, Vol. 138, No. 13LinfangioleiomiomatosisArchivos de Bronconeumología, Vol. 47, No. 2LymphangioleiomyomatosisArchivos de Bronconeumología ((English Edition)), Vol. 47, No. 2Linfangioleiomiomatosis: presentación de 6 casos y revisión de la literaturaRevista Española de Patología, Vol. 44, No. 1Entrained pulmonary clocks: epithelium and vasculature keeping paceEdwin C. Jesudason, Eli Keshet, and David Warburton1 October 2010 | American Journal of Physiology-Lung Cellular and Molecular Physiology, Vol. 299, No. 4Tuberin Regulates E-Cadherin LocalizationThe American Journal of Pathology, Vol. 177, No. 4Protein expression of urotensin II, urotensin-related peptide and their receptor in the lungs of patients with lymphangioleiomyomatosisPeptides, Vol. 31, No. 8Role of the CXCR4/CXCL12 Axis in Lymphangioleiomyomatosis and Angiomyolipoma28 June 2010 | The Journal of Immunology, Vol. 185, No. 3Lymphangioleiomyoma Cells and Lymphatic Endothelial CellsThe American Journal of Pathology, Vol. 176, No. 4Lung and PleuraVascular Endothelial Growth Factors C and D Induces Proliferation of Lymphangioleiomyomatosis Cells through Autocrine Crosstalk with EndotheliumThe American Journal of Pathology, Vol. 175, No. 4A 47-Year-Old Woman With Progressive Dyspnea and Recurrent PneumothoracesChest, Vol. 135, No. 5Lymphangioleiomyomatosis and Tuberous Sclerosis Complex12 April 2008 | Lung, Vol. 186, No. 4Lymphangioleiomyomatosis: A reviewEuropean Journal of Internal Medicine, Vol. 19, No. 5Activation of the Estrogen Receptor Contributes to the Progression of Pulmonary Lymphangioleiomyomatosis via Matrix Metalloproteinase-Induced Cell InvasivenessThe Journal of Clinical Endocrinology & Metabolism, Vol. 93, No. 5Predisposition to tetraploidy in pulmonary vascular smooth muscle cells derived from the Eker ratsYu Gui, Gordon H. He, Michael P. Walsh, and Xi-Long Zheng1 September 2007 | American Journal of Physiology-Lung Cellular and Molecular Physiology, Vol. 293, No. 3A Case of Pulmonary Lymphangioleiomyomatosis Developing 3 Years After Complete Surgical Removal of Abdominal LymphangioleiomyomaClinical Pulmonary Medicine, Vol. 14, No. 4Lymphangioléiomyomatose pulmonaire avec ou sans sclérose tubéreuse de BournevilleRevue des Maladies Respiratoires, Vol. 24, No. 6Molecular Pathogenesis of LymphangioleiomyomatosisAmerican Journal of Respiratory Cell and Molecular Biology, Vol. 36, No. 4Tuberin Nuclear Localization Can Be Regulated by Phosphorylation of Its Carboxyl Terminus17 November 2006 | Molecular Cancer Research, Vol. 4, No. 11Modulation of Cell Migration and Invasiveness by Tumor Suppressor TSC2 in LymphangioleiomyomatosisAmerican Journal of Respiratory Cell and Molecular Biology, Vol. 34, No. 4 More from this issue > Volume 286Issue 4April 2004Pages L690-L693 Copyright & PermissionsCopyright © 2004 the American Physiological Societyhttps://doi.org/10.1152/ajplung.00311.2003PubMed15003933History Published online 1 April 2004 Published in print 1 April 2004 Metrics

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call