Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Biological

Psychiatry Review

Transplantation Strategies to Enhance Maturity


and Cellular Complexity in Brain Organoids
Meiyan Wang, Fred H. Gage, and Simon T. Schafer

ABSTRACT
Human brain organoids are 3-dimensional cell aggregates that are generated from pluripotent stem cells and reca-
pitulate features of the early developing human brain. Brain organoids mainly consist of cells from the neural lineage,
such as neural progenitor cells, neurons, and astrocytes. However, current brain organoid systems lack functional
vasculature as well as other non-neuronal cells that are indispensable for oxygen and nutrient supply to the orga-
noids, causing cell stress and formation of a necrotic center. Attempts to utilize intracerebral transplantation ap-
proaches have demonstrated successful vascularization of brain organoids and robust neurodifferentiation. In this
review, we summarize recent progress and discuss ethical considerations in the field of brain organoid
transplantation.
https://doi.org/10.1016/j.biopsych.2023.01.004

Brain organoids are self-organizing 3-dimensional cultures of Another study explored a specific xenotransplantation
mainly neuroectodermal origin that resemble features of the strategy, by which human brain organoids were transplanted
developing human brain. These culture systems are usually into highly vascularized brain regions of an animal host to
derived from stem cells, such as embryonic stem cells (ESCs) vascularize the developing organoid tissue. In a recent study,
or induced pluripotent stem cells (iPSCs), that have the po- Mansour et al. (7) transplanted cerebral organoids derived from
tential to differentiate into any type of cell of the human body. human ESCs into the retrosplenial cortex of immunodeficient
The fact that brain organoids can be generated from iPSCs mice and reported highly efficient infiltration with blood vessels
derived from human subjects presents the unprecedented and the presence of functional circulation within the organoid
opportunity for using these models to study process dynamics grafts.
pertaining to the unique characteristics of human brain There are several benefits for organoid models to feature an
development and human-specific brain disorders. Brain orga- active and physiologically perfused vasculature. One advan-
noid models have been successfully used in various applica- tage is prevention of necrosis and cell death that severely
tions (1–4). However, despite a high correlation of cellular compromise the integrity and long-term culture of organoid
diversification between brain organoid models and the devel- models. Another benefit is that it supplies the organoids with
oping human brain (5), current protocols still face a number of nutrients that are otherwise missing from current in vitro par-
limitations, including incomplete maturation and cellular stress. adigms. The presence of a vascular system within brain
Of note, a recent study (6) demonstrated that cell stress is organoids provides great value, not only because it supplies
limited to a defined subpopulation of cells in the organoids and nutrients but also because it may enable faithful modeling of
does not affect neuronal specification or maturation. Never- the invasion/extravasation and interaction with different non-
theless, the lack of a microvasculature system results in low neuronal cell types.
oxygen levels and reduced nutrient supply to the center of the In this review, we focus on the benefits and purpose of
organoid, rendering the organoid’s core prone to variable de- transplantation strategies to incorporate functional vascular
grees of necrosis (7,8). systems and to allow the generation of more complex organoid
To overcome these limitations, various approaches to allow models for the study of cellular phenotypes underlying in vivo–
vascularization of brain organoids are the subjects of active like physiology.
research, including in vitro co-culture strategies using vascular
cells (9,10) or fusion to blood vessel organoids (11,12), the NEURAL TRANSPLANTS OR NEURAL CHIMERAS?
modification of culture conditions to generate vascular cells Neural and non-neuronal cells can be transplanted directly into
(13), genome-editing technologies to ectopically induce endo- the brain of a nonhuman animal during development or
thelial cells (14), and “organoid-on-a-chip” microfluidic prepa- adulthood to allow the investigation of these cells under quasi-
rations (15). Interestingly, some of these studies reported that physiological conditions (Figure 1A). As an example, trans-
the formation and/or presence of vascular-like structures in planted neural precursor cells can mature into functional
brain organoids resulted in attenuated apoptosis and acceler- neurons and integrate into the host nervous system to form
ated neuronal maturation, even in the absence of blood flow. functional connections. The grafted single neuronal cells are

616 ª 2023 Society of Biological Psychiatry.


Biological Psychiatry April 1, 2023; 93:616–621 www.sobp.org/journal ISSN: 0006-3223
Biological
Brain Organoid Transplantation Psychiatry

Figure 1. Schematic illustrating the distinction


between neural chimeras and neural transplants. (A)
Neural and glial cells or brain organoids can be
directly transplanted into the brain of a fetal or adult
animal host. (B) Neural chimeras, in contrast, are a
special form of transplant in which donor cells are
introduced into the host prior to gastrulation.

capable of receiving synaptic input from and providing syn- could be observed after 7 to 10 days in vivo; extensive
aptic input to host neurons. Different types of glia have also vascularization was observed after 14 days after engraftment
been shown to be able to integrate into and interact with host (Figure 2) (7). Importantly, using two-photon microscopy, the
cells following transplantation. A special and new form of authors were able to demonstrate that the organoid-infiltrating
neural transplantation is the concept of brain organoid trans- blood vessels showed robust blood flow. The survival of the
plantation, in which whole-brain organoids grown up to a organoid transplants was highly dependent on successful
certain stage in vitro are transplanted into an animal host to vascularization (7); very limited cell death and decreased
allow functional integration and adequate blood supply for cellular stress were observed in brain organoids in vivo (7,21).
extended periods of time (Figure 1A). In line with this finding, a recent study showed that cellular
Human neural chimeras, in contrast, are a special form of stress pathways ectopically activated in cortical organoids
transplant in which donor cells are introduced into a nonhuman in vitro could impair the proper specification of cellular sub-
host prior to gastrulation (16). Here, human donor cells inter- types and maturation of neural progenitors (22). When trans-
mingle with host cells and populate the nervous system from planted into the cortices of immunocompromised mouse pups,
the onset of neurogenesis to develop side-by-side within the dissociated organoids exhibited increased levels of cell sub-
host, enabling some level of integration (Figure 1B). Particular type specification in both outer radial glia and newborn neu-
cell types of the host may be genetically ablated, thus rons. Although the group only analyzed dissociated organoid
permitting donor cells to contribute to forming a tissue or or- cells in vivo (22), the results suggest that the transplantation
gan. An example of such an approach is blastocyst comple- was—at least in part—sufficient to alleviate the cell stress and
mentation (Figure 1B) (17). Extensive contribution of donor subtype defects observed in vitro. Nevertheless, a more
cells to an animal host’s forebrain has been reported following detailed analysis is needed to assess the differences between
neural blastocyst complementation in mice (17). dissociated and whole organoid grafts in vivo. It is important to
The distinction between neural cell or organoid transplants highlight another study that reported enhanced survival and a
and neural chimeras is vaguely defined and largely based on higher degree of vascularization in cerebral organoid grafts as
whether the cells introduced into the host animal remain compared with transplants using dissociated neural progenitor
limited to the nervous system or contribute to other organs as cells (23). Engrafted organoids featured an increased number
well. In a recent consensus study report, the National Academy of neurons and showed progressive transition from the
of Sciences suggested that a useful dividing line could be the
gastrula stage of embryogenesis, because many scientists in
the field view the introduction of exogenous cells to an or-
ganism prior to gastrulation as generating chimeras and after
gastrulation as generating transplants (16). Of note, most chi-
meras are generated using donors from the same species. The
formation of human neural chimeras in mice has not been re-
ported to date.

BRAIN ORGANOID TRANSPLANTATIONS


The first studies that attempted to induce vascularization of
brain organoids in vivo took advantage of an intracerebral
transplantation technique originally developed for trans-
planting fetal or adult tissue into the rodent brain (18–20). This
technique allowed sufficient infiltration of the transplanted Figure 2. Schematic illustrating vascularization of brain organoids in vivo,
brain organoids by the vasculature system of the host, which following engraftment.

Biological Psychiatry April 1, 2023; 93:616–621 www.sobp.org/journal 617


Biological
Psychiatry Brain Organoid Transplantation

progenitor-dominated, rosette-rich organoid tissue to a more damaged brain environment and the potential for controlling is
mature tissue that predominantly consisted of neurons and warranted.
astrocytes in vivo (7). Further research is needed to assess the
intrinsic and environmental drivers that permit sufficient
vascularization of brain organoids in vivo. Alternatively, intro- ETHICAL CONSIDERATIONS FOR HUMAN NEURAL
duction of human vasculature prior to transplantation appears TRANSPLANTS AND CHIMERAS
to be a promising strategy to potentially replace the host While advancements in brain organoid research have made
vasculature with human cells. Although not sufficiently them an ever more valuable tool for clinicians and scientists,
explored, in vitro vascularized brain organoids displayed some the very features that make them more similar to the complex
level of functional perfusion by the animal host on trans- human brain have raised some ethical concerns. This is, in
plantation (9,14). part, based on the use of human materials and the concern
Neuronal network activity in the engrafted organoids has that transplanting humanlike tissue into an animal host may
been assessed using different approaches. Longitudinal result in enhancement of brain functions due to partial hu-
measurements of AAV8-CaMKII-jRGECO1a–labeled neurons manization of the host.
in the organoid grafts by two-photon calcium imaging revealed The National Academy of Sciences has recently released a
recurrent and spontaneous rhythmic calcium transients (7). consensus study report from the Committee on Ethical, Legal,
Moreover, extracellular recordings using multielectrode arrays and Regulatory Issues Associated with Neural Chimeras and
identified neuronal action potentials and synchronized Organoids to address ethical concerns and to summarize as-
neuronal network activity (7). Cross-correlation analysis of pects related to governance and oversight of research with
neuronal activity in the same recording sites revealed a more human-animal chimeras (16). According to this report, the is-
active and coordinated network in late-stage grafts, suggest- sues specific to this form of transplantation include concerns
ing progressive neuronal maturation (7). An independent study about general animal welfare, about the distinctions between
used whole-cell patch-clamp recordings and showed that human beings and animals, and about enhancement of brain
human glutamatergic neurons indeed acquired electrophysio- functions and consciousness.
logical properties indicative of progressive cellular maturation There are a range of positions on these important issues
that occurred within the graft over time, following trans- that require discussion among people with different perspec-
plantation (24). Together, these data suggest robust neuro- tives. Thus, the research on human transplants and chimeras is
differentiation potential and extended maturation of human subject to a wide range of national and international oversight
brain organoids in vivo. mechanisms that regulate various aspects of this field. These
Extensive axonal growth of organoid grafts and long- include, but are not limited to, a collection of federal and state
distance axonal projections from the grafts to distant targets laws and regulations, professional society guidelines (e.g., In-
of the host brain have been reported. Human brain organoids ternational Society for Stem Cell Research and Guidelines for
extended axons through the cortical layers and the corpus Stem Cell Research and Clinical Translation), nonbinding
callosum (7), along the corticospinal tract (25), and to the basal consensus studies by scientific academies (e.g., National
brain regions (24). Viral tracing and optogenetics have identi- Academy of Sciences), and various conference reports. The
fied synaptic connections between human cerebral organoids legal requirements pertaining to the use of human tissues and
and mouse host neurons (7,24). Interestingly, in response to stem cells and the use and welfare of nonhuman animals are
environmental stimuli, neurons in organoid grafts demon- often implemented by research oversight committees at an
strated state-dependent firing changes (7). Mice with cerebral institutional level. In general, if research in any biotechnological
organoid grafts have also been shown to feature increased area raises substantial ethical and regulatory concerns, a
startle fear responses to auditory conditioned stimuli (24). 3-tiered oversight structure has been proposed and recom-
These data suggest that human brain organoids can integrate mended by many National Academies committees for the
and form functional circuits with the host brain. guidance of institutional committees. Most of the research
Other studies have also reported improved lesion repair involving human neural transplants falls into the first-tier
through organoid grafting in mouse and rat models of brain category because it presents no new ethical or regulatory
injuries (26–28). In a rat model of traumatic brain injury, concerns beyond those that the current oversight committee is
engrafted cortical organoids supported reconstruction of charged with addressing. Research that falls into the second
damaged motor cortex and led to improved neurologic motor tier can only proceed after additional review and approval
function and reduced brain damage (28). Similarly, in a cerebral because it may raise additional ethical concerns beyond what
artery occlusion model of stroke, engrafted cortical organoids the current oversight system can address (protection of human
supported motor cortex region–specific reconstruction in rats subjects and animal welfare). As an example, research that
(27). In both models, cells from engrafted cortical organoids renders a brain more humanlike, particularly when trans-
displayed extensive migration into different brain regions along planting human cells into the brains of nonhuman primates,
the corpus callosum (27,28). Using a controlled cortical impact may fall into this category. The third-tier category encom-
model for experimental brain injury, increased vascularization passes research that should not be permitted at present; this
and reduced glial scarring in the lesion area were observed includes experiments that are currently forbidden under federal
following brain organoid transplantation, suggesting that the law, such as the introduction of human ESCs or iPSCs into the
presence of the organoid graft was able to promote neural blastocyst of a nonhuman animal.
repair (26). Despite these promising findings, additional As indicated above, the institutional oversight committees
research on the effects that brain organoid grafts exert on the for human neural transplant and chimera research have

618 Biological Psychiatry April 1, 2023; 93:616–621 www.sobp.org/journal


Biological
Brain Organoid Transplantation Psychiatry

specific regulatory charges to ensure the protection of human originate from the yolk sac in vivo. The variability of microglia-
subjects and animal welfare. If an enhancement of brain like cells within cerebral organoids and the lack of a complete
functions or the acquisition of new capacities is suspected in recapitulation of the resting ramified state observed in vivo
animals used as a host, additional training to determine which warrant further investigation.
conditions and environments are preferred by the animals may As highlighted in this review, transplantation strategies can
be required for the evaluators. Veterinary evaluations may be provide an alternative tool for investigating human cells under
required to include ongoing monitoring for changes in specific in vivo–like conditions. However, concerns about the contri-
behaviors (e.g., activity, feeding, and socializing) and assess- bution of host cells to the human graft should be considered
ment of physiological parameters (e.g., blood pressure, cortisol when generating these models because they could potentially
level, and heart rate) to ensure animal welfare. alter the system or cell type under investigation. Thus, forward
As the research on neural transplants and chimeras de- engineering of ever more sophisticated human organoid
velops, further discussion of oversight is needed to address models would provide a path toward investigating the inter-
the ethical concerns that do not fall within the scope or action between different human non-neuronal and neural cells
expertise of the existing committees, offering opportunities for under in vivo–like conditions. Previous studies have shown
much broader discussions on a national and international level. that human glial progenitor cells can be transplanted and that
they generate functional astrocytes when transplanted
neonatally into murine hosts [e.g., (36)]. Neonatal engraftment
OUTLOOK of human glial progenitor cells allows differentiation into both
Studying prenatal human brain development remains a chal- astrocytes and myelinogenic oligodendrocytes in a hypo-
lenge due to limited access to live human brain tissue and myelinated environment (37–39). Engrafted human glial pro-
ethical constraints on research using human embryos. Brain genitor cells may even outcompete the resident glial
organoids offer a promising avenue to overcome these limi- progenitors and dominate the host brain (36,37,39). Using such
tations by allowing researchers to study human brain devel- human glial transplants revealed species-specific glial prop-
opment directly in vitro. Great progress has been made in the erties (36) as well as disease-associated changes in astrocyte
last decade to model human disorders in a dish, but challenges and oligodendrocyte differentiation (37,39). Similarly, trans-
remain that need to be overcome when attempting to translate plantation of ESC/iPSC-derived microglial or hematopoietic
clinical observations made in vivo to molecular and cellular progenitors into neonatal or postnatal mouse brains resulted in
phenotypes observed in vitro. a context-dependent differentiation of microglia (40–43).
Recent studies that performed similar organoid xeno- However, generating these glial transplant models required
transplantation experiments in mice (23) and rats (29) expression of human-specific factors such as CSF1 (colony-
demonstrated that the cellular and molecular state of maturity stimulating factor 1), which was achieved by utilizing mice
was enhanced as compared with in vitro organoids. Trans- that harbored a knockin of human CSF1 into the murine locus
planted neurons displayed increased morphological (40,41,43) or constitutive CSF1 overexpression (42). The ESC/
complexity and synaptic properties as compared with their iPSC-derived microglia acquired complex morphologies spe-
in vitro counterparts. However, further work is needed to cific to microglia and gained gene expression signatures
disentangle the exact mechanisms that support organoid shown to be induced by the in vivo brain environment (40–43).
maturation and complexity in vivo. Moreover, it remains to be Collectively, these data suggest that engrafted human glial
explored whether the increased maturity would reveal molec- cells are functional in the mouse brain. In the future, grafting
ular and cellular changes in complex neuropsychiatric and human brain organoids that contain a diverse set of resident
neurological disorders that are otherwise difficult to investigate human glial cells may provide an in vivo environment ideal for
in other cell models. studying human neuron-glia interactions.
While current brain organoids allow us to recapitulate, in A more complete in vivo brain organoid model offers many
particular, those developmental processes that emerge from advantages over current in vitro protocols regarding the
cells of the neuroectoderm, they still lack other cell types that feasibility of investigating long-term phenotypes. We advocate
are of non-ectodermal origin. Incorporation of cell types that, rather than replacing existing models, future research
forming the neurovascular unit and blood vessels (14,30) are should focus on developing complementary strategies. It will
forthcoming approaches to increase the cellular composition be the combination of both purposeful reduction and more
of these models. Additionally, most brain organoid systems authentic, complex model systems that will provide the means
lack microglial cells and, in most cases, they develop with to start disentangling the genetic, molecular, and cellular
limited or underrepresented amounts of oligodendrocytes and complexity underlying human disorders.
astrocytes. Moreover, brain organoid models that allow
specification of anatomically defined subclasses of human
astrocytes have yet to be developed. It is also worth noting ACKNOWLEDGMENTS AND DISCLOSURES
that cerebral organoids generated through unguided ap- This work was supported by the American Heart Association and the Paul G.
proaches can spontaneously develop microglia-like cells that Allen Frontiers Group (Grant No. 19PABHI34610000 [to FHG]), National
possibly emerge from mesodermal contributions within these Institute of Mental Health (Grant No. U19-MH106434 [to FHG]), JPB Foun-
dation, Annette C. Merle-Smith, Robert andMary Jane Engman Foundation,
structures (31). Additionally, short-term co-culture experiments
Lynn and Edward Streim, and the Ray and Dagmar Dolby Family Fund (to
have demonstrated that the incorporation of microglia-like FHG). MW is supported by a NARSAD Young Investigator Grant from the
cells into organoids appears feasible (32–35). However, it is Brain and Behavior Research Foundation. STS is supported by a NARSAD
not known whether these cells are similar to the ones that Young Investigator Grant from the Brain and Behavior Research Foundation

Biological Psychiatry April 1, 2023; 93:616–621 www.sobp.org/journal 619


Biological
Psychiatry Brain Organoid Transplantation

and a priority program grant from the German Research Foundation (DFG Chimeras: Science, Ethics, and Governance. Washington, DC: The
SPP 2395 [to STS]) under award No. 500300695. National Academies Press. Available at: https://doi.org/10.17226/26
We thank members of the Gage Lab for discussions, M.L. Gage for 078.
editorial suggestions and B. Coyne for administrative assistance. 17. Chang AN, Liang Z, Dai HQ, Chapdelaine-Williams AM, Andrews N,
Figures were created on BioRender.com. Bronson RT, et al. (2018): Neural blastocyst complementation enables
The authors report no biomedical financial interests or potential conflicts mouse forebrain organogenesis. Nature 563:126–130.
of interest. 18. Gage FH, Fisher LJ (1991): Intracerebral grafting: A tool for the
neurobiologist. Neuron 6:1–12.
19. Gage FH, Björklund A, Stenevi U (1984): Denervation releases a
ARTICLE INFORMATION neuronal survival factor in adult rat hippocampus. Nature 308:637–
From the Laboratory of Genetics, The Salk Institute for Biological Studies, La 639.
Jolla, California (MW, FHG, STS); Department of Psychiatry and Psycho- 20. Stenevi U, Björklund A, Svendgaard NA (1976): Transplantation of
therapy, Medical School, Technical University of Munich, Munich, Germany central and peripheral monoamine neurons to the adult rat brain:
(STS); and Center for Organoid Systems and Tissue Engineering, Technical Techniques and conditions for survival. Brain Res 114:1–20.
University of Munich, Garching, Germany (STS). 21. Huang S, Huang F, Zhang H, Yang Y, Lu J, Chen J, et al. (2022): In vivo
Address correspondence to Simon T. Schafer, Ph.D., at simon.schafer@ development and single-cell transcriptome profiling of human brain
tum.de, or Meiyan Wang, Ph.D., at mwang@salk.edu. organoids. Cell Prolif 55:e13201.
Received Jun 15, 2022; revised Nov 23, 2022; accepted Jan 9, 2023. 22. Bhaduri A, Andrews MG, Mancia Leon W, Jung D, Shin D, Allen D,
et al. (2020): Cell stress in cortical organoids impairs molecular sub-
type specification. Nature 578:142–148.
23. Daviaud N, Friedel RH, Zou H (2018): Vascularization and engraftment
REFERENCES of transplanted human cerebral organoids in mouse cortex. eNeuro 5:
1. Chiaradia I, Lancaster MA (2020): Brain organoids for the study of ENEURO.0219-18.2018.
human neurobiology at the interface of in vitro and in vivo. Nat Neu- 24. Dong X, Xu SB, Chen X, Tao M, Tang XY, Fang KH, et al. (2021):
rosci 23:1496–1508. Human cerebral organoids establish subcortical projections in the
2. Mansour AA, Schafer ST, Gage FH (2021): Cellular complexity in brain mouse brain after transplantation. Mol Psychiatry 26:2964–2976.
organoids: Current progress and unsolved issues. Semin Cell Dev Biol 25. Kitahara T, Sakaguchi H, Morizane A, Kikuchi T, Miyamoto S,
111:32–39. Takahashi J (2020): Axonal extensions along corticospinal tracts from
3. Qian X, Song H, Ming GL (2019): Brain organoids: Advances, appli- transplanted human cerebral organoids. Stem Cell Reports 15:467–
cations and challenges. Development 146:dev166074. 481.
4. Zhang DY, Song H, Ming GL (2021): Modeling neurological disorders 26. Bao Z, Fang K, Miao Z, Li C, Yang C, Yu Q, et al. (2021): Human ce-
using brain organoids. Semin Cell Dev Biol 111:4–14. rebral organoid implantation alleviated the neurological deficits of
5. Uzquiano A, Kedaigle AJ, Pigoni M, Paulsen B, Adiconis X, Kim K, traumatic brain injury in mice. Oxid Med Cell Longev 2021:6338722.
et al. (2022): Single-cell multiomics atlas of organoid development 27. Wang SN, Wang Z, Xu TY, Cheng MH, Li WL, Miao CY (2020): Cerebral
uncovers longitudinal molecular programs of cellular diversification of organoids repair ischemic stroke brain injury. Transl Stroke Res
the human cerebral cortex. bioRxiv https://doi.org/10.1101/2022.03. 11:983–1000.
17.484798. 28. Wang Z, Wang SN, Xu TY, Hong C, Cheng MH, Zhu PX, et al. (2020):
6. Vértesy Á., Eichmüller OL, Naas J, Novatchkova M, Esk C, Balmaña M, Cerebral organoids transplantation improves neurological motor
et al. (2022): Gruffi: An algorithm for computational removal of stressed function in rat brain injury. CNS Neurosci Ther 26:682–697.
cells from brain organoid transcriptomic datasets. EMBO J 29. Revah O, Gore F, Kelley KW, Andersen J, Sakai N, Chen X, et al.
41:e111118. (2022): Maturation and circuit integration of transplanted human
7. Mansour AA, Gonçalves JT, Bloyd CW, Li H, Fernandes S, Quang D, cortical organoids. Nature 610:319–326.
et al. (2018): An in vivo model of functional and vascularized human 30. Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T,
brain organoids. Nat Biotechnol 36:432–441. et al. (2019): Flow-enhanced vascularization and maturation of kidney
8. Qian X, Su Y, Adam CD, Deutschmann AU, Pather SR, Goldberg EM, organoids in vitro. Nat Methods 16:255–262.
et al. (2020): Sliced human cortical organoids for modeling distinct 31. Ormel PR, Vieira de Sá R, van Bodegraven EJ, Karst H, Harschnitz O,
cortical layer formation. Cell Stem Cell 26:766–781.e9. Sneeboer MAM, et al. (2018): Microglia innately develop within cere-
9. Pham MT, Pollock KM, Rose MD, Cary WA, Stewart HR, Zhou P, et al. bral organoids. Nat Commun 9:4167.
(2018): Generation of human vascularized brain organoids. Neuro- 32. Abud EM, Ramirez RN, Martinez ES, Healy LM, Nguyen CHH,
report 29:588–593. Newman SA, et al. (2017): iPSC-derived human microglia-like cells to
10. Shi Y, Sun L, Wang M, Liu J, Zhong S, Li R, et al. (2020): Vascularized study neurological diseases. Neuron 94:278–293.e9.
human cortical organoids (vOrganoids) model cortical development 33. Abreu CM, Gama L, Krasemann S, Chesnut M, Odwin-Dacosta S,
in vivo. PLoS Biol 18:e3000705. Hogberg HT, et al. (2018): Microglia increase inflammatory re-
11. Ahn Y, An JH, Yang HJ, Lee DG, Kim J, Koh H, et al. (2021): Human sponses in iPSC-derived human BrainSpheres. Front Microbiol
blood vessel organoids penetrate human cerebral organoids and form 9:2766.
a vessel-like system. Cells 10:2036. 34. Lin YT, Seo J, Gao F, Feldman HM, Wen HL, Penney J, et al. (2018):
12. Sun XY, Ju XC, Li Y, Zeng PM, Wu J, Zhou YY, et al. (2022): Generation APOE4 causes widespread molecular and cellular alterations associ-
of vascularized brain organoids to study neurovascular interactions. ated with Alzheimer’s disease phenotypes in human iPSC-derived
eLife 11:e76707. brain cell types [published correction appears in Neuron 2018;98:
13. Ham O, Jin YB, Kim J, Lee MO (2020): Blood vessel formation in ce- 1294]. Neuron 98:1141–1154.e7.
rebral organoids formed from human embryonic stem cells. Biochem 35. Muffat J, Li Y, Omer A, Durbin A, Bosch I, Bakiasi G, et al. (2018):
Biophys Res Commun 521:84–90. Human induced pluripotent stem cell-derived glial cells and neural
14. Cakir B, Xiang Y, Tanaka Y, Kural MH, Parent M, Kang YJ, et al. (2019): progenitors display divergent responses to Zika and dengue in-
Engineering of human brain organoids with a functional vascular-like fections. Proc Natl Acad Sci USA 115:7117–7122.
system. Nat Methods 16:1169–1175. 36. Han X, Chen M, Wang F, Windrem M, Wang S, Shanz S, et al. (2013):
15. Salmon I, Grebenyuk S, Abdel Fattah AR, Rustandi G, Pilkington T, Forebrain engraftment by human glial progenitor cells enhances
Verfaillie C, Ranga A (2022): Engineering neurovascular organoids with synaptic plasticity and learning in adult mice. Cell Stem Cell 12:342–
3D printed microfluidic chips. Lab Chip 22:1615–1629. 353.
16. National Academies of Sciences, Engineering, and Medicine (2021): 37. Osipovitch M, Asenjo Martinez A, Mariani JN, Cornwell A, Dhaliwal S,
The Emerging Field of Human Neural Organoids, Transplants, and Zou L, et al. (2019): Human ESC-derived chimeric mouse models of

620 Biological Psychiatry April 1, 2023; 93:616–621 www.sobp.org/journal


Biological
Brain Organoid Transplantation Psychiatry

Huntington’s disease reveal cell-intrinsic defects in glial progenitor cell 41. Mancuso R, Van Den Daele J, Fattorelli N, Wolfs L, Balusu S,
differentiation. Cell Stem Cell 24:107–122.e7. Burton O, et al. (2019): Stem-cell-derived human microglia trans-
38. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, Levine C, et al. planted in mouse brain to study human disease. Nat Neurosci
(2013): Human iPSC-derived oligodendrocyte progenitor cells can 22:2111–2116.
myelinate and rescue a mouse model of congenital hypomyelination. 42. Svoboda DS, Barrasa MI, Shu J, Rietjens R, Zhang S, Mitalipova M,
Cell Stem Cell 12:252–264. et al. (2019): Human iPSC-derived microglia assume a primary
39. Windrem MS, Osipovitch M, Liu Z, Bates J, Chandler-Militello D, microglia-like state after transplantation into the neonatal mouse brain.
Zou L, et al. (2017): Human iPSC glial mouse chimeras reveal glial Proc Natl Acad Sci USA 116:25293–25303.
contributions to schizophrenia. Cell Stem Cell 21:195–208.e6. 43. Xu R, Li X, Boreland AJ, Posyton A, Kwan K, Hart RP, Jiang P
40. Hasselmann J, Coburn MA, England W, Figueroa Velez DX, Kiani (2020): Human iPSC-derived mature microglia retain their identity
Shabestari S, Tu CH, et al. (2019): Development of a chimeric model to study and functionally integrate in the chimeric mouse brain. Nat Commun
and manipulate human microglia in vivo. Neuron 103:1016–1033.e10. 11:1577.

Biological Psychiatry April 1, 2023; 93:616–621 www.sobp.org/journal 621

You might also like