1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62
The American Journal of Pathology, Vol. -, No. -, - 2016
ajp.amjpathol.org
Inactivation of Tsc2 in Mesoderm-Derived Cells Causes Polycystic Kidney Lesions and Impairs Lung Alveolarization Q34
Siying Ren,*y Yongfeng Luo,y Hui Chen,y David Warburton,yz Hilaire C. Lam,x Larry Wang,y Ping Chen,* Elizabeth P. Henske,x and Wei Shiyz From the Department of Respiratory Medicine,* The Second Xiangya Hospital, Central-South University, Changsha, People’s Republic of China; the Developmental Biology and Regenerative Medicine Program,y The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California; the Department of Surgery,z Keck School of Medicine, University of Southern California, Los Angeles, California; and the Division of Pulmonary and Critical Care Medicine,x Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts Accepted for publication August 29, 2016.
Address correspondence to Wei Shi, Children’s Hospital Los Angeles, 4650 Sunset Blvd, MS 35, Los Angeles, CA 90027; or Elizabeth P. Henske, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA Q4 Q5 02115. E-mail:
[email protected]. Q6 edu or
[email protected]. edu.
Q8
The tuberous sclerosis complex (TSC) proteins are critical negative regulators of the mTORC1 pathway. Germline mutations of TSC1 or TSC2 cause TSC, affecting multiple organs, including the kidney and lung, and causing substantial morbidity and mortality. The mechanisms of organ-specific disease in TSC remain incompletely understood, and the impact of TSC inactivation on mesenchymal lineage cells has not been specifically studied. We deleted Tsc2 specifically in mesoderm-derived mesenchymal cells of multiple organs in mice using the Dermo1-Cre driver. The Dermo1-Creedriven Tsc2 conditional knockout mice had body growth retardation and died approximately 3 weeks after birth. Significant phenotypes were observed in the postnatal kidney and lung. Inactivation of Tsc2 in kidney mesenchyme caused polycystic lesions starting from the second week of age, with increased cell proliferation, tubular epithelial hyperplasia, and epithelial-mesenchymal transition. In contrast, Tsc2 deletion in lung mesenchyme led to decreased cell proliferation, reduced postnatal alveolarization, and decreased differentiation with reduced numbers of alveolar myofibroblast and type II alveolar epithelial cells. Two major findings thus result from this model: inactivation of Tsc2 in mesoderm-derived cells causes increased cell proliferation in the kidneys but reduced proliferation in the lungs, and inactivation of Tsc2 in mesoderm-derived cells causes epithelial-lined renal cysts. Therefore, Tsc2-mTOR signaling in mesenchyme is essential for the maintenance of renal structure and for lung alveolarization. (Am J Pathol 2016, -: 1e12; http://dx.doi.org/10.1016/j.ajpath.2016.08.013)
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder resulting from germline loss-offunction mutations in the TSC1 or TSC2 gene.1,2 The TSC1/TSC2 protein complex has GTPase-activating protein activity toward Rheb, an upstream regulator of mTOR, and thus negatively regulates mTORC1 signaling activity by converting active Rheb-GTP to inactive Rheb-GDP. Therefore, abrogation of TSC1 or TSC2 causes dysregulation of mTORC1-dependent pathways, including protein translation, pyrimidine and purine biosynthesis, and autophagy. In addition to neurological disease, which includes seizures and autism, TSC patients have clinical manifestations in multiple organs, including the kidney, heart, lung,
and skin. In the kidney, a variety of lesions, such as angiomyolipoma, isolated or multiple renal cysts, and renal cell carcinoma, can occur in TSC patients. In the lung, TSC patients can develop multifocal micronodular pneumocyte hyperplasia and lymphangioleiomyomatosis (LAM).3,4 LAM is a progressive disease affecting almost exclusively
Supported by NIH grants HL068597 (W.S.), HL118760 (E.P.H.), and DK096556 (E.P.H.); a Tuberous Sclerosis Alliance Research grant (W.S.); the Hunan Province Health Department Research Fund B2014-020 (S.R.); the Hunan Province Natural Science Foundation of China 14JJ7012 (S.R.); and the Engles Program in TSC and LAM Research (E.P.H.). Disclosures: None declared.
Copyright ª 2016 Published by Elsevier Inc. on behalf of the American Society for Investigative Pathology. http://dx.doi.org/10.1016/j.ajpath.2016.08.013
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 Q7 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 Q2 Q3 123 124
125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186
Ren et al women and resulting in cystic lung degeneration. Up to 80% of women with TSC have multiple lung cysts by the age of 40 years, although only a minority will develop symptomatic LAM. Men with TSC can also develop cystic lung disease, although it is almost always asymptomatic. LAM also occurs in a sporadic form in women who do not have TSC. Sporadic LAM is associated with somatic inactivation of the TSC1 or TSC2 gene.5,6 The pathogenic mechanisms underlying the development of the clinical manifestations of TSC, in particular the pathogenesis of LAM, are not completely understood. One major challenge is the limitation of existing TSC disease models because conventional homozygous Tsc1 or Tsc2 knockout mice are embryonic lethal, and heterozygous Tsc1 or Tsc2 knockout mice develop epithelial renal lesions and tumors as their primary phenotypes.7e9 This is a striking contrast to human TSC, in which mesenchymal lesions of the kidney (angiomyolipomas) and lung (LAM) are major causes of morbidity and mortality. It is still unclear how the TSC protein complex functions in different cell compartments, such as epithelial cells versus mesenchymal cells, during organ development and during post-developmental tissue homeostasis. Although genetic deletions of Tsc1 or Tsc2 in distinct cell lineages, such as neurons, neural crest-derived cells, endothelia, cardiomyocytes, and differentiated smooth muscle cells, have been reported in mice,10e14 the consequences of Tsc1 or Tsc2 inactivation in pan-mesenchymal cells has remained unknown. We have investigated the specific function of Tsc2 in mesoderm-derived cells from the beginning of development in vivo. We achieved this by crossing a mesoderm-specific Cre-expressing mouse line Dermo1-Cre with floxed-Tsc2 mice to generate mesenchyme-specific Tsc2 conditional knockout mice.15,16 Although these Tsc2 conditional knockout mice were alive postnatally, postnatal growth retardation, polycystic kidney pathology, and defective lung alveolarization were detected during postnatal development. These mice died approximately 3 weeks after birth, consistent with an indispensible role of Tsc2 in mesenchymal cells in regulating both organ development and tissue homeostasis.
Materials and Methods Mouse Strains and Breeding Q9 Q10
Q11
Mesenchyme-specific Dermo1-Cre mice were provided by Dr. David Ornitz.15 Floxed-Tsc2 (Tsc2fx/fx) mice were originally generated in Dr. Michael Gambello’s laboratory.16 mT-mG double fluorescence reporter mice and Gt(ROSA)26Sortm1Sor LacZ reporter mice were obtained from Jackson Laboratory.17,18 All mice were bred in C57BL/6 stain background, and housed in pathogen-free conditions at the animal facility of Children’s Hospital Los Angeles. The procedures for mouse breeding and tissue harvest were approved by the Institutional Animal Care and
2
Use Committee at the Saban Research Institute of Children’s Hospital Los Angeles. Mouse genotypes were determined by genomic DNA PCR using tail biopsy samples. The genotyping PCR primers were described in previous publications.16,19
Histology and Morphometric Analysis Fresh tissues at different ages were harvested from euthanized mice, and examined under dissecting microscope. Samples were then either fixed in 4% buffered paraformaldehyde or frozen in liquid nitrogen. The fixed tissues were dehydrated and embedded in paraffin, and divided into sections (5 mm thick). The tissue sections were stained with hematoxylin and eosin or periodic acid-Schiff (Sigma Aldrich, St. Louis, MO). Lung elastin was stained using Q12 Hart’s resorcin-fuchsin method, as described in our previous publication.19 For lung morphometric analysis, five sections of each tissue were randomly chosen at approximately 200-mm intervals and stained with hematoxylin and eosin. The mean linear intercept was used to measure average size of alveoli, as described previously.19 Results were analyzed with unpaired t-tests to compare the differences between mean values, and considered significant if P < 0.05. To avoid the sex differences in organ maturation, this quantitative comparison was also performed among fetuses with different sex at each time point.
Immunostaining and Detection Tissue sections were deparaffinized and hydrated, and antigen recovery was performed by boiling tissue slides in Tris-EDTA buffer (pH 9.0) for 30 minutes. The tissue sections were blocked in 5% donkey normal serum and 0.5% Triton X-100 for 1 hour, and then incubated with the following primary antibodies overnight at 4 C: mouse antiegreen fluorescent protein (GTX628528; GeneTex); Q13 goat antieplatelet endothelial cell adhesion molecule 1 (sc-1506), goat anti-CC10 (sc-9772), and goat antieSp-C (sc-7706; Santa Cruz Biotechnology, Dallas, TX); rabbit Q14 antiered fluorescent protein for Tomato protein (600-401379; Rockland); rat anti-Cdh1 (13-1900; ThermoFisher), Q15 Q16 mouse antieb-tubulin IV (MU178-UC; BioGenex); fluo- Q17 rescein-Lotus tetragonobolus lectin and rhodamineDolichos biflorus agglutinin (FL-1321 and RL-1032; Vector Laboratories); mouse anti-cytokeratin and mouse antiea- Q18 smooth muscle actin (C2562 and A2547; Sigma Aldrich); rabbit antiephospho-S6 (Ser235/236) and rabbit antivimentin (4858 and 5741; Cell Signaling Technology); Q19 and hamster anti-T1a (DSHB at the University of Iowa, Iowa City). Alexa Fluro 488e, Alexa Fluro 594e, or Alexa Fluro 647econjugated donkey secondary antibodies (Molecular Probes) were used, and cell nuclei were coun- Q20 terstained by DAPI in the mounting medium (Vector Laboratories). Fluorescence images were taken using the Zeiss LSM710 confocal microscope at the Imaging Core Q21
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248
249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310
Tsc2 in Kidney and Lung Lesions Q1 Facility of Children’s Hospital Los Angeles. Active caspase 3 staining was performed by immunohistochemistry using rabbit anti-cleaved caspase 3 antibody (9661; Cell Signaling Technology) and second anti-rabbit antibodyehorseradish peroxidase conjugate, followed by color development with 3,30 -diaminobenzidine and hematoxylin counterstaining.
Western Blot
Q22
Q23
Detection of proteins in tissue lysate by Western blot has been previously described with the following minor modification.20 Equal amounts (50 mg) of total tissue lysate proteins were separated in mini tris-glycine extended precast gels (4% to 15% gradient; Bio-Rad) and transferred into polyvinylidene difluoride membrane using Bio-Rad’s Trans-Blot Turbo Transfer System. Proteins of interest were detected using the following specific antibodies: rabbit antiTsc2 (GTX61245; GeneTex); rabbit antiephospho-S6 (Ser235/236) and mouse anti-S6 (4858 and 2317; Cell Signaling Technology); and rabbit antieb-actin (G046; Applied Biological Materials).
Cell Proliferation
Q24 Q25
Q26
To detect proliferating cells in vivo, cell nuclear 50 -ethynyl20 -deoxyuridine (EdU) labeling was used.21 Briefly, mice were i.p. injected with EdU (5 mg/kg body weight; Life Technologies) 3 hours before harvesting tissue samples. EdU is a thymidine analogue that is incorporated into newly synthesized chromosome DNA. Incorporated EdU was detected using Alexa Fluor azide (Life Technologies), and cell nuclei were counterstained with DAPI. The image analysis was performed in four random fields per slide from a total of four slides per mouse by an experienced observer blind to the mouse genotype. The ratios of EdUlabeled nuclei/total DAPI-stained nuclei were used to evaluate cell proliferation, and the experiments were repeated in more than three mice within each genotype group.
Serum Cystatin C Measurement
Q27
Blood was collected from five Tsc2 conditional knockout (CKO) and wild-type (WT) littermate control pairs around postnatal day 20 (P20). The sera were diluted 200-fold, and used for cystatin C measurement using a commerical enzyme-linked immunosorbent assay kit (Thermo). Paired sample t-test was used for the analysis.
Statistical Analysis All experiments were repeated at least three times. The quantitative data were presented as means SD. Statistical analyses were performed using t-tests, with P 0.05 considered significant.
The American Journal of Pathology
-
311 312 313 Conditional Deletion of Tsc2 in Mesoderm-Derived Cells 314 Results in Postnatal Growth Retardation and Lethality 315 316 It has been reported that Dermo1 promoter-driven Cre 317 expression is specific for certain lineages of mesoderm318 derived cells in a variety of organs in the Dermo1-Cre 319 knockin fetal mice,15 and we verified this Cre expression 320 pattern in E14.5 Dermo1-Cre fetuses using Gt(ROSA) Q28 321 26Sortm1Sor LacZ reporter mice (Figure 1A). We crossed ½F1 322 Dermo1-Cre with floxed-Tsc2 mice (Tsc2fx/fx) to specifically 323 delete Tsc2 gene in Dermo1-expressing lineages of mesen324 325 chymal cells within multiple organs/systems during devel326 opment. The mice were born with the expected mendelian 327 ratio of genotypes and sexes. All control pups had normal 328 development and weight gain with normal survival through 329 fx/fx 12 months of age. Dermo1-Cre/Tsc2 conditional 330 knockout (abbreviated as Tsc2 CKO) mice were born with 331 notably smaller body size and lower body weight than their 332 littermate controls. These differences became more obvious 333 during postnatal growth (Figure 1, B and C), suggesting 334 significant postnatal growth retardation in Tsc2 CKO mice. 335 Heterozygous Tsc2 CKO (Dermo1-Cre/Tsc2fx/þ) mice had 336 337 body weight and size comparable to their WT littermates, 338 and did not develop noticeable phenotypic abnormalities up 339 to 5 months of age (data not shown). 340 The homozygous Tsc2 CKO mice died approximately 3 341 weeks after birth, with median survival of P20 (Figure 1D). 342 No sex difference was observed for Tsc2 CKO mouse sur343 vival. Unlike the phenotypic changes seen in liver, cardiac, 344 and central neuronal systems of germline Tsc2 mutant ani345 mals,8,22,23 there were no histologically obvious abnormal346 ities in brain, liver, or heart in the Tsc2 CKO mice at P14 347 (Figure 2). In contrast, the kidney and lung were clearly ½F2 348 349 abnormal at this age (see Results below for detail). The 350 potential causes for the lethality in Tsc2 CKO mice are 351 further analyzed and discussed below. 352 353 Mesodermal Tsc2 Abrogation Results in Severe 354 355 Polycystic Kidney Lesions 356 357 Dermo1-Creedriven gene deletion, shown by green fluores358 cent protein expression in a mT-mG reporter line,17 occurred in 359 renal mesenchymal cells of both the cortical and medullary re360 gions, but not in tubular epithelial cells or endothelial cells, 361 shown by Cdh1þ (E-cadherinþ) and platelet endothelial cell 362 þ adhesion molecule 1 staining, respectively (Figure 3, A and ½F3 363 B). Reduction of Tsc2 protein and activation of the mTORC1 364 pathway (increased phosphorylation of ribosomal protein S6) 365 were verified by Western blotting of the entire Tsc2 CKO kid366 ney tissue lysate (Figure 3C). Examination of the moribund 367 Tsc2 CKO mice sacrificed at approximately 3 weeks after birth 368 369 revealed obvious changes in kidney size and appearance, which 370 looked pale, polycystic, and enlarged by gross view 371 (Figure 3D). We then analyzed the dynamic changes of kidney 372
Results
ajp.amjpathol.org
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
3
morphology in the Tsc2 CKO mice. Interestingly, no significant alterations of kidney structure and size were observed in neonatal Tsc2 CKO mice at P1 (Figure 4, A and C). Microscopic Q29½F4 cystic pathology was observed in the renal cortex from the second week after birth (P14) (Figure 4). The cystic lesions rapidly progressed and expanded into the medulla, destroying the normal renal cortex-medulla structural relationships at 3 weeks of age in all Tsc2 CKO mice (Figure 4). Glycogen accumulation was detected in the cysts by periodic acid-Schiff staining. In addition, multiple foci of tubular epithelial hyperplasia and adenomatous changes adjacent to the cysts were seen in Tsc2 CKO kidney after 2 weeks of age (Figure 4). Consistently, overall cell proliferation in Tsc2 CKO kidneys was doubled compared to that in WT littermates, shown by nucleotide analogue EdU labeling (9.0 2.4 in Tsc2 CKO versus 4.1 1.1 in WT) (Figure 5, A and B) at P7 before obvious ½F5 morphological changes occurred. The cystic changes were further analyzed by staining the tissue with L. tetragonobolus lectin (a proximal tubular marker) and D. biflorus agglutinin (a collecting duct marker). Interestingly, some of the lining epithelial cells within the cystic structures were positive for D. biflorus agglutinin (Figure 5C), but not for L. tetragonobolus lectin, which suggests that the cystic pathology originates in the distal collecting ducts. The renal endothelial networks were comparable between Tsc2 CKO and WT controls (Figure 5C). Some of the tubular epithelial cells were positively stained with vimentin in the Tsc2 CKO kidney at P21, with significant reduction of Cdh1 expression (Figure 5D). Increased vimentin protein level in the Tsc2 CKO kidney was verified by Western blot (Figure 5E), suggesting an epithelial-mesenchymal transition as a consequence of Tsc2 inactivation. In addition, increased apoptotic cells, detected by active caspase 3 immunostaining, were seen in P21 Tsc2 CKO kidney sections (Figure 5, F and G). These apoptotic cells in the cysts appeared to be shed into the lumens. Interestingly, only minor increase of serum cystatin C was detected in the live Tsc2 CKO mice around the age of P20 (10 3% greater than the littermate WT control; *P < 0.05, n Z 5 pairs), suggesting that kidney function is not severely impaired.
Mesodermal Tsc2 Deletion Causes Retardation of Pulmonary Alveolar Development
print & web 4C/FPO
373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434
Ren et al
Using mT-mG fluorescence reporter, we confirmed that most lung mesenchymal cells, except pulmonary endothelial cells, were targeted by the Dermo1-Cre driver during
Figure 1
Deletion of Tsc2 in mesoderm-derived cells resulted in reduced body growth and lethality at early life. A: LacZ staining (blue) of E14.5 Dermo1-Cre/Gt(ROSA)26Sortm1Sor reporter mice to visualize the pattern of Cre expression. B: Comparison of body sizes between Tsc2fx/fx/Dermo1-Cre [Tsc2 conditional knockout (CKO)] mice and their wild-type (WT) littermate controls. C: Comparison of whole body weights between Tsc2 CKO and their littermate controls at different ages. D: Altered survival was detected in Tsc2 CKO mice compared to the WT controls. n Z 4 per genotype (C); n Z 13 (4 females and 9 males; D). *P < 0.05. P, postnatal day.
4
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
Q30
Q31
435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496
559 labeling (6.7 1.2% in Tsc2 CKOs versus 11.0 1.8% in WT controls) (Figure 7, A and B). Differentiation analysis ½F7 560 561 using immunostaining revealed that lung alveolar myofi562 broblasts in the Tsc2 CKO mice were markedly reduced, as 563 shown by a-smooth muscle actinepositive cells in alveolar 564 septa (Figure 7C), whereas smooth muscle cells of both 565 airways and vasculatures appeared similar between Tsc2 566 CKO and the controls. Interestingly, type II alveolar 567 epithelial cells, shown by Sp-C staining, were also signifi568 569 cantly decreased, whereas type I alveolar epithelial cells, 570 shown by T1a staining, were not changed (Figure 7C). The 571 proximal airway epithelial cells, including ciliated cells and 572 Club cells, shown by b-tubulin IV and CC10 immuno573 staining, respectively, were comparable between Tsc2 CKO 574 and WT mice. The number and distribution of alveolar 575 capillary endothelial cells, detected by platelet endothelial 576 cell adhesion molecule 1 staining, appeared to be normal 577 (Figure 7C). Finally, elastin fiber deposition in the tips of 578 alveolar septa and alveolar walls was attenuated in the Tsc2 579 CKO lung during alveolar stage (P14) (Figure 7, D and E). 580 581 Therefore, reduced lung alveolarization in lung 582 mesenchyme-specific Tsc2 knockout mice is likely because 583 of both altered cell proliferation and myofibroblast differ584 entiation in the peripheral lung.
print & web 4C=FPO
497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558
Tsc2 in Kidney and Lung Lesions
Figure 2
Histological comparison of brain (A), liver (B), and heart (C) between Tsc2fx/fx/Dermo1-Cre female mice and wild-type (WT) female control mice at postnatal day 14 is shown by hematoxylin and eosinestained tissue sections. Scale bars: 100 mm (A and B); 1 mm (C). CKO, conditional knockout.
½F6
development, whereas no epithelial cells were Cre positive (Figure 6, A and B). The deletion of Tsc2 in the lung tissue of Dermo1-Cre/Tsc2fx/fx mice was then validated by reduced Tsc2 protein expression and increased mTOR pathway activity (phosphorylation of S6) using Western blot and immunostaining (Figure 6, C and D). Lung development was examined by comparing morphological structures between Tsc2 CKO mice and their WT littermate controls. Although prenatal development of the lung in Tsc2 CKO mice was not significantly affected, shown by their lung morphology at P1, significant deficiency of lung alveolarization, which normally occurs from P5 to P30 in mice,24 was detected (Figure 6E). In the Tsc2 CKO mouse lung, growth of alveolar septa was markedly decreased, which resulted in larger alveolar spaces, within the lungs from P7 to P21. This altered histology was then verified by morphometric measurement of average alveolar size (mean linear intercept) (Figure 6F). Therefore, lung mesenchymal Tsc2 has an important functional role in promoting lung alveolarization during postnatal development. To understand the potential mechanisms by which mesenchymal Tsc2 deletion inhibited lung alveolarization, we analyzed cell proliferation and differentiation. Interestingly, a significant reduction of cell proliferation was detected in P7 Tsc2 CKO mouse lungs using nuclear EdU
The American Journal of Pathology
-
Discussion TSC is a disease affecting multiple organs and systems. Germline homozygous deletion of Tsc2 in mice results in embryonic lethality, and Tsc2 heterozygous deletion results in liver, kidney, and lung tumors as the primary phenotypes in adult mice.7e9 The manifestations of human TSC include mesenchymal-lineage tumors of the kidney (angiomyolipomas) and lung (lymphangioleiomyomatosis or LAM). To elucidate the consequences of TSC2 inactivation in multiple mesenchymal lineages, we used the Dermo1-promoter-Cre that drives Cre expression in mesoderm-derived cells of multiple organs.15 Herein, we report the novel findings that pan-mesenchymal Dermo1-Creedriven homozygous Tsc2 CKO mice are viable but have growth retardation, postnatal progressive polycystic kidney disease, and defective alveolarization of the lung. Mesenchymal cells are heterogeneous, including a variety of smooth muscle cells, endothelial cells, fibroblasts, osteoblasts, and chondrocytes. The specific mesenchymal cell types and mechanisms underlying the reduced body size and weight in our Tsc2 conditional knockout mice are not known. Increased craniofacial bone mass in neural crestespecific Tsc1 knockout mice and increased postnatal skeletal bone acquisition in osteoblast-specific Tsc2 knockout mice have been reported previously.25,26 However, retardation of body growth (size or weight) was not found in those prior models. The reduced body size in our mesenchymal knockout model may be the result of aberrant bone development because of loss of Tsc2 function in both
ajp.amjpathol.org
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
5
585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620
Figure 3 Dermo1-Cre driven Tsc2 deletion in mouse kidney. A: Dermo1-Cre targeted cells in the kidney were characterized in mT-mG fluorescence protein reporter mice at postnatal day 14 (P14), in which membrane green fluorescent protein (GFP) expression (green) replaced membrane Tomato expression (red) only in cells in which Cremediated loxP DNA recombination occurred. B: Dermo1-Creemediated loxP DNA recombination in mT-mG reporter mice (green) was not detected in P14 kidney epithelial cells (Cdh1þ) or in endothelial cells [platelet endothelial cell adhesion molecule (PECAM)þ]. C: Alterations of Tsc2 protein and phosphorylation of S6, a downstream target of mTOR pathway, were detected in P7 Tsc2 conditional knockout (CKO) kidneys by Western blot. D: Gross view of kidneys isolated from Tsc2 CKO mice and wild-type (WT) control mice at P25. Scale bars: 50 mm (A and B); 1 mm (D).
print & web 4C=FPO
621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682
Ren et al
chrondrocytes and osteoblasts, and/or the consequence of impaired renal or respiratory function. In our Dermo1-Cre mice, Cre is expressed in condensed mesenchyme from which both chondrocytes and osteoblasts are derived during embryonic endochondral ossification (E11.5).15 At later stages, except in bone marrow cells and osteoclasts, Cre expression in Dermo1-Cre mice is also detected in chondrocytes of growth plate cartilage and osteoblasts of the perichondrium, periosteum, and endosteum.15 A future detailed analysis of Tsc2 in these targeted cells and skeletal development will be needed to understand the relevant mechanisms underlying the phenotype of retarded body growth. Previous studies found that deletion of Tsc1 in smooth muscle cells and cardiomyocytes using an SM22-driven Cre resulted in severe biventricular hypertrophy starting from embryonic day 15, with median survival of these Tsc1 conditional knockout mice of approximately 24 days after birth.12,13 Interestingly, modest cystadenoma pathology was also detected in the kidney and thought to be caused by minor leakage of Cre expression into renal epithelial cells. In the lung, no significant changes in alveolarization were reported; instead, significant muscularization of small peripheral pulmonary vessels was noted. In our Dermo1-Creedriven Tsc2
6
knockout mice, no morphological change in heart development was detected (Figure 2), which may be because of lack of cardiomyocyte targeting in our Dermo1-Cre driver line (Figure 1). However, in the kidney, our Tsc2 CKO mice developed severe polycystic pathology within 3 weeks after birth, resulting in destruction of normal kidney structure. In addition, tubular epithelial hyperplasia and epithelialmesenchymal transition, but no angiomyolipoma-like lesions, appeared by 3 weeks of age. Using the fluorescence reporter to track the Dermo1-Cre targeted cells, we found that Cre was expressed specifically in kidney mesenchymal cells, and not in glomerular endothelial cells nor tubular epithelial cells (Figure 3). In addition, overall cell proliferation was increased in the Tsc2 CKO kidney, consistent with epithelial hyperplasia. The potential mechanisms through which mesenchymal Tsc2 deletion results in tubular epithelial hyperplasia and cystic change may include a physical blockage of flow by these hyperplastic tubular structures. These findings suggest that Tsc2 functions within renal mesenchymal cells to maintain normal renal tubular structural homeostasis after development rather than regulating kidney morphogenesis per se. Kidney lesions occur in most TSC patients and are a common cause of morbidity. These include both
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744
Figure 4 Renal pathology in the Tsc2 conditional knockout (CKO) mice. A: Comparison of overall kidney anatomical structures between the Tsc2 CKO mice and their littermate controls at different postnatal ages. B: Hematoxylin and eosinestained kidney tissue sections at high magnification. Black dotted line highlights a hyperplastic tubule. C: Kidney cortex histology shown by periodic acid-Schiff staining. Arrow indicates multiple layers of hyperplastic tubular epithelia lining a cystic structure; arrowhead, a single layer of epithelium lining a cystic structure. Scale bars: 1 mm (A); 50 mm (B); 100 mm (C). P, postnatal day.
print & web 4C=FPO
745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806
Tsc2 in Kidney and Lung Lesions
mesenchymal and epithelial lesions: mesenchymal angiomyolipomas and epithelial polycystic kidney disease, oncocytoma, and renal cell carcinoma.27,28 The mechanisms of renal cystic disease in TSC, including whether they arise because of epithelial versus mesenchymal inactivation of TSC2, are not yet completely understood. Several findings have focused on an epithelial pathogenesis model, including that TSC2 is required for the membrane
The American Journal of Pathology
-
localization of polycystin 1 protein and that TSC2 physically interacts with polycystin 1 to regulate the primary cilium in epithelia.29,30 Our data suggest, for the first time, that some of the epithelial cystic lesions in TSC may arise through mesenchymal loss of TSC2 function, supporting a new model for the cyst pathogenesis in TSC. A contiguous gene syndrome, with severe early-onset cystic disease, occurs in human with germline deletions of both TSC2 and
ajp.amjpathol.org
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
7
807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868
print & web 4C=FPO
869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930
Ren et al
Figure 5 Cell proliferation and cystic epithelial cells in Tsc2 conditional knockout (CKO) kidney. A and B: Proliferative cells were detected by 3-hour 50 -ethynyl20 -deoxyuridine (EdU) labeling, and the percentage of EdU-positive nuclei (green) was analyzed based on DAPI-nuclear counterstaining (blue). C: Endothelial cells in the renal cortex were detected by platelet endothelial cell adhesion molecule (PECAM) 1 immunostaining. Different tubular epithelial cells were also labeled with Lotus tetragonobolus lectin (LTL; green) or Dolichos biflorus agglutinin (DBA; red)-specific binding. DBA-positive cells in the cystic epithelia are indicated with an arrow. D: Coimmunostaining of Cdh1 (green), vimentin (red), and DBA (gray) for postnatal day 21 (P21) kidneys with indicated Tsc2 genotypes. Vimentin-positive cells in tubules are indicated with arrowheads. E: Increased vimentin in total kidney tissue lysate at P21 was detected by Western blot. b-Actin is a loading control. F: Activated caspase 3 immunostaining for apoptotic cells (brown, shown with arrows) in P21 kidneys. G: The numbers of apoptotic cells per 100 mm2 area of kidney tissue sections are compared. n Z 3 in each genotype (-). *P < 0.05. Scale bar Z 50 mm (-). WT, wild type.
8
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
Q32 Q33
931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992
Figure 6 Lung mesenchyme-specific knockout of Tsc2 inhibited postnatal alveolarization. A and B: Lung cells targeted by the Dermo1-Cre driver at postnatal day 7 (P7) were analyzed in mT-mG fluorescence reporter mice. C and D: Altered Tsc2 protein expression and mTOR activation in P7 Tsc2 conditional knockout (CKO) lungs were analyzed by Western blot and immunostaining. E: Lung histological changes were evaluated by comparing hematoxylin and eosinestained lung tissue structures between Tsc2 CKO and wild-type (WT) control at different postnatal ages. F: Alveolar sizes were quantitatively analyzed by measuring mean linear intercept. n Z 4 per genotype (F). *P < 0.05. Scale bars: 50 mm (AeD); 200 mm (E). a, airway; GFP, green fluorescent protein; PECAM, platelet endothelial cell adhesion molecule; v, vasculature.
print & web 4C=FPO
993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054
Tsc2 in Kidney and Lung Lesions
the adjacent polycystic kidney disease 1 (PKD1) gene.31 More important, specific deletion of Tsc2 exon 2 to 4 in our model confirms that polycystic renal pathology can be caused by mesenchymal loss of Tsc2,16 without simultaneous deletion of the adjacent Pkd1 gene,32 which experimentally supports clinical observations that renal cysts can
The American Journal of Pathology
-
occur in patients with TSC1 or TSC2 mutation but no PKD1 deletion.33 In our Tsc2 CKO mouse lung, a striking reduction of pulmonary alveolarization was observed, without obvious changes in pulmonary vascular and airway smooth muscle cells. Yet, by examining the alveolar septa structure,
ajp.amjpathol.org
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
9
1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116
print & web 4C=FPO
1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178
Ren et al
Figure 7
Altered lung proliferation and differentiation in Tsc2 conditional knockout (CKO) mice. A and B: Proliferative cells were detected by 3-hour 50 -ethynyl-20 -deoxyuridine (EdU) labeling (green) in postnatal day 7 (P7) mice, and quantified based on cell nuclear counterstaining with DAPI (blue). C: Lung myofibroblasts/smooth muscle cells, endothelial cells, type II and type I alveolar epithelial cells, airway club cells, and ciliated cells were immunostained with the related protein markers. D: Comparison of elastin fibers (black) in lung tissues between Tsc2 CKO and wild-type (WT) control mice at P7 and P14. E: Semiquantification of elastin deposition in P14 lung tissues, measured by the percentage of stained elastin area over total lung tissue area of four tissue sections. *P < 0.05. Scale bars Z 50 mm (AeD). a, airway; PECAM, platelet endothelial cell adhesion molecule; SMA, a-smooth muscle actin; v, vasculature.
we found that a-smooth muscle actinepositive myofibroblasts in our Tsc2 CKO lung were significantly reduced, particularly in the tips of the alveolar septa. In addition, overall lung cell proliferation was significantly decreased in the Tsc2 CKO lung, which is opposite to the
10
increased cell proliferation detected in the Tsc2 CKO kidney. Therefore, it appears that loss-of-function mutation in Tsc2 and subsequent mTORC1 hyperactivation result in strikingly diverse cellular responses depending on cell lineages, organs, and stages of development and
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240
1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302
Tsc2 in Kidney and Lung Lesions homeostasis. Lung alveolarization occurs postnatally in mice, with the growth of secondary alveolar septa into primary alveolar sacs. Alveolar myofibroblasts play a critical role in driving alveolar septa formation by proliferating and migrating into the tips of the septa and by producing elastin extracellular matrices.34 Defects in alveolar myofibroblast migration to the tip because of abrogation of platelet-derived growth factor signaling results in arrest of pulmonary alveolarization.35 In our Tsc2 CKO lung, the overall number of alveolar myofibroblasts was significantly reduced, accompanied by decreased elastin deposition within the septa. Therefore, reduced proliferation, differentiation, and function of alveolar myofibroblasts because of loss of Tsc2 function and the resulting hyperactivation of mTORC1 pathway is one of the pivotal mechanisms underlying the alveolar hypoplastic lung phenotype. In contrast, alveolar capillaries were not significantly altered based on immunostaining, suggesting that alveolar angiogenesis, another driving force for pulmonary alveolarization, may not be a critical Tsc2-dependent mechanism for alveolarization. Whether defective alveolarization in these Tsc2 CKO mice contributes to pulmonary cystic lesions in adult remains unknown, because of their shortened life span. It has been reported that decreased alveolar growth in early life has a negative impact on alveolar homeostasis in an aging population, associated with alveolar destruction and emphysema.36 Therefore, it is possible that reduced alveolarization during lung development may contribute to the pulmonary cystic lesions observed in TSC patients. More important, no LAM-like nodules were detected in the female or male Tsc2 CKO mice, which died before adulthood. We therefore speculate that the high incidence pulmonary cysts in women with TSC, estimated at 80% by the age of 40 years,37 may reflect both a developmental alveolar defect involving TSC1/2 inactivation in the lung mesenchyme that is not sex specific, and sexspecific LAM celleproduced proteases and cytokines that mediate tissue destruction. This model could explain the fact that most TSC patients (both male and female) with lung cysts will not progress to symptomatic pulmonary disease. We further speculate that the mesenchymal impact of TSC2 loss cooperates with LAM celleproduced proteases to enhance the lung destruction. To address these hypotheses, further investigation of the relationship between lung mesenchymal Tsc2 inactivation and LAM-like pathology is needed.
Acknowledgments We thank Dr. David M. Ornitz for providing Dermo1-Cre mice; Dr. Michael Gambello for providing floxed-Tsc2 mice; Dr. Esteban Fernandez (Cell Imaging Core of Children’s Hospital Los Angeles) for helping with confocal imaging; Dr. Kevin Lemley for initial discussion about
The American Journal of Pathology
-
mouse kidney phenotype; and Dr. Roderick Bronson (Harvard Medical School) for review of the renal pathology.
References 1. Curatolo P, Maria BL: Tuberous sclerosis. Handb Clin Neurol 2013, 111:323e331 2. Henske EP, Józwiak S, Kingswood JC, Sampson JR, Thiele EA: Tuberous sclerosis complex. Nat Rev Dis Primers 2016, 2:16035 3. Hayashi T, Kumasaka T, Mitani K, Yao T, Suda K, Seyama K: Loss of heterozygosity on tuberous sclerosis complex genes in multifocal micronodular pneumocyte hyperplasia. Mod Pathol 2010, 23: 1251e1260 4. Carsillo T, Astrinidis A, Henske EP: Mutations in the tuberous sclerosis complex gene TSC2 are a cause of sporadic pulmonary lymphangioleiomyomatosis. Proc Natl Acad Sci U S A 2000, 97: 6085e6090 5. Henske EP, McCormack FX: Lymphangioleiomyomatosis: a wolf in sheep’s clothing. J Clin Invest 2012, 122:3807e3816 6. Harari S, Torre O, Moss J: Lymphangioleiomyomatosis: what do we know and what are we looking for? Eur Respir Rev 2011, 20:34e44 7. Kwiatkowski DJ, Zhang H, Bandura JL, Heiberger KM, Glogauer M, elHashemite N, 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 2002, 11:525e534 8. Kobayashi T, Minowa O, Kuno J, Mitani H, Hino O, Noda T: Renal carcinogenesis, hepatic hemangiomatosis, and embryonic lethality caused by a germ-line Tsc2 mutation in mice. Cancer Res 1999, 59:1206e1211 9. Onda H, Lueck A, Marks PW, Warren HB, Kwiatkowski DJ: Tsc2(þ/-) mice develop tumors in multiple sites that express gelsolin and are influenced by genetic background. J Clin Invest 1999, 104:687e695 10. Crowell B, Hwa Lee G, Nikolaeva I, Dal Pozzo V, D’Arcangelo G: Complex neurological phenotype in mutant mice lacking Tsc2 in excitatory neurons of the developing forebrain(123). eNeuro 2015, 2: ENEURO.0046-15.2015 11. Ma A, Wang L, Gao Y, Chang Z, Peng H, Zeng N, Gui YS, Tian X, Li X, Cai B, Zhang H, Xu KF: Tsc1 deficiency-mediated mTOR hyperactivation in vascular endothelial cells causes angiogenesis defects and embryonic lethality. Hum Mol Genet 2014, 23:693e705 12. Kayyali US, Larsen CG, Bashiruddin S, Lewandowski SL, Trivedi CM, Warburton RR, Parkhitko AA, Morrison TA, Henske EP, Chekaluk Y, Kwiatkowski DJ, Finlay GA: Targeted deletion of Tsc1 causes fatal cardiomyocyte hyperplasia independently of afterload. Cardiovasc Pathol 2015, 24:80e93 13. Malhowski AJ, Hira H, Bashiruddin S, Warburton R, Goto J, Robert B, Kwiatkowski DJ, Finlay GA: Smooth muscle protein-22-mediated deletion of Tsc1 results in cardiac hypertrophy that is mTORC1-mediated and reversed by rapamycin. Hum Mol Genet 2011, 20:1290e1305 14. Kwiatkowski DJ: Animal models of lymphangioleiomyomatosis (LAM) and tuberous sclerosis complex (TSC). Lymphat Res Biol 2010, 8:51e57 15. Yu K, Xu J, Liu Z, Sosic D, Shao J, Olson EN, Towler DA, Ornitz DM: Conditional inactivation of FGF receptor 2 reveals an essential role for FGF signaling in the regulation of osteoblast function and bone growth. Development 2003, 130:3063e3074 16. Hernandez O, Way S, McKenna J, Gambello MJ: Generation of a conditional disruption of the Tsc2 gene. Genesis 2007, 45:101e106 17. Muzumdar MD, Tasic B, Miyamichi K, Li L, Luo L: A global doublefluorescent Cre reporter mouse. Genesis 2007, 45:593e605 18. Soriano P: Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat Genet 1999, 21:70e71 19. Chen H, Zhuang F, Liu YH, Xu B, Del Moral P, Deng W, Chai Y, Kolb M, Gauldie J, Warburton D, Moses HL, Shi W: TGF-beta receptor II in epithelia versus mesenchyme plays distinct roles in the developing lung. Eur Respir J 2008, 32:285e295
ajp.amjpathol.org
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
11
1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364
1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399
Ren et al 20. Sun J, Zhuang FF, Mullersman JE, Chen H, Robertson EJ, Warburton D, Liu YH, Shi W: BMP4 activation and secretion are negatively regulated by an intracellular gremlin-BMP4 interaction. J Biol Chem 2006, 281:29349e29356 21. Luo Y, El Agha E, Turcatel G, Chen H, Chiu J, Warburton D, Bellusci S, Qian BP, Menke DB, Shi W: Mesenchymal adenomatous polyposis coli plays critical and diverse roles in regulating lung development. BMC Biol 2015, 13:42 22. Pajak L, Jin F, Xiao GH, Soonpaa MH, Field LJ, Yeung RS: Sustained cardiomyocyte DNA synthesis in whole embryo cultures lacking the TSC2 gene product. Am J Physiol 1997, 273:H1619eH1627 23. Rennebeck G, Kleymenova EV, Anderson R, Yeung RS, Artzt K, Walker CL: Loss of function of the tuberous sclerosis 2 tumor suppressor gene results in embryonic lethality characterized by disrupted neuroepithelial growth and development. Proc Natl Acad Sci U S A 1998, 95:15629e15634 24. Warburton D, Bellusci S, Del Moral PM, Kaartinen V, Lee M, Tefft D, Shi W: Growth factor signaling in lung morphogenetic centers: automaticity, stereotypy and symmetry. Respir Res 2003, 4:5 25. Fang F, Sun S, Wang L, Guan JL, Giovannini M, Zhu Y, Liu F: Neural crest-specific TSC1 deletion in mice leads to sclerotic craniofacial bone lesion. J Bone Miner Res 2015, 30:1195e1205 26. Riddle RC, Frey JL, Tomlinson RE, Ferron M, Li Y, DiGirolamo DJ, Faugere MC, Hussain MA, Karsenty G, Clemens TL: Tsc2 is a molecular checkpoint controlling osteoblast development and glucose homeostasis. Mol Cell Biol 2014, 34:1850e1862 27. Henske EP: Tuberous sclerosis and the kidney: from mesenchyme to epithelium, and beyond. Pediatr Nephrol 2005, 20:854e857 28. Yang P, Cornejo KM, Sadow PM, Cheng L, Wang M, Xiao Y, Jiang Z, Oliva E, Jozwiak S, Nussbaum RL, Feldman AS, Paul E, Thiele EA, Yu JJ, Henske EP, Kwiatkowski DJ, Young RH, Wu CL: Renal cell carcinoma in tuberous sclerosis complex. Am J Surg Pathol 2014, 38:895e909 29. Kleymenova E, Ibraghimov-Beskrovnaya O, Kugoh H, Everitt J, Xu H, Kiguchi K, Landes G, Harris P, Walker C: Tuberin-dependent
12
30.
31.
32.
33.
34. 35.
36. 37.
membrane localization of polycystin-1: a functional link between polycystic kidney disease and the TSC2 tumor suppressor gene. Mol Cell 2001, 7:823e832 Shillingford JM, Murcia NS, Larson CH, Low SH, Hedgepeth R, Brown N, Flask CA, Novick AC, Goldfarb DA, Kramer-Zucker A, Walz G, Piontek KB, Germino GG, Weimbs T: The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease. Proc Natl Acad Sci U S A 2006, 103: 5466e5471 Brook-Carter PT, Peral B, Ward CJ, Thompson P, Hughes J, Maheshwar MM, Nellist M, Gamble V, Harris PC, Sampson JR: Deletion of the TSC2 and PKD1 genes associated with severe infantile polycystic kidney disease: a contiguous gene syndrome. Nat Genet 1994, 8:328e332 Piontek K, Menezes LF, Garcia-Gonzalez MA, Huso DL, Germino GG: A critical developmental switch defines the kinetics of kidney cyst formation after loss of Pkd1. Nat Med 2007, 13: 1490e1495 Sampson JR, Maheshwar MM, Aspinwall R, Thompson P, Cheadle JP, Ravine D, Roy S, Haan E, Bernstein J, Harris PC: Renal cystic disease in tuberous sclerosis: role of the polycystic kidney disease 1 gene. Am J Hum Genet 1997, 61:843e851 Morrisey EE, Hogan BL: Preparing for the first breath: genetic and cellular mechanisms in lung development. Dev Cell 2010, 18:8e23 Bostrom H, Willetts K, Pekny M, Leveen P, Lindahl P, Hedstrand H, Pekna M, Hellstrom M, Gebre-Medhin S, Schalling M, Nilsson M, Kurland S, Tornell J, Heath JK, Betsholtz C: PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell 1996, 85:863e873 Fletcher C, Peto R: The natural history of chronic airflow obstruction. Br Med J 1977, 1:1645e1648 Cudzilo CJ, Szczesniak RD, Brody AS, Rattan MS, Krueger DA, Bissler JJ, Franz DN, McCormack FX, Young LR: Lymphangioleiomyomatosis screening in women with tuberous sclerosis. Chest 2013, 144:578e585
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.4.0 DTD AJPA2462_proof 18 October 2016 7:52 pm EO: AJP16_0197
1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434