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. -, - 2019
ajp.amjpathol.org
Q1
Q37
Host Lymphotoxin-b Receptor Signaling Is Crucial for Angiogenesis of Metanephric Tissue Transplanted into Lymphoid Sites Maria Giovanna Francipane,*y Bing Han,* and Eric Lagasse* From the McGowan Institute for Regenerative Medicine and Department of Pathology,* University of Pittsburgh, Pittsburgh, Pennsylvania; and the Ri.MED Foundation,y Palermo, Italy Accepted for publication August 20, 2019. Address correspondence to Maria Giovanna Francipane, Ph.D., 450 Technology Dr., Ste. 333, Pittsburgh, PA 15219; or Eric Lagasse, Pharm.D., Ph.D., 450 Technology Dr., Ste. 300, Pittsburgh, PA 15219. E-mail:
[email protected] or lagasse@ pitt.edu.
Q6
The mouse lymph node (LN) can provide a niche to grow metanephric kidney to maturity. Here, we show that signaling through the lymphotoxin-b receptor (LTbR) is critical for kidney organogenesis both in the LN and the omentum. By transplanting kidney rudiments either in the LNs of mice undergoing LTbR antagonist treatment or in the omenta of LTBR knockout (LTbR/) mice, the host LTbR signals were found to be crucial for obtaining a well-vascularized kidney graft. Indeed, defective LTbR signaling correlated with decreased expression of endothelial and angiogenic markers in kidney grafts as well as structural alterations. Because the number of glomerular endothelial cells expressing the LTbR target nuclear factor kBeinducing kinase (NIK) decreased in the absence of a functional LTbR, it was speculated that an LTbR/NIK axis mediated the angiogenetic signals required for successful ectopic kidney organogenesis, given the established role of NIK in neovascularization. However, the transplantation of kidney rudiments in omenta of NIK/ mice revealed that NIK is dispensable for ectopic kidney vascular integration and maturation. Finally, defective LTbR signaling impaired compensatory glomerular adaptation to renal mass reduction, indicating that kidney regeneration approaches, besides whole kidney reconstruction, might benefit from the presence of LTbR signals. (Am J Pathol 2019, -: 1e18; https://doi.org/10.1016/j.ajpath.2019.08.018)
Chronic and end-stage kidney disease impose a large burden on human health worldwide. Projected prevalence trend lines are not comforting,1 suggesting an urgent need to explore new approaches to overcome limitations associated with traditional interventions. Dialysisdthe most common method used to treat advanced kidney failuredcan only replace 10% to 20% of kidney function and is associated with increased left ventricular mass, intramyocardial cell fibrosis, and capillary loss.2,3 Orthotopic kidney transplantation offers better quality of life and longer survivals,4 yet it is limited by organ shortage and the risks associated with immunosuppression, including increased vulnerability to infection and cancer, as well as organ toxicity.5 One novel solution to the shortage of human organs available for transplantation envisions growing new organs in situ via transplantation of developing anlagen from animal embryos.6 A few sites have been considered for renal anlage transplantation, and, paradoxically, the orthotopic
environment seems not to be the most appropriate. Indeed, transplantation of metanephric tissue into the adult mouse kidney does not lead to glomerular or tubular differentiation.7 Other sites have also been considered, including the anterior eye chamber,8 the omentum,9 the paraaortic area,10 and, recently, the lymph node (LN) has been identified as an attractive site for the maturation of mouse metanephroi into functional glomeruli and tubules.11,12 Here, we show that host lymphotoxin-b receptor (LTbR) pathway generates angiogenic cues to support kidney organogenesis in both LNs and omenta. Our hypothesis that
Supported by the Ri.MED Foundation (M.G.F.); NIH grant R01 DK085711 (E.L.); and The Pediatric Device InitiativeeMcGowan Institute for Regenerative Medicine, University of Pittsburgh e Commonwealth of Pennsylvania grant SAP4100073573 (E.L., M.G.F.). Disclosures: E.L. is the scientific founder and the Chief Scientific Officer of LyGenesis, Inc.
Copyright ª 2019 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. https://doi.org/10.1016/j.ajpath.2019.08.018
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
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 Q5 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 Q2 Q3 122 Q4 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
Francipane et al
Q7
LTbR could affect kidney organogenesis in these sites sprang from the central role of LTbR in the development13,14 and homeostasis15 of lymphoid organs, as well as from our finding that glomeruli maturing in both LNs and omenta contained, during the early stages of engraftment, endothelial cells expressing the nuclear factor kB-inducing kinase (NIK), a downstream target, among others, of LTbR.16 Given an established role for NIK in neovascularization,17 and the presence of host endothelial cells in ectopic kidney grafts, it was speculated that a host LTbR/ NIK axis could allow successful kidney organogenesis. In accordance, defective LTbR signaling was found to be associated with decreased numbers of glomerular NIK-expressing endothelial cells, decreased absolute vascularization/angiogenesis, and structural alterations of the kidney grafts. However, kidney organogenesis was not affected in the absence of NIK. This result might suggest either the existence of different targets downstream of LTbR, leading to angiogenesis, or the activation of compensatory angiogenic pathways in the absence of NIK. Finally, the absence of LTbR signals impaired compensatory glomerular adaptation to renal mass reduction, indicating that kidney regeneration approaches, besides whole kidney reconstruction, might benefit from the presence of LTbR signals.
mice (n Z 10 per group) or in the omenta of wild-type or NIK/ C57BL/6 mice (n Z 5 per group). Kidneys were split longitudinally in two pieces before transplantation. The kidney preparation was placed onto the exposed omentum, and the omentum was later folded onto itself. Growth Factor Reduced Matrigel (356231; Corning, Corning, NY) was used to glue the transplanted omenta. Intraomental kidney grafts from wild-type and LTbR/ mice were collected at different time points (2 weeks: n Z 1; 3 weeks: n Z 4; 4 weeks: n Z 1; 5 weeks: n Z 1; 6 weeks: n Z 3), whereas intraomental kidney grafts from wild-type and NIK/ mice were all collected 3 weeks after transplantation. Wild-type or LTbR/ C57BL/6 mice were also transplanted with GFPþ newborn (postnatal day 2) kidneys in their omenta (n Z 3 per group). Half of a kidney was transplanted in each mouse as described in the paragraph above. One mouse from each group was sacrificed every week, starting at 2 weeks after transplantation up to the fourth week. Images of kidney grafts were obtained with both the Olympus IX71 inverted fluorescence microscope (Tokyo, Japan) and the Leica EZ4D stereomicroscope (Wetzlar, Germany).
Materials and Methods
To assess LTbR pathway contribution to kidney organogenesis inside the LN, recipient mice were treated intraperitoneally with 100 mg of mouse LTbR-Fc fusion protein (71122; BPS Bioscience, San Diego, CA) or control immunoglobulin (PI31205; Pierce Biotechnology, Rockford, IL) 2 days before transplantation. Mice were further treated at day 8 and 15, and LNs were collected at day 22. LTbR-Fc concentration and exposure times were chosen based on previously published studies.19,20 LTbR blockage was confirmed by decreased glomerular NIK expression.
Ethics Statement
Q8 Q9
Mice were purchased from Charles River Laboratories (Wilmington, MA) or The Jackson Laboratory (Bar Harbor, ME), and bred and housed in the Division of Laboratory Animal Resources facility at the University of Pittsburgh. Experimental protocols followed NIH’s Guide for the Care and Use of Laboratory Animals,18 and they were approved by the University of Pittsburgh Institutional Animal Care and Use Committee. Mixed sexes were used.
Tissues and Transplantations E14 kidneys were retrieved from timed-pregnant green fluorescent protein (GFP)þ or wild-type C57BL/6 mice under a dissecting microscope. GFPþ kidneys served for transplantation of wild-type C57BL/6 LNs to investigate host-derived vascularization (CD31 stain) and expression of LTbR, erasmus university rotterdamdthymic reticulum antibody 7 (ER-TR7), LT-b, and NIK (n Z 6) or to test the effect of LTbR-Fc treatment (n Z 6) (see below). Wild-type kidneys served for transplantation of wild-type C57BL/6 LNs (n Z 3) to analyze glomerular NIK expression. LN transplantation was performed as previously described,11 and LN kidney grafts were collected 3 or 6 weeks after transplantation, depending on the experiment. Alternatively, paired GFPþ E14 kidneys were transplanted in the omenta of wild-type or LTbR/ C57BL/6
2
LTbR-Fc Treatments
Stains and Quantifications Engrafted LNs and omenta and mouse kidneys were fixed in 4% paraformaldehyde, embedded in optimal cutting temperature compound or paraffin, and cut into 4e to 7em-thick sections for further analysis. Hematoxylin and eosin (H&E) staining was performed as described elsewhere.21 All other stains were performed as follows. Fluorescent Immunohistochemistry Staining of Frozen Tissue Sections Frozen sections were equilibrated at room temperature for 15 minutes, then rehydrated in phosphate-buffered saline (PBS) for 15 minutes, and permeabilized with cold 0.1% Triton X-100 for 10 minutes. After three washes in PBS supplemented with 0.05% Tween 20 (PBS-T), a solution of 3% bovine serum albumin was applied to the sections for 30 minutes to prevent nonspecific binding. Sections were then incubated with specific primary
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
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
Growing Kidneys in Ectopic Sites antibodies at least 1 hour at room temperature or overnight at 4 C. After three washes in PBS-T, sections were incubated with Alexa Fluor 488 or 594 secondary antibodies (Thermo Fisher, Pittsburgh, PA) for 20 minutes. Sections were washed three times, and the nuclei were counterstained with Hoechst 33342 (Life Technologies, Frederick, MD). A solution of 50% glycerol in Hanks’ balanced salt solution was used as mounting medium. Fluorescent Immunocytochemistry Staining CD45-depleted stromal cells from omenta were cytospun onto glass slides (see below for isolation), fixed with acetone for 8 minutes, and stained with specific antibodies according to the same protocol described for frozen sections. Chromogenic Immunohistochemistry Staining of ParaffinEmbedded Tissue Sections Paraffin sections were deparaffinized in xylene and rehydrated in graded alcohol. Endogenous peroxidases were blocked with the use of 3% hydrogen peroxide for 10 minutes. Antigen unmasking was performed with 10 mmol/L sodium citrate buffer pH 6.0. Nonspecific binding was prevented by incubating the slides in 3% bovine serum albumin in PBS-T for 30 minutes. The slides were exposed to primary antibodies overnight at 4 C, incubated with biotinylated secondary antibody, and later with streptavidinhorseradish peroxidase (R.T.U. Vectastain Kit; Vector, Burlingame, CA). 3-Amino-9-ethylcarbazole (HK092-5KE; BioGenex, Fremont, CA) was used as a chromogen. Immunostained sections were counterstained with Mayer’s hematoxylin (Dako, Carpinteria, CA) and mounted in permount mounting medium (SP15-500; Thermo Fisher). A list of antibodies used is provided in Table 1. Isotype½T1 matched antibodies were used as negative controls. Images were obtained with the Olympus IX71 inverted fluorescence microscope.
Q10
Fluorescent Quantifications Areas of LN and intraomental kidney grafts were quantified by measuring pixel counts of GFP signal with the use of Photoshop CS5 (Adobe, San Jose, CA). The mean fluorescence intensity (MFI) of podoplanin (PDPLN) and the fluorescence intensities of CD31, CD105, and Ly-76 were measured among regions of interest drawn around the Bowman capsule with the use of ImageJ version 1.52a (NIH, Bethesda, MD; https://imagej.nih.gov/ij), as previously described.22 Glomerular areas were quantified at the time of PDPLN MFI measurement. The corrected total glomerular fluorescence for CD31, CD105, and Ly-76 was later calculated with the following formula: Integrated density e (area of selected glomerulus mean fluorescence of background readings). Length of lotus tetragonolobus lectin (LTL)þ tubules was obtained with ImageJ version 1.52a by drawing a line over the LTLþ stain (circular tubular structures were not
The American Journal of Pathology
-
Table 1
List of Reagents Used in This Study
Antibody
Catalog number
Supplier
CD105 CD31 CD34 CDH1 CIV EFNB2 ER-TR7 Ki-67 LT-b LTbR LTL Ly-76 NIK PAI-1 PDPLN
550546 ab28364 MA1-82073 sc-7870 1340-01 ab131536 ab51824 ab15580 ab64835 ab70063 B-1325 ab91113 ab216409 ab66705 NBP1-90211
BD Biosciences (San Jose, CA) Abcam (Cambridge, MA) ThermoFisher (Pittsburgh, PA) Santa Cruz Biotechnology (Dallas, TX) SouthernBiotech (Birmingham, AL) Abcam (Cambridge, MA) Abcam (Cambridge, MA) Abcam (Cambridge, MA) Abcam (Cambridge, MA) Abcam (Cambridge, MA) Vector Laboratories (Burlingame, CA) Abcam (Cambridge, MA) Abcam (Cambridge, MA) Abcam (Cambridge, MA) Novus Biologicals (Centennial, CO)
CDH1, cadherin 1; CIV, collagen IV; EFNB2, ephrin B2; ER-TR7, erasmus university rotterdamethymic reticulum antibody 7; LT-b, lymphotoxin-b; LTbR, lymphotoxin-b receptor; LTL, lotus tetragonolobus lectin; NIK, nuclear factor kBeinducing kinase; PAI-1, plasminogen activator inhibitor 1; PDPLN, podoplanin.
considered) and later converting the obtained numbers into microns. Glomeruli and tubules from at least five sections per sample were considered. ER-TR7 MFI was measured in 6 sections per sample with the use of ImageJ version 1.52a. The number of NIKþ cells in LN and intraomental grafts was obtained by analyzing glomeruli from 7 to 13 microscopic fields depending on the experiment. The number of plasminogen activator inhibitor 1 (PAI-1)þ tubules in LNs and intraomental grafts was obtained by analyzing 12 to 20 microscopic fields. Histologic Quantifications Quantitative measurement of glomerular diameters was performed by taking the maximum diameter from the hilum to the end of the Bowman capsule of 18 H&E-stained glomeruli derived from three sections per mouse, using the measurement tool in cellSens Standard 1.9 software (Olympus). Ki67þ nuclei were counted in at least five random sites within the cortex across two separate stained sections per mouse. The number of Ki-67þ nuclei was finally expressed as a percentage of the total number of nuclei. All sections were randomly selected and analyzed under Q11 blinded conditions.
Stromal Cell Isolation and Flow Cytometry Stromal cells were isolated from GFPþ C57BL/6 mouse mesenteric LNs (n Z 10) or omenta (n Z 10) according to the method described by Turley laboratory.23 Briefly, tissues were collected and digested with an enzyme mix comprised of RPMI 1640 medium that contained 0.8 mg/mL dispase (17105-041; Gibco, Gaithersburg, MD), 0.2 mg/mL collagenase P (11213857001; Sigma-Aldrich, St. Louis, MO),
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
3
311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372
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
Francipane et al
Q12
and 1 mg/mL DNase I (89836; Thermo Scientific, Pittsburgh, PA). After digestion, anti-CD45 immunomagnetic beads (130-052-301; Miltenyi Biotec, Auburn, CA) were used to deplete hematopoietic cells from the bulk population. CD45-depleted stromal cells were then stained with Sytox Blue (S11348; Molecular Probes, Eugene, OR), phycoerythrin-cyanine 7 CD45 (552848; BD Biosciences, Bedford, MA), phycoerythrin Podoplanin (127407; BioLegend, San Diego, CA), and peridinin chlorophyll protein/ cyanine 5.5 CD31 (102419; BioLegend). Flow cytometric analysis was performed with a BD FACSAria cytometer and FlowJo software version 9.6 (TreeStar, Ashland, OR), according to standard procedures. Isotype antibodies were used as negative controls. Table 2
Molecular Analysis RNA isolation was performed with the RNeasy Mini Kit (74106; Qiagen, Germantown, MD) according to the manufacturer’s instructions (including the optional oncolumn DNase digestion). RNA purity and concentration were measured with a NanoDrop 2000/c spectrophotometer (Thermo Fisher Scientific, Pittsburgh, PA). cDNAs were synthesized with the RT2 First Strand Kit (330401; Qiagen), and expression of 84 genes modulating the biological processes of angiogenesis were measured with the Mouse Angiogenesis RT2 Profiler PCR Array (PAMM-024Z; Qiagen) on a StepOnePlus Real-Time PCR System (Applied Biosystems, Foster City, CA),
List of Primer Sequences Used in This Study NCBI reference
Gene ID
Sequence
ANGPT1
F: 50 -AGTGCGACAGAGCAGTACAA-30 R: 50 -TGGGCCATCTCCGACTTCAT-30 F: 50 -TGTACCGTGGAGAAGCCTGT-30 R: 50 -CCTCACGGAACTTGGGATTTGA-30 F: 50 -CAAATGGGTCTTTGGAGGGC-30 R: 50 -ACGTCTTGTTGGACCGTGAT-30 F: 50 -GCCCATTACCCTGCTGTTTG-30 R: 50 -CACAGCCGACTCTTTCTGC-30 F: 50 -AGTCTTCTGCTCGCCCTGCTC-30 R: 50 -CGGACTCCACCACAACAGTTCC-30 F: 50 -AACCTGTGGCTGTGGAACAAC-30 R: 50 -GGGAAGTGATGTCCAGGAAGT-30 F: 50 -GTGGAAGTGTCCTCCCTTGAG-30 R: 50 -GGAGCCTTCCGTTCTTAGGGT-30 F: 50 -CAGGTCCTAGCTGGGTTGG-30 R: 50 -CGTTGAAAGGCACCACTTCC-30 F: 50 -CGGACACATGCAGTCTCTGT-30 R: 50 -CCTTCTCCAATACAGCGTCCA-30 F: 50 -CAACCCGACAGAGACAATCCT-30 R: 50 -TGAAGGCGTCTCTTCCCACT-30 F: 50 -CCAAGGCAGGCAGACGAAT-30 R: 50 -TAGTGTACTCGGCCAGGTGT-30 F: 50 -CCGGCTTAGTTCTCTGTGGA-30 R: 50 -GCAATGCAGGTGAGGGATGT-30 F: 50 -CTCTGGGTACGTGGGTGTTC-30 R: 50 -CACCACTCACAGTGTTTGCG-30 F: 50 -GATCACCACAACCCACACCT-30 R: 50 -CCAGGTTGCGGAAGCAGTAA-30 F: 50 -TTGGCGACTGGAGCAATACA-30 R: 50 -GAGACGGAACCTACCACAGC-30 F: 50 -GATTGAGACCCTGGTGGACATC-30 R: 50 -CATCTCTCCTATGTGCTGGCT-30 F: 50 -TGGCTGGGTGTGGTATGAAC-30 R: 50 -GGTGACCTCCCTCATGTTCC-30 F: 50 -TGCTAAGTCTGGCACGAAGG-30 R: 50 -ACACCAAACTGCCCGATCAT-30
EFNA1 EFNB2 ENG JAG1 NOS3 PECAM1 PGF PTK2 SERPINE1 TBX1 TEK TGFA TGFB3 TIE1 VEGFA GUSB HSP90AB1
NM_009640.4 NM_010107.4 NM_010111.5 NM_007932.2 NM_013822.5 NM_008713.4 NM_008816.3 NM_008827.3 NM_007982.2 NM_008871.2 NM_011532.2 NM_013690.3 NM_031199.4 NM_009368.3 NM_011587.2 NM_001287056.1 NM_010368.2 NM_008302.3
Primer sequences were designed with National Center for Biotechnology Information’s (NCBI) Primer-BLAST (Basic Local Alignment Search Tool) (https:// www.ncbi.nlm.nih.gov/tools/primer-blast, last accessed April 27, 2018). The NCBI Reference sequences that were entered in the Primer-BLAST submission form are indicated for each gene and were retrieved from NCBI’s Nucleotide database (https://www.ncbi.nlm.nih.gov/refseq). F, forward; R, reverse.
4
ajp.amjpathol.org
Q34
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
Q35
Q36
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
according to the manufacturer’s instructions. The CT values from all plates and wells were then exported from the Real-Time PCR instrument and formatted into a new Excel spreadsheet (Microsoft, Redman, WA) (Supplemental Table S1). The data were uploaded in the RT2 Profiler PCR Array Data Analysis Webportal (https:// dataanalysis.qiagen.com/pcr/arrayanalysis.php, last accessed April 4, 2018) and analyzed. GUSB and HSP90AB1 were automatically selected as optimal housekeeping genes. Genes with fold changes >2 and P values < 0.05 were later validated with Real-Time RTPCR. Primers were designed with NCBI’s Primer-Blast (https://www.ncbi.nlm.nih.gov/tools/primer-blast, last accessed April 27, 2018). Primers bridged an exoneexon junction or spanned introns. Free online Beacon Designer software (http://www.premierbiosoft. com/qOligo/Oligo.jsp?PIDZ1, last accessed April 27, 2018) was used to evaluate possible secondary structures such as dimers or hairpins. To check the
Q13
Q14
Q15
559 primer specificity, before performing Real-Time 560 RT-PCR, conventional PCR was performed, and ampli561 cons were separated on agarose gels. A melting curve 562 analysis was performed after each Real-Time RT-PCR to 563 further confirm that each assay produced a single, specific 564 ½T2 565 product. Primers sequences are given in Table 2.
Unilateral Nx Wild-type (n Z 3) and LTbR/ (n Z 4) C57BL/6 mice underwent unilateral nephrectomy (Nx). After exposing the left kidney, the renal artery, vein, and ureter were tied off. The kidney was finally excised distal to the ligatures.
Statistical Analysis Statistical analysis was performed with the unpaired t-test (*P < 0.05, **P < 0.01, ***P < 0.001,
Figure 1 Vascularization of glomeruli engrafted in the mouse lymph node (LN) might be driven by host lymphotoxin-b receptor (LTbR)/nuclear factor kB-inducing kinase (NIK) axis. A: Left column: Frozen sections of a representative green fluorescent protein (GFP)þ metanephric (embryonic day 14) kidney graft 3 weeks after transplantation. Merged GFP signal with Hoechst stain (blue; top panel), merged GFP signal with CD31 (endothelial cell) stain (red; middle panel), and merged CD31 stain (red) with Hoechst stain (blue; bottom panel). Boxed and circled areas are shown at higher magnification to the right and demonstrate the presence of a host (GFP) CD31þ cell in an engrafted glomerulus. Right panel: Cartoon depicting the localization of glomerular endothelial cells (image adapted from http://whatwhen-how.com/acp-medicine/glomerular-diseasespart-1). B: Representative immunofluorescence stain for CD34 (endothelial cell marker, red) and NIK (green) in an engrafted wild-type (GFP) glomerulus 3 weeks after transplantation. Arrows point to double positive cells. C: Representative immunofluorescence stain for LTbR (red), erasmus university rotterdamdthymic reticulum antibody 7 (ER-TR7; LN fibroblastic reticular cells, red), or lymphotoxin b (LT-b, red) in engrafted GFPþ glomeruli 3 weeks after transplantation. Data presented in this figure are representative of one technical replicate of a minimum of three technical replicates per sample. n Z 3. Scale bars: 50 mm (A); 25 mm (B and C).
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
Growing Kidneys in Ectopic Sites
The American Journal of Pathology
-
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
5
Q20
Q21
Q22
566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620
****P < 0.0001). Statistical power analysis was also performed to assess error rates for tubule length measurements. For all studies, the power of a two-tailed t-test was >0.9, given an a level of 0.05 (5%). A volcano plot of RT2 Profiler PCR Array data was used to identify genes differentially expressed in kidneys engrafted in LTbR/ versus wild-type omenta. Correlation between RT2 Profiler PCR Array and Real-Time RT-PCR was calculated by linear regression and Pearson correlation. Both SABiosciences’ data analysis (Frederick, MD) and GraphPad Prism 7 software (GraphPad Inc., San Diego, CA) were used.
Results Vascularization of Glomeruli Engrafted in the Mouse LN Might Be Driven by Host LTbR/NIK Axis With the use of GFP-expressing mouse metanephric kidneys, successful nephron maturation inside a host mouse LN has been demonstrated.11,12 The LN-engrafted nephrons could rely on a remarkable vascular network originating from both host- and graft-derived vascular cells.11,12 Even more impressive were the data of human fetal kidney engraftment inside the LN, which clearly indicated that,
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
Francipane et al
Figure 2
Treatment with lymphotoxin-b receptor antagonist (LTbR-Fc) impaired expansion of kidneys engrafted in lymph node (LN) and caused a decrease in nuclear factor kB-inducing kinase (NIK)-expressing glomerular cells. A: Schematic representation of LN transplantation assay with the course of mouse treatments. Green K stands for green fluorescent protein (GFP)þ metanephric (embryonic day 14) kidney. B: Merged fluorescence/bright-field images of whole mount kidney-bearing LNs isolated from either control Ige or LTbR-Fcetreated mice (GFPþ areas indicate kidney engraftment). C: Bar graph showing fold changes of GFP intensity (pixel counts obtained with Adobe Photoshop) in LNs as in B (blue bars: Control Ig; green bars: LTbR-Fc). D: Immunofluorescence stain for NIK (red) in glomeruli of engrafted kidneys as in B. E: Line graph showing the percentage of NIK-expressing cells in engrafted glomeruli as in B. Glomeruli were divided into five groups based on the number of NIK-expressing cells (0; 1 to 3; 4 to 6; 7 to 9; or 10 positive cells/glomerulus). Data presented in D and E are representative of one technical replicate of a minimum of three technical replicates per sample. n Z 3 per group (D and E). **P < 0.01. Scale bars: 500 mm (B); 25 mm (D).
6
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
Q23 Q24
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
although the donor and host endothelial cells could coexist in the early stages of engraftment, over time the grafts were entirely re-vascularized by host cells.24 ½F1 Figure 1A shows abundance of host (GFP) endothelial cells (CD31þ) in the interstitium of a GFPþ mouse metanephric kidney 3 weeks after transplantation in the LN, as
well as the presence of a host endothelial cell in an engrafted glomerulus. Most glomerular endothelial cells (CD34þ) expressed NIK (Figure 1B). Of interest, the number of NIKþ cells and/or NIK fluorescence intensity were significantly decreased in 6-week engrafted glomeruli (Supplemental Figure S1, A and B), suggesting that NIK
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
Growing Kidneys in Ectopic Sites
Figure 3 Treatment with lymphotoxin-b receptor antagonist (LTbR-Fc) affected lymph node (LN) kidney graft architecture and vasculature. A: Representative immunofluorescence stains for podoplanin (PDPLN; podocytes and lymphatic endothelia, red), lotus tetragonolobus lectin (LTL; proximal tubules, red), CD31 (endothelial cells, red), CD105 (newly formed vessels, red), and Ly-76 (erythrocytes, red) on serial frozen sections of kidney grafts isolated from either Control Ig- or LTbR-Fcetreated mice. Insets show a representative glomerulus for each group. B: Bar graph showing PDPLN mean fluorescence intensity (MFI; plum bars) in glomeruli of kidney grafts as in A, measured with ImageJ version 1.52a (each bar represents a glomerulus). Gray bars indicate the area of each glomerulus considered (ImageJ). C: Scatter dot plot with bar graphs SD showing the length of LTLþ tubules in kidney grafts as in A, measured across sections, using ImageJ. Each dot represents a tubule. DeF: Scatter dot plot with bar graphs SD presenting the corrected fluorescence intensities of glomerular (CTGF) CD31, CD105, and Ly-76 in kidney grafts as in A, measured with ImageJ. Each dot represents a glomerulus. Data presented in this figure are representative of one technical replicate of a minimum of three technical replicates per sample (n Z 3 per group). *P < 0.05, ****P < 0.0001. Power.t.test Z 1, sig.level Z 0.05 (C). Scale bars: 100 mm (A).
The American Journal of Pathology
-
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
7
Q25
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
expression could mark activated host endothelial cells during the early stages of kidney vascular integration in the host LN. NIK was activated in response to stimulation of tumor necrosis factor receptor superfamily members such as LTbR.25 LTbR was widely expressed on the three major CD45 LN stromal cell populationsdfibroblastic reticular cells (FRCs), lymphatic endothelial cells, and blood endothelial cellsdand was important for several adult LN functions.15 LTbR signals on FRCs regulated LN vascular
endothelial growth factor levels and, in turn, LN endothelial cell proliferation.26 Likewise, LTbR triggering on LN endothelial cells affected vasculature development.27 We therefore hypothesized that host LTbR/NIK signaling facilitated glomerular vascularization inside the LN. In accordance with our hypothesis, engrafted glomeruli were found to be embedded in a host LTbR reactive cellular scaffold, compatible with ER-TR7þ FRCs (Figure 1C). Intraglomerular LTbR expression could also be observed, which, together with a rare intraglomerular expression of the
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
Francipane et al
Figure 4 Metanephric kidney engraftment in omentum: similarity with the lymph node (LN) system. A: Frozen section of a representative green fluorescent protein (GFP)þ glomerulus 3 weeks after transplantation in the omentum. Merged GFP signal with Hoechst stain (blue; left panel), merged GFP signal with CD31 (endothelial cell) stain (red; middle panel), and merged CD31 stain (red) with Hoechst stain (blue; right panel). Circled areas are shown at higher magnification below and demonstrate the presence of a host (GFP) CD31þ cell in the engrafted glomerulus. B: Merged GFP signal with CD31 (red, left column) or nuclear factor kB-inducing kinase (NIK; red, right column) stain in serial sections of GFPþ kidney graft in the omentum (3 weeks after transplantation). Top row shows expressions of CD31 and NIK in glomeruli; bottom row, expressions of CD31 and NIK in a vessel. Arrows point to a host (GFP) CD31þ/NIKþ cell. C: Flow cytometric profiles of CD45 cells isolated from LN (left panel) or omentum (right panel). From the podoplanin (PDPLN) and CD31 expression, distinct stromal cell populations can be identified: fibroblastic reticular cells (FRCs; PDPLNþ/CD31), lymphatic endothelial cells (LECs; PDPLNþ/ CD31þ), and blood endothelial cells (BECs; PDPLN/CD31þ). D: Representative immunofluorescence stain for lymphotoxin-b receptor (LTbR; red) and PDPLN (green) on CD45 omental cells. White asterisks indicate LTbRþ/PDPLN cells and yellow asterisks indicate LTbRþ/PDPLNþ cells. Nuclei were counterstained with Hoechst (blue). Data presented in A and B are representative of one of three technical replicates per sample. n Z 3 (A and B). Flow cytometric profiles and LTbR/PDPLN stain were generated by pooling cells from 10 mice. Scale bars: 25 mm (A); 50 mm (B); 100 mm (D). Cy, cyanine; PE, phycoerythrin; PerCP, peridinin chlorophyll protein.
8
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
Q26
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
membrane-anchored LTbR ligand LT-b (Figure 1C), strengthened the idea that the LTbR/NIK axis could play a role in glomerular vascularization inside the LN.
Inhibition of LTbR Signaling Decreases Glomerular Vascularization/Angiogenesis in the LN To determine the role of LTbR signaling on kidney engraftment in the LN, 2 days before metanephric kidney
1055 transplantation, recipient mice were treated with a recom1056 binant LTbR-Fc fusion proteindwhich antagonized LTbR1057 mediated effects by engaging soluble and cell surface forms 1058 of LTbR ligandsdor control Ig (day 1). Mice were treated 1059 again at day 8 and 15, and LNs were collected at day 22 1060 (Figure 2A). LTbR-Fc treatment resulted in reduced ½F2 1061 expansion of transplanted GFPþ metanephric kidneys 1062 (Figure 2, B and C) and decreased numbers of NIKþ 1063 glomerular cells (Figure 2D). Indeed, although glomeruli 1064
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
Growing Kidneys in Ectopic Sites
Defective lymphotoxin-b receptor (LTbR) signaling in the omentum resulted in reduced kidney graft expansion. A: Merged fluorescence/brightfield (left panels) or dissecting scope (right panels) images of whole mount omenta from wild-type (WT) or LTbR/ mice 2 to 6 weeks after transplantation of green fluorescent protein (GFP)þ metanephric (embryonic day 14) kidneys. Grafts from 3 mice are shown at 6 weeks after transplantation in comparison with an adult mouse kidney. Red arrows point to the engrafted kidneys in WT or LTbR/ mice. B: Bar graph showing fold changes of GFP intensity (pixel counts obtained with Adobe Photoshop) in transplanted omenta as in A (blue bars: WT; green bars: LTbR/). Dotted lines separate the samples based on the time (weeks) elapsed from transplantation (2 weeks: n Z 1 per group; 3 weeks: n Z 4 per group; 4 weeks: n Z 1 per group; 5 weeks: n Z 1 per group; 6 weeks: n Z 3 per group). Scale bars Z 500 mm (A).
Figure 5
The American Journal of Pathology
-
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
9
Q27
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
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
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
Francipane et al
10
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
Growing Kidneys in Ectopic Sites 1303 1241 devoid of NIKþ cells could be observed in both experipossibility that the observed effects could be due to LTbR/ 1304 1242 mental groups, the number of such glomeruli was doubled herpesvirus entry mediator inhibition on donor kidney cells 1305 1243 in grafts from LTbR-Fcetreated mice (Figure 2E). Moreor other cell types present in the donor tissue at the time of 1306 1244 over, although both the groups had similar percentages of transplantation, including stromal and hematopoietic cells. 1307 1245 þ / 13 glomeruli that contained one to three NIK cells, the mice did not have LNs, the omentum Because LTbR 1308 1246 number of glomeruli with four to six NIKþ cells was was used as an alternative site for transplantation. Similarly 1309 1247 to the LN, the omentum supported the maturation of GFPdrastically reduced after LTbR-Fc treatment (Figure 2E). 1310 1248 expressing metanephric kidneys. Intraomental kidney Overall, glomeruli engrafted in LTbR-Fcetreated mice had 1311 1249 þ grafts contained host-derived (GFP) glomerular endothe1312 1250 Q16 a mean of 1.48 0.25 NIK cells, whereas glomeruli 1251 engrafted in control mice had a mean of 2.88 0.33 NIKþ lial cells (CD31þ) (Figure 4A) as well as NIK-expressing ½F4 1313 1314 1252 endothelial cells (CD31þ) in renal glomeruli and blood cells (P Z 0.001). 1315 1253 When graft maturity was investigated through immunovessels (Figure 4B). We therefore hypothesized that LN and 1316 1254 fluorescence staining for PDPLN to highlight the glomeruli, omentum supported kidney organogenesis through a similar 1317 1255 or LTL to visualize the proximal tubules, no obvious cellular/molecular mechanism. Our hypothesis was champ1318 1256 structural differences could be found between treated and ioned by the finding that both sites shared similar FRC/ 1319 1257 lymphatic endothelial cell/blood endothelial cell flow cyto½F3 control mice (Figure 3A). Further analysis confirmed that 1320 1258 metric profiles (Figure 4C). Moreover, LTbRþ/PDPLNþ neither significant changes in total surface area of glomeruli 1321 1259 nor significant changes in PDPLN expression had occurred and LTbRþ/PDPLN cell subsets could be identified in 1322 1260 (Figure 3B). Conversely, LTbR-Fc treatment resulted in omental cell suspensions, indicating the existence of FRCs 1323 1261 decreased average LTLþ tubule length, calculated in cross and blood endothelial cells that mediated LT signals in this 1324 1262 1325 1263 site as well (Figure 4D). sections (Figure 3C). The extent of vascularization/angio1326 1264 genesis was also evaluated by staining for CD31 and the Consistent with our previous findings (Figure 2, B and þ 1327 1265 neo-angiogenic marker CD105 (Figure 3A). The intraC), expansion of GFP metanephric kidneys was affected in 1328 1266 / glomerular CD31 and CD105 fluorescence intensities were the LTbR omentum. Not only did the grafts expand less 1329 1267 quantified, and a significantly weaker intensity was found over a 6-week period (Figure 5), but they also were ½F5 1330 1268 for both markers in glomeruli of LTbR-Fcetreated mice significantly less vascularized (Figure 6A). The glomerular ½F6 1331 1269 (Figure 3, D and E). Nevertheless, no significant difference CD31, CD105, and Ly-76 fluorescence intensities were 1332 1270 in glomerular blood perfusion was observed (Ly-76 stain), quantified; significant changes could be observed at every 1333 1271 although fewer erythrocytes were located within blood time point analyzed (Figure 6B). Of note, no significant 1334 1272 vessels of treated grafts (Figure 3, A and F). Thus, LTbR 1335 1273 changes in Ly-76 expression had been observed in LN signaling might mediate vascularization/angiogenesis of the 1336 1274 kidney grafts isolated from mice undergoing LTbR-Fc 1337 1275 kidneys transplanted in the LN. treatment with respect to their control counterparts 1338 1276 (Figure 3F). The cause for this different outcome remains 1339 1277 unknown. Glomerular Vascularization/Angiogenesis Is Decreased 1340 1278 CD31 and CD105 fluorescence intensities were also in Kidneys Engrafted in the LTbR/ Omentum 1341 1279 compared over time (Figure 6, C and D). CD31 expression 1342 1280 was stable up to 4 weeks in glomeruli engrafted in the wildThe LTbR ligands included the heterotrimeric LT a1b2 1343 1281 type omentum and decreased afterward (Figure 6C). (LTa1b2)28 and LT-related inducible ligand.29 Although 1344 1282 Conversely, CD31 expression began falling soon after the LTa1b2 bound uniquely to LTbR, LT-related inducible 1345 1283 second week of transplantation in glomeruli engrafted in the 1346 1284 ligand may also interact with herpesvirus entry mediator.30 1347 1285 LTbR/ omentum (Figure 6C). CD105 expression had a To rule out the possibility that impaired graft angiogenesis 1348 1286 similar trend in both experimental groups, with the highest resulted from LTbR-Fc interference with signaling path1349 1287 expression registered at 2 weeks after transplantation ways other than those mediated by LTbR, the outcome of þ 1350 1288 (Figure 6D). At this stage, significant differences in NIK kidney transplantation in an LTbR defective environment, 1351 1289 / glomerular cell numbers could also be observed between the such as that offered by the LTbR mouse, was investi1352 1290 two kidney groups (Supplemental Figure S1C and gated. In addition, this approach allowed excluding the 1353 1291 1354 1292 1355 1293 Figure 6 Defective lymphotoxin-b receptor (LTbR) signaling in the omentum resulted in reduced kidney graft vascularization/angiogenesis and decreased numbers of nuclear factor kBeinducing kinase (NIK)-expressing glomerular cells. A: Representative immunofluorescence stains for CD31 (endothelial cells, 1356 1294 red), CD105 (newly formed vessels, red), and Ly-76 (erythrocytes, red) on serial frozen sections of intraomental kidney grafts from wild-type (WT) or LTbR/ 1357 1295 mice over the course of 6 weeks. B: Scatter dot plot with bar graphs SD presenting the corrected fluorescence intensities of intraglomerular (CTGF) CD31, 1358 1296 CD105, and Ly-76 in kidney grafts as in A, measured with ImageJ version 1.52a. Each dot represents a glomerulus. C and D: Line graphs SEM presenting CD31 1359 1297 / and CD105 CTGFs over the time course (blue line: WT; green line: LTbR ). E: Line graph showing the percentage of NIK-expressing cells in glomeruli 2 weeks 1360 1298 / after transplantation in WT (blue line) or LTbR (green line) omentum. Glomeruli were divided into five groups based on the number of NIK-expressing cells 1361 1299 (0; 1 to 3; 4 to 6; 7 to 9; or 10 positive cells/glomerulus). Data presented in this figure are representative of one of three technical replicates per sample 1362 1300 (n Z 10 per group; 2 weeks: n Z 1 per group; 3 weeks: n Z 4 per group; 4 weeks: n Z 1 per group; 5 weeks: n Z 1 per group; 6 weeks: n Z 3 per group). 1301 Q28 Q29 *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Scale bars Z 100 mm (A). 1302 1363
The American Journal of Pathology
-
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
11
Figure 6E). Indeed, the number of glomeruli devoid of NIKþ cells increased in the LTbR/ omentum (Figure 6E). Moreover, although almost an equal number of glomeruli had either one to three or four to six NIKþ cells in the wildtype omentum, the number of glomeruli with one to three NIKþ cells increased in the LTbR/ omentum, to the detriment of glomeruli with four to six NIKþ cells (Figure 6E). Finally, glomeruli with seven to nine NIKþ cells were more abundant in the wild-type omentum than in the LTbR/ omentum, and glomeruli with up to 10 NIKþ cells could only be observed in the wild-type omentum
(Figure 6E). Overall, glomeruli engrafted in the LTbR/ omentum had a mean of 2.04 0.23 NIKþ cells, whereas glomeruli engrafted in the wild-type omentum had a mean of 4.04 0.30 NIKþ cells (P < 0.0001). Over the time course, the number of glomeruli devoid of NIKþ cells progressively increased in both groups, and the number of NIKþ cells per positive glomerulus also decreased (Supplemental Figure S1, D and E). Four weeks after transplantation, no more than three NIKþ cells could be observed in glomeruli engrafted in the LTbR/ omentum, whereas a small percentage of glomeruli engrafted in the
print & web 4C=FPO
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 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
Francipane et al
Figure 7 Defective lymphotoxin-b receptor (LTbR) signaling in the omentum affected kidney graft architecture. A: Representative immunofluorescence stains for collagen IV (CIV; glomerular basement membranes and mesangial cells, red), podoplanin (PDPLN; podocytes and lymphatic endothelia, red), and lotus tetragonolobus lectin (LTL; proximal tubules, red) on serial frozen sections of intraomental kidney grafts from wild-type (WT) or LTbR/ mice over the course of 6 weeks. B: Bar graph showing PDPLN mean fluorescence intensity (MFI; plum bars) in glomeruli 2 weeks after transplantation in WT or LTbR/ omentum, measured with ImageJ version 1.52a (each bar represents a glomerulus). Gray bars indicate the area of each glomerulus considered (ImageJ). C: Scatter dot plot with bar graphs SD showing the length of LTLþ tubules 2 weeks after transplantation in WT or LTbR/ omentum, measured across sections with ImageJ. Each dot represents a tubule. Power.t.test Z 1, sig.level Z 0.05. Data presented in this figure are representative of one of three technical replicates per sample (n Z 10 per group; 2 weeks: n Z 1 per group; 3 weeks: n Z 4 per group; 4 weeks: n Z 1 per group; 5 weeks: n Z 1 per group; 6 weeks: n Z 3 per group). ****P < 0.0001. Scale bars Z 100 mm (A).
12
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
Q30
1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487
Growing Kidneys in Ectopic Sites
print & web 4C=FPO
1488 wild-type omentum had four to six NIKþ cells 1489 (Supplemental Figure S1, D and E). These results suggested 1490 a critical time window between 2 and 3 weeks after trans1491 plantation for LTbR/NIK actions on the vascular remodel1492 ing of the transplanted glomeruli. 1493 1494 Structural Changes in Kidneys Engrafted in the 1495 1496 LTbR/ Omentum 1497 1498 With respect to their control counterparts, the kidneys 1499 engrafted in the LTbR/ omentum also showed structural 1500 changes overtime, with complete loss of glomeruli and tu1501 ½F7 bules by 6 weeks (Figure 7A). 1502 The time point of 2 weeks was specifically studied. 1503 Although no significant changes in total surface area of 1504 glomeruli were observed, highly significant changes 1505 affected PDPLN expression (Figure 7B) and LTLþ tubule 1506 length (Figure 7C). 1507 Tubulointerstitial injury has been described as a mediator 1508 1509 of progression of kidney disease.31 It was therefore studied 1510 whether the observed glomerular and tubular loss in later 1511 stages of engraftment could be due to the expansion of renal 1512 interstitial fibroblasts. To test this hypothesis, staining was 1513 performed for ER-TR7, which besides outlining the various 1514 compartments of peripheral lymphoid organs by detecting 1515 the reticular fibroblasts, also identifies interstitial fibroblasts 1516 in the kidney (Supplemental Figure S2A).32 Significant 1517 changes were not observed between the kidneys engrafted in 1518 the wild-type omentum versus those engrafted in the 1519 LTbR/ omentum over the 6-week period (Supplemental 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549
The American Journal of Pathology
-
Figure S2B). Moreover, in each group, ER-TR7 levels were quite stable until the fifth week, after which time they decreased (Supplemental Figure S2C). Thus, the precise mechanism of glomerular and tubular loss in late-stage grafts remains unknown. Similar results were obtained when using newborn kidneys, instead of metanephric kidneys (Supplemental Figure S3), indicating that host-driven angiogenesis was uncoupled from the maturation stage of the transplanted tissue (ie, ongoing versus completed angiogenesis).Of interest, when using newborn kidneys, more pronounced structural changes were detected between the two mouse groups, starting as early as the second week after transplantation (Supplemental Figure S3). Thus, these results pinpointed the importance of host LTbR signaling for successful vascularization/angiogenesis and maturation of the kidney grafts in lymphoid sites.
Reduced Expression of Angiogenesis-Related Genes in Kidneys Engrafted in the LTbR/ Omentum PCR array profiles of angiogenesis-related genes confirmed that host LTbR signaling sustains ectopic kidney angiogenesis. Indeed, when six distinct transplants engrafted either in the LTbR/ omentum or in the wild-type omentum 3 weeks after transplantation were compared, several proangiogenic factors were found to be down-regulated in the absence of functional LTbR signaling (Figure 8 and Supplemental Figure S4). These factors included components of the angiopoietineTie signaling system (ANGPT1,
Figure 8 Down-regulation of pro-angiogenic genes in kidneys engrafted in the lymphotoxin-b receptor (LTbR)/ omentum. A: The volcano plot displays genes differentially expressed in the kidneys engrafted in the LTbR/ omentum as compared with the kidneys engrafted in the wildtype (WT) omentum. Plotted along the x axis is the mean of log2 fold-change; along the y axis is the negative logarithm to the base 10 of the P values. The vertical dashed lines and the horizontal line reflect the filtering criteria (fold change 2.0 and P values 0.05). Green dots mark genes with decreased expression. B: Heatmap of the 14 statistically significant differentially expressed genes. C: Correlation between RT2 Profiler PCR Array and real-time RT-PCR data. Relative expression levels determined by RT2 Profiler PCR Array (x axis) and real-time RT-PCR (y axis) are plotted for a subset of 16 genes (LTbR/ 3 versus WT 3). Solid line indicates linear regression; both the squared and the Pearson correlation coefficients are given. D: Representative scatter plot (LTbR/ 3 versus WT 3) showing the gene expression patterns (presented as fold change) of selected genes by RT2 Profiler PCR Array (PCR Array) and real-time RT-PCR (RT-qPCR).
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
13
1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 ½F8 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 Q31 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611
1674 TEK, and TIE1); the epidermal growth factor signaling 1675 system (TGFA); the Epheephrin signaling system (EFNA1 1676 and EFNB2); the transforming growth factor b signaling 1677 system (ENG and TGFB3); the vascular endothelial growth 1678 factor signaling system (PGF ); and targets shared among 1679 these systems (JAG1, NOS3, PTK2, SERPINE1, TBX1). 1680 Expression of EFNB2 and SERPINE1 products was later 1681 analyzed in sections of 3-week intraomental kidney grafts. 1682 EFNB2 was found to be expressed in glomerular cells 1683 1684 (presumably podocytes and parietal epithelial cells), and 1685 PAI-1, the product of SERPINE1, was expressed in renal tubules (presumably proximal tubules) (Figure 9A). ½F9 1686 1687 Although EFNB2 fluorescence intensity was similar in both 1688 groups, a significant lower number of tubules were positive 1689 / for PAI-1 in the kidneys engrafted in the LTbR omen1690 tum (Figure 9B). A similar trend could be observed in 1691 kidneys engrafted in LNs under LTbR-Fc treatment 1692 (Figure 9, C and D), confirming that a similar cellular/mo1693 lecular mechanism drove kidney organogenesis in LNs and 1694 omenta. These results also suggested that, although the 1695 1696 lymphoid microenvironment provided signaling cues that 1697 affected the vasculature of the maturing kidney, the kidney 1698 graft self-sustains by producing angiogenic molecules. 1699 Production of these molecules appeared to be facilitated by 1700 an environment that mediated LT signals.
print & web 4C/FPO
1612 1613 1614 1615 1616 1617 1618 1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673
Francipane et al
Host NIK Is Not Required for Ectopic Kidney Organogenesis A significant reduction in the number of NIK-expressing glomerular cells had been observed in kidneys engrafted in both LNs of mice undergoing LTbR antagonist treatment (Figure 2, D and E) and omenta of LTbR/ mice (Figure 6E and Supplemental Figure S1, CeE). To elucidate whether a functional NIK was required for successful ectopic kidney organogenesis, GFPþ metanephric kidneys were transplanted in wild-type or NIK/ omenta. Significant differences in graft size, structure, vascularization, or angiogenesis were not observed at 3 weeks after Figure 9 Pro-angiogenic gene expression in kidneys engrafted in the omentum and lymph node (LN). A: Representative immunofluorescence stains for ephrin B2 (EFNB2; red) and plasminogen activator inhibitor-1 (PAI-1; red) on serial frozen sections of kidneys engrafted in wild-type (WT; top left panels) or lymphotoxin-b receptor knockout (LTbR/; bottom left panels) omenta (3 weeks after transplantation). Boxed areas are shown at higher magnification to the right. B: Scatter dot plot with bar graphs SD presenting the number of PAI-1þ tubules per microscopic field of sections stained as in A. C: Representative immunofluorescence stains for EFNB2 and PAI-1 (red) on serial frozen sections of LN kidney grafts isolated from either mice treated with Control Ig (top left panels) or LTbRFc (bottom left panels) (3 weeks after transplantation). Boxed areas are shown at higher magnification to the right. D: Scatter dot plot with bar graphs SD presenting the number of PAI-1þ tubules per microscopic field of sections stained as in C. Data presented in this figure are representative of one of three technical replicates per sample (n Z 3 per group). ***P < 0.001, ****P < 0.0001. Scale bars: 100 mm (A and C). GFP, green fluorescent protein.
14
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
Q32
1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735
transplantation (Supplemental Figure S5, AeK), indicating that, although NIK could be activated by LTbR signaling in this system, it was dispensable for ectopic kidney organogenesis. Moreover, NIK-expressing cells could still be detected in kidneys engrafted in the NIK/ omentum 3 weeks after transplantation (Supplemental Figure S6), although NIK fluorescence intensity was much lower than that observed in kidneys engrafted in the wild-type omentum at the same stage. These cells were likely donor cells, which might have been trying to compensate for the missing host NIK function.
LTbR Plays a Role in Glomerular Adaptive Responses to Nx Demonstration that LTbR signaling was important for adult kidney repair would strengthen our hypothesis that kidney rebuilding approaches might benefit from the presence of a microenvironment that mediated LTbR signals. To our knowledge, there are no studies that investigated LTbR physiological role in kidney development or during compensatory growth. The structure of LTbR/ kidneys versus wild-type kidneys was first compared. LTbR did not appear to be essential for kidney development during embryogenesis because LTbR/ mice had normal kidneys with what appeared to be normal vascularization and structure (Supplemental Figure S7). This finding likely suggested
1798 differences between ectopic organogenesis and normal 1799 organogenesis. Indeed, although LTbR-angiogenetic sig1800 nals appeared to be important for ectopic kidney organ1801 ogenesis, in normal kidney organogenesis (which 1802 occurs mainly through vasculogenesis), LTbR was not so 1803 crucial. 1804 LTbR relative contribution to compensatory renal growth 1805 after removal of one kidney was therefore investigated to 1806 corroborate the idea that not only was LTbR signaling 1807 1808 beneficial for glomerular vascularization/angiogenesis in 1809 lymphoid sites, but it also might play a role in glomerular 1810 adaptation to renal mass reduction. 1811 Contralateral compensatory renal growth and increased 1812 renal function followed contralateral Nx in experimental 1813 33 models. Accordingly, although kidneys of wild-type 1814 and LTbR/ mice had similar weights before surgery, 1815 LTbR/ mice showed significantly lower weights of their 1816 remnant kidneys 10 days after Nx (Figure 10A). The ½F101817 contralateral kidneys in wild-type mice showed glomerular 1818 enlargement as assessed in H&E-stained sections 1819 1820 (Figure 10B). These glomeruli often contained more eryth1821 rocytes, indicating an increase of renal blood flow. Differ1822 ences in adaptive responses seemed to be confined to the 1823 glomerulus, because tubular epithelial cells from both 1824 mouse groups showed a similar widespread positivity for 1825 the Ki-67 proliferation marker (Figure 10C). These data 1826 indicated a more efficient glomerular cell response to uni1827 lateral Nx in the presence of functional LTbR signaling. 1828
print & web 4C=FPO
1736 1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775 1776 1777 1778 1779 1780 1781 1782 1783 1784 1785 1786 1787 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797
Growing Kidneys in Ectopic Sites
Figure 10 Lymphotoxin-b receptor (LTbR) played a role in renal responses to nephrectomy (Nx). A: Scatter dot plot with bar graph SD showing kidney weights before and after Nx (PreNx and PostNx, respectively) in both wild-type (WT) and LTbR/ mice. Each dot represents a mouse. B: Left panel: Scatter dot plot with bar graph SD showing glomerular diameters in WT and LTbR/ PostNx kidneys. Each dot represents a glomerulus. Right panel: Representative hematoxylin and eosin (H&E) stain in Sham, WT_PostNx, or LTbR/_PostNx kidney paraffin sections. C: Left panel: Scatter dot plot with bar graph SD showing the percentage of Ki-67þ tubular cell nuclei from WT and LTbR/ PostNx kidneys. Each dot represents one stained section. Right panel: Representative immunohistochemistry for Ki-67 in WT and LTbR/ PostNx kidney paraffin sections (3-amino-9-ethylcarbazole, red). *P < 0.05, **P < 0.01, ***P < 0.001. Scale bars: 50 mm (B and C).
The American Journal of Pathology
-
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
15
Q33
1829 1830 1831 1832 1833 1834 1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855 1856 1857 1858 1859
1860 1861 1862 1863 1864 1865 1866 1867 1868 1869 1870 1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888 1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919 1920 1921
Francipane et al
Discussion LNs and omenta can be used as bioreactors to grow organs. LN grafting is a unique approach pioneered by our laboratory,11,12,24,34,35 with no previous records in the literature. Conversely, the greater omentum has long been described as an ideal transplantation site mainly for its easy accessibility and large vascular network.36 Nevertheless, to our knowledge, cellular and molecular mechanisms that allow tissue and organ development within these locations have not been explored yet. By transplanting kidney rudiments either in the LNs of mice undergoing LTbR antagonist treatment or in the omenta of LTbR/ mice, it has been shown that host LTbR signals are crucial for obtaining a well-vascularized kidney graft. Indeed, defective host LTbR signaling correlated with decreased expression of endothelial and angiogenic markers in the transplanted tissues as well as structural alterations. Although the number of glomerular endothelial cells expressing the LTbR target NIK decreased in the absence of a functional LTbR, the results from the transplantation of kidney rudiments in the omenta of NIK/ mice indicated that host NIK is not required for successful ectopic kidney organogenesis. NIK and subsequent non-canonical NF-kB signaling have been described to regulate both inflammation-induced and tumor-associated angiogenesis.17 From this perspective, kidney angiogenesis in lymphoid tissues shares features with pathologic angiogenesis. However, the progressive reduction in NIK-expressing cells that was observed in glomeruli engrafted in LNs and omenta seems to suggest a tight regulation of angiogenesis in the ectopic kidney grafts, whereby NIK likely elicits a transient signal. In the wildtype omentum, for example, NIK and the neoangiogenic marker CD105 shared a similar pattern, with decreased expressions in engrafted glomeruli starting as early as the second week after transplantation, a pattern different from that of the endothelial cell marker CD31, which was quite stable for 4 weeks. These data seem to suggest that NIK is required during the early stages of engraftment, coinciding with early angiogenesis and tissue vascular integration, but is no longer needed in later stages of kidney maturation. NIK-mediated signals, however, are not necessary for kidney organogenesis in lymphoid sites, because overall vascularization/angiogenesis was not affected in NIK/ recipients. Besides activating non-canonical NF-kB pathway through NIK,16 LTbR could also trigger the canonical NF-kB route to stimulate endothelial cells.37 Moreover, other angiogenic signals elicited by the donor tissue itself must not be underestimated, as indicated by the data of PAI-1 production by the kidney tubular cells. How host LTbR signals can influence donor tissue production of PAI-1 and possibly of other angiogenetic molecules remains unknown.
16
Kidney tissue integration after transplantation likely involves extensive vascular remodeling, whereby several players are involved, including endothelial cells, smooth muscle cells, and fibroblasts. To record these dynamic processes is technically demanding, but not impossible. Transplanting GFPþ tissue in a wild-type recipient has been an effective method for visualizing the engrafted tissue later on, but the use of mice with constitutive or inducible endogenous fluorescent reporters, in combination with tracer injections and/or mosaic genetic recombination,38 would better allow characterizing the multicellular dynamics of angiogenesis and vascular remodeling in the transplanted tissues. Three-dimensional reconstruction of the engrafted kidneys that was recently performed on human fetal kidneys engrafted in the LN24 would also allow us to better quantify the differences in tubular elongation across the experimental groups. Indeed, the current adopted strategy of LTLþ tubule length quantification on two-dimensional sections cannot be considered accurate, because the apparent tubule length depends on the level at which the tissue was sectioned and observed. However, this strategy highlighted some important changes in the absence of a functional LTbR signal that warrant future study. Despite these limitations, the findings of host lymphoid stromal microenvironment contribution to ectopic kidney organogenesis seem to suggest implications for future development of new approaches to treat kidney disease. The renal sub-capsular space has served as the preferred site for kidney regeneration from embryonic metanephroi39,40 and recently kidney organoids.41e43 However, there is no convincing evidence that this approach can improve kidney function after a major injury. The inflammatory and fibrotic milieu of a host’s diseased kidney could constitute an adverse environment for cell and tissue engraftment and ultimately function. Finally, clinical progress in renal subcapsular transplantation has lagged behind other techniques because of the inelastic and tight nature of the human renal sub-capsular space.44 Additional strategies to engineer functional kidneys include repopulating synthetic/decellularized scaffolds with kidney-specific cells and transplanting these constructs orthotopically.45 Unfortunately, these approaches have often failed to recapitulate native kidney functions because of inadequate blood supply. Indeed, despite efforts to endow the scaffolds with an endothelium,45 it is still not possible to generate vascularized and perfused renal tissues that produce urine and/or have endocrine functions such as generation erythropoietin,46 necessary for erythropoiesis.47 Although reconstructing a fully functional kidney in the LN or the omentum for clinical purposes is challenging, stromal cells from these sites could be exploited to create an artificial scaffold for renal progenitor cells and thus provide an auxiliary aid during renal failure. A major concern about this approach might arise from the LTbR documented role in kidney disease.
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
1922 1923 1924 1925 1926 1927 1928 1929 1930 1931 1932 1933 1934 1935 1936 1937 1938 1939 1940 1941 1942 1943 1944 1945 1946 1947 1948 1949 1950 1951 1952 1953 1954 1955 1956 1957 1958 1959 1960 1961 1962 1963 1964 1965 1966 1967 1968 1969 1970 1971 1972 1973 1974 1975 1976 1977 1978 1979 1980 1981 1982 1983
1984 1985 1986 1987 1988 1989 1990 1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034 2035 2036 2037 2038 2039 2040 2041 2042 2043 2044 2045
Growing Kidneys in Ectopic Sites Renal biopsies from lupus nephritis and IgA nephropathy patients showed elevated LT/LTbR levels, and LTbR inhibition improved renal function in a murine lupus model.48 LTbR-Ig was efficacious when given from week 3 to 7, whereas delaying the treatment until 5 weeks did not lead to significant results, highlighting a dynamic role for LTbR across different stages of disease.48 LTbR signaling in lupus likely results from the formation of tertiary lymphoid organs. Unlike secondary lymphoid organs such as the LN, tertiary lymphoid organs arise in response to nonresolving inflammation over the course of several years. Therefore, although tertiary lymphoid organ-mediated LTbR signaling leads to a prolonged, deregulated, and maladaptive response in injured kidneys that involves active inflammation and repeated unsuccessful attempts to repair the tissue, secondary lymphoid organ-mediated LTbR signaling might be exploited to support kidney development. The finding that glomerular adaptive responses to loss of renal mass were impaired in the absence of a functional LTbR indicated that LTbR can participate to repair responses after injury, although we cannot rule out that these responses might become maladaptive over time. In conclusion, although our results do not allow any unbiased conclusion on the precise cellular/molecular machineries that support kidney organogenesis in lymphoid sites, they point to an important function of the lymphoid stromal microenvironment in sustaining angiogenesis that warrants future investigation. Better understanding of the unique properties of this microenvironment might have implications for future engineering of functional organs.
Acknowledgments We thank Hannah Liu and Adam DeDionisio for technical support with tissue sectioning and immunofluorescence/ immunohistochemistry staining and Lynda Guzik (McGowan Institute Flow Cytometry Facility) for help with flow cytometric profiling of lymph node and omental stromal cell populations and for proofreading the manuscript. M.G.F. designed the study, performed most of the experiments, analyzed the data, prepared the figures, and wrote the manuscript; B.H. managed mouse colonies, performed the mouse surgeries, isolated omental stromal cells, and analyzed them by immunofluorescence; E.L. designed and supervised the study; all authors discussed the results and approved the final version of the manuscript.
Supplemental Data Supplemental material for this article can be found at http://doi.org/10.1016/j.ajpath.2019.08.018.
The American Journal of Pathology
-
References 1. Glassock RJ, Warnock DG, Delanaye P: The global burden of chronic kidney disease: estimates, variability and pitfalls. Nat Rev Nephrol 2017, 13:104e114 2. Neild GH: Life expectancy with chronic kidney disease: an educational review. Pediatr Nephrol 2017, 32:243e248 3. Glassock RJ, Pecoits-Filho R, Barberato SH: Left ventricular mass in chronic kidney disease and ESRD. Clin J Am Soc Nephrol 2009, 4 Suppl 1:S79eS91 4. Abecassis M, Bartlett ST, Collins AJ, Davis CL, Delmonico FL, Friedewald JJ, Hays R, Howard A, Jones E, Leichtman AB, Merion RM, Metzger RA, Pradel F, Schweitzer EJ, Velez RL, Gaston RS: Kidney transplantation as primary therapy for end-stage renal disease: a National Kidney Foundation/Kidney Disease Outcomes Quality Initiative (NKF/KDOQITM) conference. Clin J Am Soc Nephrol 2008, 3:471e480 5. Christians U, Klawitter J, Klawitter J, Brunner N, Schmitz V: Biomarkers of immunosuppressant organ toxicity after transplantation: status, concepts and misconceptions. Expert Opin Drug Metab Toxicol 2011, 7:175e200 6. Hammerman MR: Organogenesis of kidneys following transplantation of renal progenitor cells. Transpl Immunol 2004, 12: 229e239 7. Woolf AS, Palmer SJ, Snow ML, Fine LG: Creation of a functioning chimeric mammalian kidney. Kidney Int 1990, 38:991e997 8. Hyink DP, Tucker DC, St John PL, Leardkamolkarn V, Accavitti MA, Abrass CK, Abrahamson DR: Endogenous origin of glomerular endothelial and mesangial cells in grafts of embryonic kidneys. Am J Physiol 1996, 270:F886eF899 9. Rogers SA, Lowell JA, Hammerman NA, Hammerman MR: Transplantation of developing metanephroi into adult rats. Kidney Int 1998, 54:27e37 10. Matsumoto K, Yokoo T, Yokote S, Utsunomiya Y, Ohashi T, Hosoya T: Functional development of a transplanted embryonic kidney: effect of transplantation site. J Nephrol 2012, 25:50e55 11. Francipane MG, Lagasse E: The lymph node as a new site for kidney organogenesis. Stem Cells Transl Med 2015, 4:295e307 12. Francipane MG, Lagasse E: Regenerating a kidney in a lymph node. Pediatr Nephrol 2016, 31:1553e1560 13. De Togni P, Goellner J, Ruddle NH, Streeter PR, Fick A, Mariathasan S, Smith SC, Carlson R, Shornick LP, StraussSchoenberger J, Russell JH, Karr R, Chaplin DD: Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science 1994, 264:703e707 14. Futterer A, Mink K, Luz A, Kosco-Vilbois MH, Pfeffer K: The lymphotoxin beta receptor controls organogenesis and affinity maturation in peripheral lymphoid tissues. Immunity 1998, 9:59e70 15. Browning JL, Allaire N, Ngam-Ek A, Notidis E, Hunt J, Perrin S, Fava RA: Lymphotoxin-beta receptor signaling is required for the homeostatic control of HEV differentiation and function. Immunity 2005, 23:539e550 16. Sun SC: Non-canonical NF-kappaB signaling pathway. Cell Res 2011, 21:71e85 17. Noort AR, van Zoest KP, Weijers EM, Koolwijk P, Maracle CX, Novack DV, Siemerink MJ, Schlingemann RO, Tak PP, Tas SW: NFkappaB-inducing kinase is a key regulator of inflammation-induced and tumour-associated angiogenesis. J Pathol 2014, 234:375e385 18. Committee for the Update of the Guide for the Care and Use of Laboratory AnimalsNational Research Council: Guide for the Care and Use of Laboratory Animals: Eighth Edition. Washington, DC, National Academies Press, 2011 19. Boehm T, Scheu S, Pfeffer K, Bleul CC: Thymic medullary epithelial cell differentiation, thymocyte emigration, and the control of autoimmunity require lympho-epithelial cross talk via LTbetaR. J Exp Med 2003, 198:757e769
ajp.amjpathol.org
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
17
2046 2047 2048 2049 2050 2051 2052 2053 2054 2055 2056 2057 2058 2059 2060 2061 2062 2063 2064 2065 2066 2067 2068 2069 2070 2071 2072 2073 2074 2075 2076 2077 2078 2079 2080 2081 2082 2083 2084 2085 2086 2087 2088 2089 2090 2091 2092 2093 2094 2095 2096 2097 2098 2099 2100 2101 2102 2103 2104 2105 2106 2107
2108 2109 2110 2111 2112 2113 2114 2115 2116 2117 2118 2119 2120 2121 2122 2123 2124 2125 2126 2127 2128 2129 2130 2131 2132 2133 2134 2135 2136 2137 2138 2139 2140 2141 2142 2143 2144 2145 2146 2147 2148 2149 2150 2151 2152 2153 2154 2155 2156 2157 2158 2159 2160 2161 2162 2163 2164 2165 2166 2167 2168 2169
Francipane et al 20. Franki AS, Van Beneden K, Dewint P, Hammond KJ, Lambrecht S, Leclercq G, Kronenberg M, Deforce D, Elewaut D: A unique lymphotoxin {alpha}beta-dependent pathway regulates thymic emigration of V{alpha}14 invariant natural killer T cells. Proc Natl Acad Sci U S A 2006, 103:9160e9165 21. Cardiff RD, Miller CH, Munn RJ: Manual hematoxylin and eosin staining of mouse tissue sections. Cold Spring Harb Protoc 2014, 2014:655e658 22. Haddad G, Zhabyeyev P, Farhan M, Zhu LF, Kassiri Z, Rayner DC, Vanhaesebroeck B, Oudit GY, Murray AG: Phosphoinositide 3kinase beta mediates microvascular endothelial repair of thrombotic microangiopathy. Blood 2014, 124:2142e2149 23. Fletcher AL, Malhotra D, Acton SE, Lukacs-Kornek V, BellemarePelletier A, Curry M, Armant M, Turley SJ: Reproducible isolation of lymph node stromal cells reveals site-dependent differences in fibroblastic reticular cells. Front Immunol 2011, 2:35 24. Francipane MG, Han B, Oxburgh L, Sims-Lucas S, Li Z, Lagasse E: Kidney-in-a-lymph node: a novel organogenesis assay to model human renal development and test nephron progenitor cell fates. J Tissue Eng Regen Med 2019, 13:1724e1731 25. Matsushima A, Kaisho T, Rennert PD, Nakano H, Kurosawa K, Uchida D, Takeda K, Akira S, Matsumoto M: Essential role of nuclear factor (NF)-kappaB-inducing kinase and inhibitor of kappaB (IkappaB) kinase alpha in NF-kappaB activation through lymphotoxin beta receptor, but not through tumor necrosis factor receptor I. J Exp Med 2001, 193:631e636 26. Chyou S, Ekland EH, Carpenter AC, Tzeng TC, Tian S, Michaud M, Madri JA, Lu TT: Fibroblast-type reticular stromal cells regulate the lymph node vasculature. J Immunol 2008, 181:3887e3896 27. Onder L, Danuser R, Scandella E, Firner S, Chai Q, Hehlgans T, Stein JV, Ludewig B: Endothelial cell-specific lymphotoxin-beta receptor signaling is critical for lymph node and high endothelial venule formation. J Exp Med 2013, 210:465e473 28. Upadhyay V, Fu YX: Lymphotoxin signalling in immune homeostasis and the control of microorganisms. Nat Rev Immunol 2013, 13: 270e279 29. Zhai Y, Guo R, Hsu TL, Yu GL, Ni J, Kwon BS, Jiang GW, Lu J, Tan J, Ugustus M, Carter K, Rojas L, Zhu F, Lincoln C, Endress G, Xing L, Wang S, Oh KO, Gentz R, Ruben S, Lippman ME, Hsieh SL, Yang D: LIGHT, a novel ligand for lymphotoxin beta receptor and TR2/HVEM induces apoptosis and suppresses in vivo tumor formation via gene transfer. J Clin Invest 1998, 102:1142e1151 30. Mauri DN, Ebner R, Montgomery RI, Kochel KD, Cheung TC, Yu GL, Ruben S, Murphy M, Eisenberg RJ, Cohen GH, Spear PG, Ware CF: LIGHT, a new member of the TNF superfamily, and lymphotoxin alpha are ligands for herpesvirus entry mediator. Immunity 1998, 8:21e30 31. Nangaku M: Mechanisms of tubulointerstitial injury in the kidney: final common pathways to end-stage renal failure. Intern Med 2004, 43:9e17 32. Tsujie M, Isaka Y, Ando Y, Akagi Y, Kaneda Y, Ueda N, Imai E, Hori M: Gene transfer targeting interstitial fibroblasts by the artificial viral envelope-type hemagglutinating virus of Japan liposome method. Kidney Int 2000, 57:1973e1980 33. Hayslett JP: Effect of age on compensatory renal growth. Kidney Int 1983, 23:599e602
18
34. Komori J, Boone L, DeWard A, Hoppo T, Lagasse E: The mouse lymph node as an ectopic transplantation site for multiple tissues. Nat Biotechnol 2012, 30:976e983 35. Francipane MG, Lagasse E: Maturation of embryonic tissues in a lymph node: a new approach for bioengineering complex organs. Organogenesis 2014, 10:323e331 36. Smink AM, Faas MM, de Vos P: Toward engineering a novel transplantation site for human pancreatic islets. Diabetes 2013, 62: 1357e1364 37. Dejardin E, Droin NM, Delhase M, Haas E, Cao Y, Makris C, Li ZW, Karin M, Ware CF, Green DR: The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways. Immunity 2002, 17:525e535 38. Wang Y, Jin Y, Lavina B, Jakobsson L: Characterization of multicellular dynamics of angiogenesis and vascular remodelling by intravital imaging of the wounded mouse cornea. Sci Rep 2018, 8: 10672 39. Hammerman MR: Renal organogenesis from transplanted metanephric primordia. J Am Soc Nephrol 2004, 15:1126e1132 40. Imberti B, Corna D, Rizzo P, Xinaris C, Abbate M, Longaretti L, Cassis P, Benedetti V, Benigni A, Zoja C, Remuzzi G, Morigi M: Renal primordia activate kidney regenerative events in a rat model of progressive renal disease. PLoS One 2015, 10:e0120235 41. Xinaris C, Benedetti V, Rizzo P, Abbate M, Corna D, Azzollini N, Conti S, Unbekandt M, Davies JA, Morigi M, Benigni A, Remuzzi G: In vivo maturation of functional renal organoids formed from embryonic cell suspensions. J Am Soc Nephrol 2012, 23:1857e1868 42. Sharmin S, Taguchi A, Kaku Y, Yoshimura Y, Ohmori T, Sakuma T, Mukoyama M, Yamamoto T, Kurihara H, Nishinakamura R: Human induced pluripotent stem cell-derived podocytes mature into vascularized glomeruli upon experimental transplantation. J Am Soc Nephrol 2016, 27:1778e1791 43. van den Berg CW, Ritsma L, Avramut MC, Wiersma LE, van den Berg BM, Leuning DG, Lievers E, Koning M, Vanslambrouck JM, Koster AJ, Howden SE, Takasato M, Little MH, Rabelink TJ: Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo. Stem Cell Reports 2018, 10:751e765 44. Sakata N, Yoshimatsu G, Kodama S: The spleen as an optimal site for islet transplantation and a source of mesenchymal stem cells. Int J Mol Sci 2018, 19:E1391 45. Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti JP, Ott HC: Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med 2013, 19:646e651 46. Obara N, Suzuki N, Kim K, Nagasawa T, Imagawa S, Yamamoto M: Repression via the GATA box is essential for tissue-specific erythropoietin gene expression. Blood 2008, 111:5223e5232 47. Adamson JW: Regulation of red blood cell production. Am J Med 1996, 101:4Se6S 48. Seleznik G, Seeger H, Bauer J, Fu K, Czerkowicz J, Papandile A, Poreci U, Rabah D, Ranger A, Cohen CD, Lindenmeyer M, Chen J, Edenhofer I, Anders HJ, Lech M, Wuthrich RP, Ruddle NH, Moeller MJ, Kozakowski N, Regele H, Browning JL, Heikenwalder M, Segerer S: The lymphotoxin beta receptor is a potential therapeutic target in renal inflammation. Kidney Int 2016, 89:113e126
ajp.amjpathol.org
-
The American Journal of Pathology
FLA 5.6.0 DTD AJPA3220_proof 19 October 2019 12:33 am EO: 2019_49
2170 2171 2172 2173 2174 2175 2176 2177 2178 2179 2180 2181 2182 2183 2184 2185 2186 2187 2188 2189 2190 2191 2192 2193 2194 2195 2196 2197 2198 2199 2200 2201 2202 2203 2204 2205 2206 2207 2208 2209 2210 2211 2212 2213 2214 2215 2216 2217 2218 2219 2220 2221 2222 2223 2224 2225 2226 2227 2228 2229 2230 2231