2 Human CNS Barrier-Forming Organoids With Cerebrospinal Fluid Production

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

HOME  SCIENCE  VOL. 369, NO. 6500  HUMAN CNS BARRIER-FORMING ORGANOIDS WITH CEREBROSPINAL FLUID PRODUCTION

RESEARCH ARTICLE
 󡀆 

Human CNS barrier-forming organoids with cerebrospinal fluid


production
LAURA PELLEGRINI
, CLAUDIA BONFIO , JESSICA CHADWICK, FARIDA BEGUM, MARK SKEHEL , AND , MADELINE A. LANCASTER  Authors Info & Affiliations

SCIENCE • 10 Jul 2020 • Vol 369, Issue 6500 • DOI: 10.1126/science.aaz5626

5 60     GET ACCESS

Brain barrier and support in a dish

Deep within the brain, the choroid plexus filters blood and secretes cerebrospinal fluid (CSF), a nutrient-rich
liquid that bathes and supports the brain and protects it from entry of toxic compounds. Current understand-
ing of this vital tissue in humans is limited. Pellegrini et al. developed choroid plexus organoids that quantita-
 
tively predict human brain permeability of small molecules and secrete an isolated CSF-like fluid (see the

Perspective by Silva-Vargas and Doetsch). This CSF model reveals secretion of developmental factors and dis-
ease-related biomarkers by key cell types and provides a testing ground for drug entry into the brain.

Science, this issue p. eaaz5626; see also p. 143 




Structured Abstract 

INTRODUCTION

The choroid plexus is a secretory epithelial tissue of the central nervous system (CNS) responsible for cere-
brospinal fluid (CSF) production and functions as a barrier that regulates entry of compounds and nutrients
into the brain. The CSF plays key roles in the delivery of nutrients to the brain, circulation of instructive sig-
naling molecules, and clearance of toxic by-products such as protein aggregates.

RATIONALE

Current understanding of the choroid plexus and CSF has primarily come from animal models or CSF collected
from human volunteers. These have yielded insight into general CSF composition, but the specific cellular and
tissue sources of various secreted proteins have remained elusive. There is also limited understanding of the
development of the choroid plexus in humans and of the relative changes in CSF composition over time. All of
these deficiencies in our understanding come from a lack of experimental access to the human choroid plexus.
Although several previous studies have successfully generated cells with a choroid plexus identity from human
pluripotent stem cells, none have been able to recapitulate the morphology, maturation, and function of the
choroid plexus, and currently, no in vitro model exists for authentic human CSF. Knowledge of the processes
that regulate choroid plexus development and CSF composition could provide better strategies to manipulate
and therapeutically target this vital brain tissue.

RESULTS

To study the development and function of the human choroid plexus, we developed a pluripotent stem cell–
derived organoid model. Choroid plexus organoids recapitulate key morphological and functional features of
human choroid plexus. First, organoids form a tight barrier that selectively regulates the entry of small mole-
cules such as dopamine. We demonstrate that organoids can qualitatively and quantitatively predict the per-
meability of new drugs, and we take advantage of this system to reveal a potential toxic accumulation of BIA
10-2474, a drug that caused severe neurotoxicity only in humans and not in animal models tested. Second,
https://www.science.org/doi/10.1126/science.aaz5626 1/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

choroid plexus organoids secrete a CSF-like fluid containing proteins and known biomarkers within self-con-
tained compartments. We examine changes in secretion of CSF proteins over time and identify distinct cell
types within the epithelium that contribute to dynamic changes in CSF composition. We find that these cell
types can be traced to rather obscure descriptions in the literature of “dark” and “light” cells, and we demon-
strate that these cells exhibit opposing features related to mitochondria and cilia. We also uncover a previously
unidentified cell type in the choroid plexus: myoepithelial cells. These interacting subpopulations exhibit dis-
tinct secretory roles in CSF production and reveal previously uncharacterized human-specific secreted proteins
that may play important roles in human brain development.

CONCLUSION

Human choroid plexus organoids provide an easily tractable system to study the key functions of this organ:
CSF secretion and selective transport into the CNS. As such, they can predict CNS permeability of new com-
pounds to aid in the development of neurologically relevant therapeutics. They also provide a source of more
authentic CSF and can be used to understand development of this key organ in brain development and
homeostasis.

 





CSF-producing choroid plexus organoids predict CNS permeability of drugs.


Choroid plexus organoids develop highly intricate folded tissue morphology (section stained for choroid plexus markers shown at top
right) similar to choroid plexus tissue in vivo (top left) and, later, self-contained fluid-filled compartments containing a CSF-like fluid (top
middle) that is separate from media. (Bottom left) Choroid plexus organoids accurately predict the permeability of small molecules such
as dopamine and levodopa (l-dopa) and quantitatively predict the permeability of a range of therapeutic molecules. The graph shows the
correlation between permeability in vivo and in vitro for the drugs tested. R2, coefficient of determination. (Bottom right) Single-cell RNA
sequencing reveals newly identified epithelial subtypes (colored dark and light) that participate in filtration and specialized secretion of
CSF proteins. The Venn diagram shows overlap between proteins detected in CSF in vivo and in the organoid.

https://www.science.org/doi/10.1126/science.aaz5626 2/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

Abstract

Cerebrospinal fluid (CSF) is a vital liquid, providing nutrients and signaling molecules and clearing out toxic
by-products from the brain. The CSF is produced by the choroid plexus (ChP), a protective epithelial barrier
that also prevents free entry of toxic molecules or drugs from the blood. Here, we establish human ChP
organoids with a selective barrier and CSF-like fluid secretion in self-contained compartments. We show that
this in vitro barrier exhibits the same selectivity to small molecules as the ChP in vivo and that ChP-CSF
organoids can predict central nervous system (CNS) permeability of new compounds. The transcriptomic and
proteomic signatures of ChP-CSF organoids reveal a high degree of similarity to the ChP in vivo. Finally, the
intersection of single-cell transcriptomics and proteomic analysis uncovers key human CSF components pro-
duced by previously unidentified specialized epithelial subtypes.

Get full access to this article


View all available purchase options and get full access to this article.

 GET ACCESS

ALREADY A SUBSCRIBER? SIGN IN

 

Supplementary Material 

Summary

Materials and Methods

Figs. S1 to S13

Tables S1 to S5

References (50–128)

MDAR Reproducibility Checklist

Data S1 to S3

Resources

File (aaz5626_data_s1.xlsx)

DOWNLOAD

File (aaz5626_data_s2.xlsx)

DOWNLOAD

File (aaz5626_data_s3.xlsx)

DOWNLOAD

File (aaz5626_mdar_reproducibility_checklist.pdf)

DOWNLOAD

File (aaz5626_pellegrini_sm.pdf)

DOWNLOAD

https://www.science.org/doi/10.1126/science.aaz5626 3/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

File (aaz5626_pellegrini_sm_rev1.pdf)

DOWNLOAD

File (pap.pdf)

DOWNLOAD

View/request a protocol for this paper from Bio-protocol.

Correction (9 July 2020): A new reference 98 was added to the supplementary reference list.

References and Notes


1 M. P. Lun, E. S. Monuki, M. K. Lehtinen, Development and functions of the choroid plexus-cerebrospinal fluid system. Nat. Rev. Neurosci. 16,
445–457 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

2 M. K. Lehtinen, C. A. Walsh, Neurogenesis at the brain-cerebrospinal fluid interface. Annu. Rev. Cell Dev. Biol. 27, 653–679 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

3 adult
V. Silva-Vargas, A. R. Maldonado-Soto, D. Mizrak, P. Codega, F. Doetsch, Age-dependent niche signals from the choroid plexus regulate
neural stem cells. Cell Stem Cell 19, 643–652 (2016).
CROSSREF • PUBMED • GOOGLE SCHOLAR


4 M. P. Lun, M. B. Johnson, K. G. Broadbelt, M. Watanabe, Y. J. Kang, K. F. Chau, M. W. Springel, A. Malesz, A. M. M. Sousa, M. Pletikos, T.

Adelita, M. L. Calicchio, Y. Zhang, M. J. Holtzman, H. G. W. Lidov, N. Sestan, H. Steen, E. S. Monuki, M. K. Lehtinen, Spatially heterogeneous
choroid plexus transcriptomes encode positional identity and contribute to regional CSF production. J. Neurosci. 35, 4903–4916 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR 

5 J. F. Ghersi-Egea, N. Strazielle, M. Catala, V. Silva-Vargas, F. Doetsch, B. Engelhardt, Molecular anatomy and functions of the choroidal
blood-cerebrospinal fluid barrier in health and disease. Acta Neuropathol. 135, 337–361 (2018). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

6 Z. Redzic, Molecular biology of the blood-brain and the blood-cerebrospinal fluid barriers: Similarities and differences. Fluids Barriers CNS 8,
3 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

7 G. J. Dohrmann, P. C. Bucy, Human choroid plexus: A light and electron microscopic study. J. Neurosurg. 33, 506–516 (1970).
CROSSREF • PUBMED • GOOGLE SCHOLAR

8 M. A. Lancaster, M. Renner, C. A. Martin, D. Wenzel, L. S. Bicknell, M. E. Hurles, T. Homfray, J. M. Penninger, A. P. Jackson, J. A. Knoblich,
Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).
CROSSREF • PUBMED • GOOGLE SCHOLAR

9 G. Quadrato, T. Nguyen, E. Z. Macosko, J. L. Sherwood, S. Min Yang, D. R. Berger, N. Maria, J. Scholvin, M. Goldman, J. P. Kinney, E. S. Boyden,
J. W. Lichtman, Z. M. Williams, S. A. McCarroll, P. Arlotta, Cell diversity and network dynamics in photosensitive human brain organoids.
Nature 545, 48–53 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

10 S. P. Pașca, The rise of three-dimensional human brain cultures. Nature 553, 437–445 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

11 H. Sakaguchi, T. Kadoshima, M. Soen, N. Narii, Y. Ishida, M. Ohgushi, J. Takahashi, M. Eiraku, Y. Sasai, Generation of functional hippocam-
pal neurons from self-organizing human embryonic stem cell-derived dorsomedial telencephalic tissue. Nat. Commun. 6, 8896 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

12 R. M. Fame, M. K. Lehtinen, Emergence and developmental roles of the cerebrospinal fluid system. Dev. Cell 52, 261–275 (2020).
CROSSREF • PUBMED • GOOGLE SCHOLAR

13 M. A. Lancaster, N. S. Corsini, S. Wolfinger, E. H. Gustafson, A. W. Phillips, T. R. Burkard, T. Otani, F. J. Livesey, J. A. Knoblich, Guided self-
organization and cortical plate formation in human brain organoids. Nat. Biotechnol. 35, 659–666 (2017).

https://www.science.org/doi/10.1126/science.aaz5626 4/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

CROSSREF • PUBMED • GOOGLE SCHOLAR

14 S. L. Giandomenico, S. B. Mierau, G. M. Gibbons, L. M. D. Wenger, L. Masullo, T. Sit, M. Sutcliffe, J. Boulanger, M. Tripodi, E. Derivery, O.
Paulsen, A. Lakatos, M. A. Lancaster, Cerebral organoids at the air-liquid interface generate diverse nerve tracts with functional output. Nat.
Neurosci. 22, 669–679 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

15 M. Watanabe, Y.-J. Kang, L. M. Davies, S. Meghpara, K. Lau, C.-Y. Chung, J. Kathiriya, A.-K. Hadjantonakis, E. S. Monuki, BMP4 sufficiency to
induce choroid plexus epithelial fate from embryonic stem cell-derived neuroepithelial progenitors. J. Neurosci. 32, 15934–15945 (2012).
CROSSREF • PUBMED • GOOGLE SCHOLAR

16 D. S. Currle, X. Cheng, C. M. Hsu, E. S. Monuki, Direct and indirect roles of CNS dorsal midline cells in choroid plexus epithelia formation.
Development 132, 3549–3559 (2005).
CROSSREF • PUBMED • GOOGLE SCHOLAR

17 M. K. Lehtinen, C. S. Bjornsson, S. M. Dymecki, R. J. Gilbertson, D. M. Holtzman, E. S. Monuki, The choroid plexus and cerebrospinal fluid:
Emerging roles in development, disease, and therapy. J. Neurosci. 33, 17553–17559 (2013).
CROSSREF • PUBMED • GOOGLE SCHOLAR

18 S. A. Liddelow, Development of the choroid plexus and blood-CSF barrier. Front. Neurosci. 9, 32 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

19 M. Sathyanesan, M. J. Girgenti, M. Banasr, K. Stone, C. Bruce, E. Guilchicek, K. Wilczak-Havill, A. Nairn, K. Williams, S. Sass, J. G. Duman, S.
 S. Newton, A molecular characterization of the choroid plexus and stress-induced gene regulation. Transl. Psychiatry 2, e139–e9 (2012). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

N. Dani, R. H. Herbst, N. Habib, J. Head, D. Dionne, L. Nguyen, C. McCabe, J. Cui, F. B. Shipley, A. Jang, C. Rodman, S. J. Riesenfeld, F.
20

Zhang, O. Rozenblatt-Rosen, A. Regev, M. K. Lehtinen, A cellular and spatial map of the choroid plexus across brain ventricles and ages.
bioRxiv 627539 [Preprint]. 5 May 2019; . 
CROSSREF • GOOGLE SCHOLAR

21 T. J. Nowakowski, A. Bhaduri, A. A. Pollen, B. Alvarado, M. A. Mostajo-Radji, E. Di Lullo, M. Haeussler, C. Sandoval-Espinosa, S. J. Liu, D.

Velmeshev, J. R. Ounadjela, J. Shuga, X. Wang, D. A. Lim, J. A. West, A. A. Leyrat, W. J. Kent, A. R. Kriegstein, Spatiotemporal gene expres-
sion trajectories reveal developmental hierarchies of the human cortex. Science 358, 1318–1323 (2017). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

22 J. Cao, M. Spielmann, X. Qiu, X. Huang, D. M. Ibrahim, A. J. Hill, F. Zhang, S. Mundlos, L. Christiansen, F. J. Steemers, C. Trapnell, J.
Shendure, The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

23 R. Duelli, B. E. Enerson, D. Z. Gerhart, L. R. Drewes, Expression of large amino acid transporter LAT1 in rat brain endothelium. J. Cereb.
Blood Flow Metab. 20, 1557–1562 (2000).
CROSSREF • PUBMED • GOOGLE SCHOLAR

24 H. Uchino, Y. Kanai, D. K. Kim, M. F. Wempe, A. Chairoungdua, E. Morimoto, M. W. Anders, H. Endou, Transport of amino acid-related com-
pounds mediated by L-type amino acid transporter 1 (LAT1): Insights into the mechanisms of substrate recognition. Mol. Pharmacol. 61,
729–737 (2002).
CROSSREF • PUBMED • GOOGLE SCHOLAR

25 R. Durso, J. E. Evans, E. Josephs, G. Szabo, B. Evans, H. H. Fernandez, T. R. Browne, Variable absorption of carbidopa affects both peripheral
and central levodopa metabolism. J. Clin. Pharmacol. 40, 854–860 (2000).
CROSSREF • PUBMED • GOOGLE SCHOLAR

26 V. V. Rao, J. L. Dahlheimer, M. E. Bardgett, A. Z. Snyder, R. A. Finch, A. C. Sartorelli, D. Piwnica-Worms, Choroid plexus epithelial expression
of MDR1 P glycoprotein and multidrug resistance-associated protein contribute to the blood–cerebrospinal-fluid drug-permeability barrier.
Proc. Natl. Acad. Sci. U.S.A. 96, 3900–3905 (1999).
CROSSREF • PUBMED • GOOGLE SCHOLAR

27 C. J. Ek, A. Wong, S. A. Liddelow, P. A. Johansson, K. M. Dziegielewska, N. R. Saunders, Efflux mechanisms at the developing brain barriers:
ABC-transporters in the fetal and postnatal rat. Toxicol. Lett. 197, 51–59 (2010).
CROSSREF • PUBMED • GOOGLE SCHOLAR

28 Y. Uchida, Z. Zhang, M. Tachikawa, T. Terasaki, Quantitative targeted absolute proteomics of rat blood-cerebrospinal fluid barrier trans-
porters: Comparison with a human specimen. J. Neurochem. 134, 1104–1115 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

https://www.science.org/doi/10.1126/science.aaz5626 5/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

29 S. G. Summerfield, Y. Zhang, H. Liu, Examining the uptake of central nervous system drugs and candidates across the blood-brain barrier. J.
Pharmacol. Exp. Ther. 358, 294–305 (2016).
CROSSREF • PUBMED • GOOGLE SCHOLAR

30 L. Angelov, N. D. Doolittle, D. F. Kraemer, T. Siegal, G. H. Barnett, D. M. Peereboom, G. Stevens, J. McGregor, K. Jahnke, C. A. Lacy, N. A.
Hedrick, E. Shalom, S. Ference, S. Bell, L. Sorenson, R. M. Tyson, M. Haluska, E. A. Neuwelt, Blood-brain barrier disruption and intra-arterial
methotrexate-based therapy for newly diagnosed primary CNS lymphoma: A multi-institutional experience. J. Clin. Oncol. 27, 3503–3509
(2009).
CROSSREF • PUBMED • GOOGLE SCHOLAR

31 E. Zylber-Katz, J. M. Gomori, A. Schwartz, A. Lossos, F. Bokstein, T. Siegal, Pharmacokinetics of methotrexate in cerebrospinal fluid and
serum after osmotic blood-brain barrier disruption in patients with brain lymphoma. Clin. Pharmacol. Ther. 67, 631–641 (2000).
CROSSREF • PUBMED • GOOGLE SCHOLAR

32 I. Das, A. Krzyzosiak, K. Schneider, L. Wrabetz, M. D. Antonio, N. Barry, A. Sigurdardottir, A. Bertolotti, Preventing proteostasis diseases by
selective inhibition of a phosphatase regulatory subunit. 348, 239–243 (2015).
GOOGLE SCHOLAR

33 C. H. I. H. Wong, K. W. E. I. Siah, A. W. Lo, Estimation of clinical trial success rates and related parameters. Biostatistics 20, 273–286 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

34 R. Kaur, P. Sidhu, S. Singh, What failed BIA 10-2474 phase I clinical trial? Global speculations and recommendations for future phase I trials.
J. Pharmacol. Pharmacother. 7, 120–126 (2016).
 CROSSREF • PUBMED • GOOGLE SCHOLAR 

35 
K. Otrubova, C. Ezzili, D. L. Boger, The discovery and development of inhibitors of fatty acid amide hydrolase (FAAH). Bioorg. Med. Chem.
Lett. 21, 4674–4685 (2011).

CROSSREF • PUBMED • GOOGLE SCHOLAR


36 D. Butler, E. Callaway, Scientists in the dark after French clinical trial proves fatal. Nature 529, 263–264 (2016).
CROSSREF • PUBMED • GOOGLE SCHOLAR


37 S. M. Bird, R. A. Bailey, A. P. Grieve, S. Senn, Statistical issues in first-in-human studies on BIA 10-2474: Neglected comparison of protocol
against practice. Pharm. Stat. 16, 100–106 (2017). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

38 E. Kida, S. Palminiello, A. A. Golabek, M. Walus, T. Wierzba-Bobrowicz, A. Rabe, G. Albertini, K. E. Wisniewski, Carbonic anhydrase II in the
developing and adult human brain. J. Neuropathol. Exp. Neurol. 65, 664–674 (2006).
CROSSREF • PUBMED • GOOGLE SCHOLAR

39 J. B. Wollack, B. Makori, S. Ahlawat, R. Koneru, S. C. Picinich, A. Smith, I. D. Goldman, A. Qiu, P. D. Cole, J. Glod, B. Kamen, Characterization
of folate uptake by choroid plexus epithelial cells in a rat primary culture model. J. Neurochem. 104, 1494–1503 (2008).
CROSSREF • PUBMED • GOOGLE SCHOLAR

40 V. Ulloa, N. Saldivia, L. Ferrada, K. Salazar, F. Martínez, C. Silva-Alvarez, R. Magdalena, M. J. Oviedo, H. Montecinos, P. Torres-Vergara, M.
Cifuentes, F. Nualart, Basal sodium-dependent vitamin C transporter 2 polarization in choroid plexus explant cells in normal or scorbutic
conditions. Sci. Rep. 9, 14422 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

41 A. Roy, M. M. Al-bataineh, N. M. Pastor-Soler, Collecting duct intercalated cell function and regulation. Clin. J. Am. Soc. Nephrol. 10, 305–
324 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

42 A. A. Pollen, T. J. Nowakowski, J. Chen, H. Retallack, C. Sandoval-Espinosa, C. R. Nicholas, J. Shuga, S. J. Liu, M. C. Oldham, A. Diaz, D. A.
Lim, A. A. Leyrat, J. A. West, A. R. Kriegstein, Molecular identity of human outer radial glia during cortical development. Cell 163, 55–67
(2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

43 J.-M. Nicolas, in Blood-Brain Barrier in Drug Discovery: Optimizing Brain Exposure of CNS Drugs and Minimizing Brain Side Effects for Peripheral
Drugs, L. Di, E. H. Kerns, Eds. (Wiley, 2015).
GOOGLE SCHOLAR

44 M. A. Lancaster, J. A. Knoblich, Generation of cerebral organoids from human pluripotent stem cells. Nat. Protoc. 9, 2329–2340 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

https://www.science.org/doi/10.1126/science.aaz5626 6/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

45 J. A. Miller, S.-L. Ding, S. M. Sunkin, K. A. Smith, L. Ng, A. Szafer, A. Ebbert, Z. L. Riley, J. J. Royall, K. Aiona, J. M. Arnold, C. Bennet, D.
Bertagnolli, K. Brouner, S. Butler, S. Caldejon, A. Carey, C. Cuhaciyan, R. A. Dalley, N. Dee, T. A. Dolbeare, B. A. C. Facer, D. Feng, T. P. Fliss,
G. Gee, J. Goldy, L. Gourley, B. W. Gregor, G. Gu, R. E. Howard, J. M. Jochim, C. L. Kuan, C. Lau, C.-K. Lee, F. Lee, T. A. Lemon, P. Lesnar, B.
McMurray, N. Mastan, N. Mosqueda, T. Naluai-Cecchini, N.-K. Ngo, J. Nyhus, A. Oldre, E. Olson, J. Parente, P. D. Parker, S. E. Parry, A.
Stevens, M. Pletikos, M. Reding, K. Roll, D. Sandman, M. Sarreal, S. Shapouri, N. V. Shapovalova, E. H. Shen, N. Sjoquist, C. R.
Slaughterbeck, M. Smith, A. J. Sodt, D. Williams, L. Zöllei, B. Fischl, M. B. Gerstein, D. H. Geschwind, I. A. Glass, M. J. Hawrylycz, R. F.
Hevner, H. Huang, A. R. Jones, J. A. Knowles, P. Levitt, J. W. Phillips, N. Šestan, P. Wohnoutka, C. Dang, A. Bernard, J. G. Hohmann, E. S.
Lein, Transcriptional landscape of the prenatal human brain. Nature 508, 199–206 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

46 U. Raudvere, L. Kolberg, I. Kuzmin, T. Arak, P. Adler, H. Peterson, J. Vilo, g:Profiler: a web server for functional enrichment analysis and con-
versions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

47 L. Dayon, O. Cominetti, J. Wojcik, A. N. Galindo, A. Oikonomidi, H. Henry, E. Migliavacca, M. Kussmann, G. L. Bowman, J. Popp, Proteomes
of paired human cerebrospinal fluid and plasma: Relation to blood-brain barrier permeability in older adults. J. Proteome Res. 18, 1162–1174
(2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

48 L. Guo, H. Ren, H. Zeng, Y. Gong, X. Ma, Proteomic analysis of cerebrospinal fluid in pediatric acute lymphoblastic leukemia patients: A pi-
lot study. OncoTargets Ther. 12, 3859–3868 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

49 M. D. Zappaterra, S. N. Lisgo, S. Lindsay, S. P. Gygi, C. A. Walsh, B. A. Ballif, A comparative proteomic analysis of human and rat embryonic
 cerebrospinal fluid. J. Proteome Res. 6, 3537–3548 (2007). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

50 
B. J. Stewart, J. R. Ferdinand, M. D. Young, T. J. Mitchell, K. W. Loudon, A. M. Riding, N. Richoz, G. L. Frazer, J. U. L. Staniforth, F. A. Vieira
Braga, R. A. Botting, D.-M. Popescu, R. Vento-Tormo, E. Stephenson, A. Cagan, S. J. Farndon, K. Polanski, M. Efremova, K. Green, M. Del
Castillo Velasco-Herrera, C. Guzzo, G. Collord, L. Mamanova, T. Aho, J. N. Armitage, A. C. P. Riddick, I. Mushtaq, S. Farrell, D. Rampling,  J.
Nicholson, A. Filby, J. Burge, S. Lisgo, S. Lindsay, M. Bajenoff, A. Y. Warren, G. D. Stewart, N. Sebire, N. Coleman, M. Haniffa, S. A.
Teichmann, S. Behjati, M. R. Clatworthy, Spatiotemporal immune zonation of the human kidney. Science 365, 1461–1466 (2019). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

51 
D. N. Perkins, D. J. C. Pappin, D. M. Creasy, J. S. Cottrell, Probability-based protein identification by searching sequence databases using
mass spectrometry data. Electrophoresis 20, 3551–3567 (1999).
CROSSREF • PUBMED • GOOGLE SCHOLAR

52 A. Keller, A. I. Nesvizhskii, E. Kolker, R. Aebersold, Empirical statistical model to estimate the accuracy of peptide identifications made by
MS/MS and database search. Anal. Chem. 74, 5383–5392 (2002).
CROSSREF • PUBMED • GOOGLE SCHOLAR

53 A. Lex, N. Gehlenborg, Sets and intersections. Nat. Methods 11, 779 (2014).
CROSSREF • GOOGLE SCHOLAR

54 M. Fridén, S. Winiwarter, G. Jerndal, O. Bengtsson, H. Wan, U. Bredberg, M. Hammarlund-Udenaes, M. Antonsson, Structure-brain exposure
relationships in rat and human using a novel data set of unbound drug concentrations in brain interstitial and cerebrospinal fluids. J. Med.
Chem. 52, 6233–6243 (2009).
CROSSREF • PUBMED • GOOGLE SCHOLAR

55 D. E. Vetter, J. R. Mann, P. Wangemann, J. Liu, K. J. McLaughlin, F. Lesage, D. C. Marcus, M. Lazdunski, S. F. Heinemann, J. Barhanin, Inner
ear defects induced by null mutation of the isk gene. Neuron 17, 1251–1264 (1996).
CROSSREF • PUBMED • GOOGLE SCHOLAR

56 W. E. Evans, P. R. Hutson, C. F. Stewart, D. A. Cairnes, W. P. Bowman, G. Rivera, W. R. Crom, Methotrexate cerebrospinal fluid and serum
concentrations after intermediate-dose methotrexate infusion. Clin. Pharmacol. Ther. 33, 301–307 (1983).
CROSSREF • PUBMED • GOOGLE SCHOLAR

57 R. N. Golden, M. V. Rudorfer, M. A. Sherer, M. Linnoila, W. Z. Potter, Bupropion in depression. I. Biochemical effects and clinical response.
Arch. Gen. Psychiatry 45, 139–143 (1988).
CROSSREF • PUBMED • GOOGLE SCHOLAR

58 A. Onasch, A. Tanzeem, F. Isgro, D. Böning, G. Strobel, Effect of intravenous dopamine infusion on plasma concentrations of dopamine and
dopamine sulfate in men, during and up to 18 h after infusion. Eur. J. Clin. Pharmacol. 55, 755–759 (2000).
CROSSREF • PUBMED • GOOGLE SCHOLAR

https://www.science.org/doi/10.1126/science.aaz5626 7/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

59 N. Rizzo, J.-M. Scherrmann, C. Padoin, S. Palombo, C. Girre, Omeprazole and lansoprazole are not inducers of cytochrome P4501A2 under
conventional therapeutic conditions. Eur. J. Clin. Pharmacol. 49, 491–495 (1996).
CROSSREF • PUBMED • GOOGLE SCHOLAR

60 P. Benetello, M. Furlanut, G. Zara, M. Baraldo, E. Hassan, Plasma levels of levodopa and its main metabolites in parkinsonian patients after
conventional and controlled-release levodopa-carbidopa associations. Eur. Neurol. 33, 69–73 (1993).
CROSSREF • PUBMED • GOOGLE SCHOLAR

61 D. V. Jackson Jr., V. S. Sethi, C. L. Spurr, J. M. McWhorter, Pharmacokinetics of vincristine in the cerebrospinal fluid of humans. Cancer Res.
41, 1466–1468 (1981).
PUBMED • GOOGLE SCHOLAR

62 I. Seri, T. Tulassay, J. Kiszel, T. Machay, S. Csömör, Cardiovascular response to dopamine in hypotensive preterm neonates with severe hya-
line membrane disease. Eur. J. Pediatr. 142, 3–9 (1984).
CROSSREF • PUBMED • GOOGLE SCHOLAR

63 V. R. Varma, S. Varma, Y. An, T. J. Hohman, S. Seddighi, R. Casanova, A. Beri, E. B. Dammer, N. T. Seyfried, O. Pletnikova, A. Moghekar, M. R.
Wilson, J. J. Lah, R. J. O’Brien, A. I. Levey, J. C. Troncoso, M. S. Albert, M. Thambisetty, Alpha-2 macroglobulin in Alzheimer’s disease: A
marker of neuronal injury through the RCAN1 pathway. Mol. Psychiatry 22, 13–23 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

64 S. Westwood, E. Leoni, A. Hye, S. Lynham, M. R. Khondoker, N. J. Ashton, S. J. Kiddle, A. L. Baird, R. Sainz-Fuertes, R. Leung, J. Graf, C. T.
Hehir, D. Baker, C. Cereda, C. Bazenet, M. Ward, M. Thambisetty, S. Lovestone, Blood-based biomarker candidates of cerebral amyloid using
 PiB PET in non-demented elderly. J. Alzheimers Dis. 52, 561–572 (2016). 
CROSSREF • PUBMED • GOOGLE SCHOLAR


65 A. Kim, L. Ji, C. Balmaceda, B. Diez, S. J. Kellie, I. J. Dunkel, S. L. Gardner, R. Sposto, J. L. Finlay, The prognostic value of tumor markers in
newly diagnosed patients with primary central nervous system germ cell tumors. Pediatr. Blood Cancer 51, 768–773 (2008).

CROSSREF • PUBMED • GOOGLE SCHOLAR

66 
G. Legault, J. C. Allen, Potential role of ventricular tumor markers in CNS germ cell tumors. Pediatr. Blood Cancer 60, 1647–1650 (2013).
CROSSREF • PUBMED • GOOGLE SCHOLAR

67 
A. Quaranta, I. Karlsson, L. Ndreu, F. Marini, M. Ingelsson, G. Thorsén, Glycosylation profiling of selected proteins in cerebrospinal fluid
from Alzheimer’s disease and healthy subjects. Anal. Methods 11, 3331–3340 (2019).
CROSSREF • GOOGLE SCHOLAR

68 M. Costa, R. Horrillo, A. M. Ortiz, A. Pérez, A. Mestre, A. Ruiz, M. Boada, S. Grancha, Increased albumin oxidation in cerebrospinal fluid and
plasma from Alzheimer’s disease patients. J. Alzheimers Dis. 63, 1395–1404 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

69 P. Lewczuk, A. Gaignaux, O. Kofanova, N. Ermann, F. Betsou, S. Brandner, B. Mroczko, K. Blennow, D. Strapagiel, S. Paciotti, J. Vogelgsang,
M. H. Roehrl, S. Mendoza, J. Kornhuber, C. Teunissen, Interlaboratory proficiency processing scheme in CSF aliquoting : Implementation
and assessment based on biomarkers of Alzheimer’s disease. Alzheimers Res. Ther. 10, 87 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

70 G. C. Li, L. Zhang, M. Yu, H. Jia, T. Tian, J. Wang, F. Wang, L. Zhou, Identification of novel biomarker and therapeutic target candidates for
acute intracerebral hemorrhage by quantitative plasma proteomics. Clin. Proteomics 14, 14 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

71 L. M. Schilde, S. Kösters, S. Steinbach, K. Schork, M. Eisenacher, S. Galozzi, M. Turewicz, K. Barkovits, B. Mollenhauer, K. Marcus, C. May,
Protein variability in cerebrospinal fluid and its possible implications for neurological protein biomarker research. PLOS ONE 13, e0206478
(2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

72 A. Batista Muñoz, S. Hadley, M. Iriondo Sanz, T. Agut Quijano, M. Camprubí Camprubí, Role of beta-2-microglobulin as a biomarker in very
preterm and extremely preterm infants with CNS inflammation. PLOS ONE 14, e0216498 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

73 J. Svatoňová, K. Bořecká, P. Adam, V. Lánská, Beta2-microglobulin as a diagnostic marker in cerebrospinal fluid: A follow-up study. Dis.
Markers 2014, 495402 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

74 X. Huang, J. Xu, Y. Shen, L. Zhang, M. Xu, M. Chen, J. Ren, L. Zhou, H. Gong, P. Zhong, Protein profiling of cerebrospinal fluid from patients
undergoing vestibular schwannoma surgery and clinical significance. Biomed. Pharmacother. 116, 108985 (2019).

https://www.science.org/doi/10.1126/science.aaz5626 8/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

CROSSREF • PUBMED • GOOGLE SCHOLAR

75 F. W. Khwaja, J. D. L. Nolen, S. E. Mendrinos, M. M. Lewis, J. J. Olson, J. Pohl, E. G. Van Meir, J. C. Ritchie, D. J. Brat, Proteomic analysis of
cerebrospinal fluid discriminates malignant and nonmalignant disease of the central nervous system and identifies specific protein markers.
Proteomics 6, 6277–6287 (2006).
CROSSREF • PUBMED • GOOGLE SCHOLAR

76 W. Jongbloed, K. D. van Dijk, S. D. Mulder, W. D. J. van de Berg, M. A. Blankenstein, W. van der Flier, R. Veerhuis, Clusterin levels in plasma
predict cognitive decline and progression to Alzheimer’s disease. J. Alzheimers Dis. 46, 1103–1110 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

77 H. Kessler, F.-G. Pajonk, P. Meisser, T. Schneider-Axmann, K.-H. Hoffmann, T. Supprian, W. Herrmann, R. Obeid, G. Multhaup, P. Falkai, T.
A. Bayer, Cerebrospinal fluid diagnostic markers correlate with lower plasma copper and ceruloplasmin in patients with Alzheimer’s disease.
J. Neural Transm. 113, 1763–1769 (2006).
CROSSREF • PUBMED • GOOGLE SCHOLAR

78 A. R. Kallianpur, H. Gittleman, S. Letendre, R. Ellis, J. S. Barnholtz-Sloan, W. S. Bush, R. Heaton, D. C. Samuels, D. R. Franklin Jr., D. Rosario-
Cookson, D. B. Clifford, A. C. Collier, B. Gelman, C. M. Marra, J. C. McArthur, J. A. McCutchan, S. Morgello, I. Grant, D. Simpson, J. R.
Connor, T. Hulgan; CHARTER Study Group, Cerebrospinal fluid ceruloplasmin, haptoglobin, and vascular endothelial growth factor are as-
sociated with neurocognitive impairment in adults with HIV infection. Mol. Neurobiol. 56, 3808–3818 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

79 S. Tsuji-Akimoto, I. Yabe, M. Niino, S. Kikuchi, H. Sasaki, Cystatin C in cerebrospinal fluid as a biomarker of ALS. Neurosci. Lett. 452, 52–55
(2009).
 
CROSSREF • PUBMED • GOOGLE SCHOLAR


80 Y. Ren, W. Zhu, F. Cui, F. Yang, Z. Chen, L. Ling, X. Huang, Measurement of cystatin C levels in the cerebrospinal fluid of patients with amy-
otrophic lateral sclerosis. Int. J. Clin. Exp. Pathol. 8, 5419–5426 (2015). 
PUBMED • GOOGLE SCHOLAR

81 
A. A. Albayar, A. Roche, P. Swiatkowski, S. Antar, N. Ouda, E. Emara, D. H. Smith, A. K. Ozturk, B. I. Awad, Biomarkers in spinal cord injury:
Prognostic insights and future potentials. Front. Neurol. 10, 27 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR


82 C. Zenzmaier, J. Marksteiner, A. Kiefer, P. Berger, C. Humpel, Dkk-3 is elevated in CSF and plasma of Alzheimer’s disease patients. J.
Neurochem. 110, 653–661 (2009).
CROSSREF • PUBMED • GOOGLE SCHOLAR

83 W. E. Heywood, D. Galimberti, E. Bliss, E. Sirka, R. W. Paterson, N. K. Magdalinou, M. Carecchio, E. Reid, A. Heslegrave, C. Fenoglio, E.
Scarpini, J. M. Schott, N. C. Fox, J. Hardy, K. Bhatia, S. Heales, N. J. Sebire, H. Zetterberg, K. Mills, Identification of novel CSF biomarkers for
neurodegeneration and their validation by a high-throughput multiplexed targeted proteomic assay. Mol. Neurodegener. 10, 64 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

84 K. E. McLimans, A. A. Willette; Alzheimer’s Disease Neuroimaging Initiative, Autotaxin is related to metabolic dysfunction and predicts
Alzheimer’s disease outcomes. J. Alzheimers Dis. 56, 403–413 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

85 L. Higginbotham, L. Ping, E. B. Dammer, D. M. Duong, Integrated proteomics reveals brain-based cerebrospinal fluid biomarkers in asymp-
tomatic and symptomatic Alzheimer’s disease. bioRxiv 806752 [Preprint]. 12 November 2019; .
CROSSREF • GOOGLE SCHOLAR

86 E. M. Castaño, A. E. Roher, C. L. Esh, T. A. Kokjohn, T. Beach, Comparative proteomics of cerebrospinal fluid in neuropathologically-con-
firmed Alzheimer’s disease and non-demented elderly subjects. Neurol. Res. 28, 155–163 (2006).
CROSSREF • PUBMED • GOOGLE SCHOLAR

87 A. E. Roher, C. L. Maarouf, L. I. Sue, Y. Hu, J. Wilson, T. G. Beach, Proteomics-derived cerebrospinal fluid markers of autopsy-confirmed
Alzheimer’s disease. Biomarkers 14, 493–501 (2009).
CROSSREF • PUBMED • GOOGLE SCHOLAR

88 F. Shen, Y. Zhang, Y. Yao, W. Hua, H. S. Zhang, J. S. Wu, P. Zhong, L. F. Zhou, Proteomic analysis of cerebrospinal fluid: Toward the identifi-
cation of biomarkers for gliomas. Neurosurg. Rev. 37, 367–380 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

89 F. H. Hochberg, N. A. Atai, D. Gonda, M. S. Hughes, B. Mawejje, L. Balaj, R. S. Carter, Glioma diagnostics and biomarkers: An ongoing chal-
lenge in the field of medicine and science. Expert Rev. Mol. Diagn. 14, 439–452 (2014).

https://www.science.org/doi/10.1126/science.aaz5626 9/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

CROSSREF • PUBMED • GOOGLE SCHOLAR

90 F. Martínez-Ricarte, R. Mayor, E. Martínez-Sáez, C. Rubio-Pérez, E. Pineda, E. Cordero, M. Cicuéndez, M. A. Poca, N. López-Bigas, S. Ramon
Y Cajal, M. Vieito, J. Carles, J. Tabernero, A. Vivancos, S. Gallego, F. Graus, J. Sahuquillo, J. Seoane, Molecular diagnosis of diffuse gliomas
through sequencing of cell-free circulating tumor DNA from cerebrospinal fluid. Clin. Cancer Res. 24, 2812–2819 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

91 L. W. Bonham, E. G. Geier, N. Z. R. Steele, D. Holland, B. L. Miller, A. M. Dale, R. S. Desikan, J. S. Yokoyama; Alzheimer’s Disease
Neuroimaging Initiative, Insulin-like growth factor binding protein 2 is associated with biomarkers of Alzheimer’s disease pathology and
shows differential expression in transgenic mice. Front. Neurosci. 12, 476 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

92 D. Åberg, P. Johansson, J. Isgaard, A. Wallin, J.-O. Johansson, U. Andreasson, K. Blennow, H. Zetterberg, N. D. Åberg, J. Svensson, Increased
cerebrospinal fluid level of insulin-like growth factor-II in male patients with Alzheimer’s disease. J. Alzheimers Dis. 48, 637–646 (2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

93 M. Dzieciatkowska, G. Qi, J. You, K. G. Bemis, H. Sahm, H. M. Lederman, T. O. Crawford, L. M. Gelbert, C. Rothblum-Oviatt, M. Wang,
Proteomic characterization of cerebrospinal fluid from ataxia-telangiectasia (A-T) patients using a LC/MS-based label-free protein quantifi-
cation technology. Int. J. Proteomics 2011, 578903 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

94 A. Poljak, M. Hill, R. J. Hall, A. M. MacLullich, M. J. Raftery, J. Tai, S. Yan, G. A. Caplan, Quantitative proteomics of delirium cerebrospinal
fluid. Transl. Psychiatry 4, e477 (2014).

 CROSSREF • PUBMED • GOOGLE SCHOLAR


95 
N. Khosroshahi, P. Alizadeh, M. Khosravi, P. Salamati, K. Kamrani, Spinal fluid lactate dehydrogenase level differentiates between structural
and metabolic etiologies of altered mental status in children. Iran. J. Child. Neurol. 9, 31–36 (2015).
PUBMED • GOOGLE SCHOLAR


96 L. Parnetti, A. Castrioto, D. Chiasserini, E. Persichetti, N. Tambasco, O. El-Agnaf, P. Calabresi, Cerebrospinal fluid biomarkers in Parkinson
disease. Nat. Rev. Neurol. 9, 131–140 (2013). 
CROSSREF • PUBMED • GOOGLE SCHOLAR

97 M. S. Rotunno, M. Lane, W. Zhang, P. Wolf, P. Oliva, C. Viel, A.-M. Wills, R. N. Alcalay, C. R. Scherzer, L. S. Shihabuddin, K. Zhang, S. P.
Sardi,
Cerebrospinal fluid proteomics implicates the granin family in Parkinson’s disease. Sci. Rep. 10, 2479 (2020).
CROSSREF • PUBMED • GOOGLE SCHOLAR

98 L. Dayon, A. Núñez Galindo, J. Wojcik, O. Cominetti, J. Corthésy, A. Oikonomidi, H. Henry, M. Kussmann, E. Migliavacca, I. Severin, G. L.
Bowman, J. Popp, Alzheimer disease pathology and the cerebrospinal fluid proteome. Alzheimers Res. Ther. 10, 66 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

99 X. Yan, L. Mai, C. Lin, W. He, G. Yin, J. Yu, L. Huang, S. Pan, CSF-based analysis for identification of potential serum biomarkers of neural
tube defects. Neurosci. Bull. 33, 436–444 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

100 C. Chen, D. Xiao, W. Zhou, Q. Shi, H.-F. Zhang, J. Zhang, C. Tian, J.-Z. Zhang, X.-P. Dong, Global protein differential expression profiling of
cerebrospinal fluid samples pooled from Chinese sporadic CJD and non-CJD patients. Mol. Neurobiol. 49, 290–302 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

101 G. Sathe, C. H. Na, S. Renuse, A. K. Madugundu, M. Albert, A. Moghekar, A. Pandey, Quantitative proteomic profiling of cerebrospinal fluid
to identify candidate biomarkers for Alzheimer’s disease. Proteomics Clin. Appl. 13, e1800105 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

102 S. Vuletic, H. Kennedy, J. J. Albers, J. Killestein, H. Vrenken, D. Lütjohann, C. E. Teunissen, Cerebrospinal fluid apolipoprotein E and phos-
pholipid transfer protein activity are reduced in multiple sclerosis; relationships with the brain MRI and CSF lipid variables. Mult. Scler.
Relat. Disord. 3, 533–541 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

103 S. Vuletic, E. R. Peskind, S. M. Marcovina, J. F. Quinn, M. C. Cheung, H. Kennedy, J. A. Kaye, L. W. Jin, J. J. Albers, Reduced CSF PLTP activ-
ity in Alzheimer’s disease and other neurologic diseases; PLTP induces ApoE secretion in primary human astrocytes in vitro. J. Neurosci.
Res. 80, 406–413 (2005).
CROSSREF • PUBMED • GOOGLE SCHOLAR

104 H. Kanamaru, F. Kawakita, F. Nakano, Y. Miura, M. Shiba, R. Yasuda, N. Toma, H. Suzuki; pSEED group, Plasma periostin and delayed cere-
bral ischemia after aneurysmal subarachnoid hemorrhage. Neurotherapeutics 16, 480–490 (2019).

https://www.science.org/doi/10.1126/science.aaz5626 10/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

CROSSREF • PUBMED • GOOGLE SCHOLAR

105 Y. Manevich, S. Hutchens, P. V. Halushka, K. D. Tew, D. M. Townsend, E. C. Jauch, K. Borg, Peroxiredoxin VI oxidation in cerebrospinal fluid
correlates with traumatic brain injury outcome. Free Radic. Biol. Med. 72, 210–221 (2014).
CROSSREF • PUBMED • GOOGLE SCHOLAR

106 C. Kim, H. Choi, E. S. Jung, W. Lee, S. Oh, N. L. Jeon, I. Mook-Jung, HDAC6 inhibitor blocks amyloid beta-induced impairment of mitochon-
drial transport in hippocampal neurons. PLOS ONE 7, e42983 (2012).
CROSSREF • PUBMED • GOOGLE SCHOLAR

107 D. Martins-De-Souza, T. Wobrock, I. Zerr, A. Schmitt, J. Gawinecka, T. Schneider-Axmann, P. Falkai, C. W. Turck, Different apolipoprotein
E, apolipoprotein A1 and prostaglandin-H2 D-isomerase levels in cerebrospinal fluid of schizophrenia patients and healthy controls. World
J. Biol. Psychiatry 11, 719–728 (2010).
CROSSREF • PUBMED • GOOGLE SCHOLAR

108 R. J. Perrin, R. Craig-Schapiro, J. P. Malone, A. R. Shah, P. Gilmore, A. E. Davis, C. M. Roe, E. R. Peskind, G. Li, D. R. Galasko, C. M. Clark, J.
F. Quinn, J. A. Kaye, J. C. Morris, D. M. Holtzman, R. R. Townsend, A. M. Fagan, Identification and validation of novel cerebrospinal fluid
biomarkers for staging early Alzheimer’s disease. PLOS ONE 6, e16032 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

109 H. M. Nielsen, L. Minthon, E. Londos, K. Blennow, E. Miranda, J. Perez, D. C. Crowther, D. A. Lomas, S. M. Janciauskiene, Plasma and CSF
serpins in Alzheimer disease and dementia with Lewy bodies. Neurology 69, 1569–1579 (2007).
CROSSREF • PUBMED • GOOGLE SCHOLAR

 
110 C. Ditzen, N. Tang, A. M. Jastorff, L. Teplytska, A. Yassouridis, G. Maccarrone, M. Uhr, T. Bronisch, C. A. Miller, F. Holsboer, C. W. Turck,
Cerebrospinal fluid biomarkers for major depression confirm relevance of associated pathophysiology. Neuropsychopharmacology 37,
1013–
1025 (2012).
CROSSREF • PUBMED • GOOGLE SCHOLAR


111 L. Winer, D. Srinivasan, S. Chun, D. Lacomis, M. Jaffa, A. Fagan, D. M. Holtzman, E. Wancewicz, C. F. Bennett, R. Bowser, M. Cudkowicz, T.

M. Miller, SOD1 in cerebral spinal fluid as a pharmacodynamic marker for antisense oligonucleotide therapy. JAMA Neurol. 70, 201–207
(2013).
CROSSREF • PUBMED • GOOGLE SCHOLAR


112 L. T. Vu, R. Bowser, Fluid-based biomarkers for amyotrophic lateral sclerosis. Neurotherapeutics 14, 119–134 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

113 B. Vafadar-Isfahani, G. Ball, C. Coveney, C. Lemetre, D. Boocock, L. Minthon, O. Hansson, A. K. Miles, S. M. Janciauskiene, D. Warden, A. D.
Smith, G. Wilcock, N. Kalsheker, R. Rees, B. Matharoo-Ball, K. Morgan, Identification of SPARC-like 1 protein as part of a biomarker panel
for Alzheimer’s disease in cerebrospinal fluid. J. Alzheimers Dis. 28, 625–636 (2012).
CROSSREF • PUBMED • GOOGLE SCHOLAR

114 F. Mo, X. Ma, X. Liu, R. Zhou, Y. Zhao, H. Zhou, Altered CSF proteomic profiling of paediatric acute lymphocytic leukemia patients with
CNS infiltration. J. Oncol. 2019, 3283629 (2019).
CROSSREF • PUBMED • GOOGLE SCHOLAR

115 E. Agah, A. Zardoui, A. Saghazadeh, M. Ahmadi, A. Tafakhori, N. Rezaei, Osteopontin (OPN) as a CSF and blood biomarker for multiple
sclerosis: A systematic review and meta-analysis. PLOS ONE 13, e0190252 (2018).
CROSSREF • PUBMED • GOOGLE SCHOLAR

116 S. Futakawa, K. Nara, M. Miyajima, A. Kuno, H. Ito, H. Kaji, K. Shirotani, T. Honda, Y. Tohyama, K. Hoshi, Y. Hanzawa, S. Kitazume, R.
Imamaki, K. Furukawa, K. Tasaki, H. Arai, T. Yuasa, M. Abe, H. Arai, H. Narimatsu, Y. Hashimoto, A unique N-glycan on human transferrin
in CSF: A possible biomarker for iNPH. Neurobiol. Aging 33, 1807–1815 (2012).
CROSSREF • PUBMED • GOOGLE SCHOLAR

117 A. Singh, A. J. Beveridge, N. Singh, Decreased CSF transferrin in sCJD: A potential pre-mortem diagnostic test for prion disorders. PLOS
ONE 6, e16804 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

118 M. Bjerke, H. Zetterberg, Å. Edman, K. Blennow, A. Wallin, U. Andreasson, Cerebrospinal fluid matrix metalloproteinases and tissue in-
hibitor of metalloproteinases in combination with subcortical and cortical biomarkers in vascular dementia and Alzheimer’s disease. J.
Alzheimers Dis. 27, 665–676 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

119 F. H. Duits, M. Hernandez-Guillamon, J. Montaner, J. D. C. Goos, A. Montañola, M. P. Wattjes, F. Barkhof, P. Scheltens, C. E. Teunissen, W.

https://www.science.org/doi/10.1126/science.aaz5626 11/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

M. van der Flier, Matrix metalloproteinases in Alzheimer’s disease and concurrent cerebral microbleeds. J. Alzheimers Dis. 48, 711–720
(2015).
CROSSREF • PUBMED • GOOGLE SCHOLAR

120 F. Streijger, M. A. Skinnider, J. C. Rogalski, R. Balshaw, C. P. Shannon, A. Prudova, L. Belanger, L. Ritchie, A. Tsang, S. Christie, S. Parent, J.-
M. Mac-Thiong, C. Bailey, J. Urquhart, T. Ailon, S. Paquette, M. Boyd, J. Street, C. G. Fisher, M. F. Dvorak, C. H. Borchers, L. J. Foster, B. K.
Kwon, A targeted proteomics analysis of cerebrospinal fluid after acute human spinal cord injury. J. Neurotrauma 34, 2054–2068 (2017).
CROSSREF • PUBMED • GOOGLE SCHOLAR

121 K. Schultz, K. Nilsson, J. E. Nielsen, S. G. Lindquist, L. E. Hjermind, B. B. Andersen, A. Wallin, C. Nilsson, A. Petersén, Transthyretin as a
potential CSF biomarker for Alzheimer’s disease and dementia with Lewy bodies: Effects of treatment with cholinesterase inhibitors. Eur. J.
Neurol. 17, 456–460 (2010).
CROSSREF • PUBMED • GOOGLE SCHOLAR

122 S. Ranganathan, E. Williams, P. Ganchev, V. Gopalakrishnan, D. Lacomis, L. Urbinelli, K. Newhall, M. E. Cudkowicz, R. H. Brown Jr., R.
Bowser, Proteomic profiling of cerebrospinal fluid identifies biomarkers for amyotrophic lateral sclerosis. J. Neurochem. 95, 1461–1471
(2005).
CROSSREF • PUBMED • GOOGLE SCHOLAR

123 S. B. Lewis, R. Wolper, Y. Y. Chi, L. Miralia, Y. Wang, C. Yang, G. Shaw, Identification and preliminary characterization of ubiquitin C termi-
nal hydrolase 1 (UCHL1) as a biomarker of neuronal loss in aneurysmal subarachnoid hemorrhage. J. Neurosci. Res. 88, 1475–1484 (2010).
CROSSREF • PUBMED • GOOGLE SCHOLAR

124 R. Dobson, J. Topping, A. Davis, E. Thompson, G. Giovannoni, Cerebrospinal fluid and urinary biomarkers in multiple sclerosis. Acta
Neurol. Scand. 128, 321–327 (2013).
CROSSREF • PUBMED • GOOGLE SCHOLAR 

125 Z. Guo, Y. Zhang, L. Zou, D. Wang, C. Shao, Y. Wang, W. Sun, L. Zhang, A proteomic analysis of individual and gender variations in normal

human urine and cerebrospinal fluid using iTRAQ quantification. PLOS ONE 10, e0133270 (2015).

CROSSREF • PUBMED • GOOGLE SCHOLAR


126 T. Ruland, J. Wolbert, M. G. Gottschalk, S. König, A. Schulte-Mecklenbeck, J. Minnerup, S. G. Meuth, C. C. Groß, H. Wiendl, G. Meyer Zu
Hörste, Cerebrospinal fluid concentrations of neuronal proteins are reduced in primary angiitis of the central nervous system. Front.
Neurol. 9, 407 (2018).

CROSSREF • PUBMED • GOOGLE SCHOLAR

127 D. Morales, R. Hechavarria, V. Wojna, S. F. Acevedo, YWHAE/14-3-3ε: A potential novel genetic risk factor and CSF biomarker for HIV neu-
rocognitive impairment. J. Neurovirol. 19, 471–478 (2013).
CROSSREF • PUBMED • GOOGLE SCHOLAR

128 J. M. Lubieniecka, F. Streijger, J. H. T. Lee, N. Stoynov, J. Liu, R. Mottus, T. Pfeifer, B. K. Kwon, J. R. Coorssen, L. J. Foster, T. A. Grigliatti, W.
Tetzlaff, Biomarkers for severity of spinal cord injury in the cerebrospinal fluid of rats. PLOS ONE 6, e19247 (2011).
CROSSREF • PUBMED • GOOGLE SCHOLAR

eLetters (0)

No eLetters have been published for this article yet.

eLetters is an online forum for ongoing peer review. Submission of eLetters are open to all. eLetters are not edited, proofread, or
indexed. Please read our Terms of Service before submitting your own eLetter.

SUBMIT A RESPONSE TO THIS ARTICLE

CURRENT ISSUE
Social evolution in mammals
BY
TIM CLUTTON-BROCK

https://www.science.org/doi/10.1126/science.aaz5626 12/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

Serum amyloid A delivers retinol to intestinal myeloid cells to promote


adaptive immunity
BY
YE-JI BANG,
ZEHAN HU,
ET AL.

Immune correlates of protection by mRNA-1273 vaccine against SARS-


CoV-2 in nonhuman primates
BY
KIZZMEKIA S. CORBETT,
MARTHA C. NASON,
ET AL.

TABLE OF CONTENTS 

LATEST NEWS
ADVERTISING FEATURE 17 SEP 2021 SCIENCESHOTS 17 SEP 2021
Post-pandemic: The power of crisis manage- A new fleet of Moon landers will set sail next
ment planning year, backed by private companies

SCIENCEINSIDER 17 SEP 2021 NEWS FEATURE 16 SEP 2021

Peru’s new president is controversial. Here’s why A huge forest experiment aims to reduce wild-
scientists have high hopes for him fires. Can it unite loggers and environmental-
ists?

NEWS 16 SEP 2021 SIFTER 16 SEP 2021


Biden’s vaccine mandates, bad Facebook data, Australian bushfires had widespread impacts
 and a doctored portrait across the globe 


RECOMMENDED 
RESEARCH ARTICLES NOVEMBER 2017

Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells

RESEARCH ARTICLES MARCH 2019

Human pluripotent stem cell–derived brain pericyte–like cells induce blood-brain barrier properties

PERSPECTIVES JANUARY 2019

Assembling human brain organoids

FOLLOW US

     󡀆

NEWS CAREERS COMMENTARY JOURNALS AUTHORS & REVIEWERS

All News Careers Articles Opinion Science Information for Authors

ScienceInsider Find Jobs Analysis Science Advances Information for Reviewers

News Features Employer Profiles Blogs Science Immunology

Science Robotics

Science Signaling

Science Translational Medicine

Science Partner Journals

LIBRARIANS ADVERTISERS RELATED SITES ABOUT US HELP

FAQs
https://www.science.org/doi/10.1126/science.aaz5626 13/14
20/9/21 12:10 Human CNS barrier-forming organoids with cerebrospinal fluid production

Manage Your Institutional Advertising Kits


Custom Publishing Info AAAS.org
AAAS Communities Leadership
Work at AAAS Access and Subscriptions
Subscription
Library Admin Portal Post a Job EurekAlert! Prizes and Awards Order a Single Issue
Request a Quote Science in the Classroom Reprints and Permissions
Librarian FAQs Contact Us

© 2021 American Association for the Advancement of Science. All rights reserved. AAAS is a partner of HINARI, AGORA, OARE, CHORUS, CLOCKSS, CrossRef and COUNTER. Science ISSN
0036-8075.

Terms of Service
|
Privacy Policy
|
Accessibility

 





https://www.science.org/doi/10.1126/science.aaz5626 14/14

You might also like