Selective localization of bone marrow-derived ramified cells in the brain adjacent to the attachments of choroid plexus

https://doi.org/10.1016/j.bbi.2012.12.010Get rights and content

Abstract

Although the immune system modulates higher functions of the brain under non-inflammatory conditions, how immune cells interact with brain parenchymal cells remains to be determined. Using bone marrow chimeric mice in which the recipients’ immune system was reconstituted by marrow cells derived from GFP-transgenic mice by syngeneic intra-bone marrow-bone marrow transplantation (IBM-BMT) and by intravenous (IV)-BMT, we examined the distribution, density and differentiation of donor-derived marrow cells in the brain parenchyma 2 weeks and 1, 4 and 8 months after BMT. Marrow-derived cells started to populate discrete brain regions from 1 to 4 months after BMT, exhibited ramified morphology and expressed Iba-1. The ramified marrow-derived cells were distributed in more brain regions and for a longer time after IBM-BMT than IV-BMT. Most of these discrete regions were adjacent to the attachments of choroid plexus that comprised thinned brain parenchyma consisting of astroglial processes in the narrow channel between the ependyma and pia. These specific portions of astroglial processes expressed fractalkine. In the choroid plexus stroma, not only Iba-1+ myeloid cells but also non-myeloid CXCL12-expressing cells were of bone marrow-origin. Transcripts of fractalkine, CXCL12 and their related molecules such as CX3CR1, ADAM10 and CXCR4 were detected in the tissue consisting of the choroid plexus, the attachments and adjacent brain parenchyma. Thus, bone marrow cells selectively enter the discrete brain regions adjacent to the attachments of choroid plexus and differentiate into ramified myeloid cells. Fractalkine in the attachments of choroid plexus and CXCL12 in the choroid plexus stroma may be involved in these brain–immune interactions.

Highlight

Bone marrow cells enter the selective brain regions adjacent to the attachments of choroid plexus and differentiate into ramified myeloid cells.

Introduction

It has been established that the immune system modulates the functional and behavioral processes of the central nervous system (CNS) (Yirmiya and Goshen, 2011). Exposure to pathogens stimulates the peripheral immune system and induces inflammatory responses, including the elevated expression of pro-inflammatory cytokines such as interleukin (IL)-1 (Derijk et al., 1991, Oppenheim et al., 1986), IL-6, and tumor necrosis factor (TNF)-α (Roth et al., 1993). Increased levels of pro-inflammatory cytokines in peripheral tissues induce the local synthesis of pro-inflammatory cytokines within the brain parenchyma (Ban et al., 1992, Fontana et al., 1984, Hagan et al., 1993), leading to behavioral alteration such as sickness behavior (Hart, 1988, Kent et al., 1992, Rothwell, 1991) and impaired learning (Yirmiya et al., 2002).

The immune system also plays an important role in maintaining the higher functions of the brain under non-inflammatory conditions. For example, severe combined immune deficiency (SCID) mice and nude mice that are deficient in mature T cells exhibit cognitive deficits and behavioral abnormalities that are remedied by T cell restoration (Kipnis et al., 2004). In addition, systemic depletion of CD4-positive T cells leads to reduced hippocampal neurogenesis and impaired learning in the Morris water maze (Wolf et al., 2009). T cells are not normally present in the brain parenchyma under non-inflammatory conditions. But during cognitive task performance, it is necessary that T cells accumulate in the meninges and express high levels of IL-4, which skews meningeal myeloid cells toward an anti-inflammatory phenotype (Derecki et al., 2010, Derecki et al., 2011). However, the question of whether peripheral immune cells located in the meningeal spaces interact with cells within the brain parenchyma via cell-to-cell contact and, if so, what is the site of interaction, remains.

One of the most powerful tools for addressing these questions is the bone marrow chimera in which the recipients’ immune system is reconstituted with donors’ labeled bone marrow cells (BMCs) by transplantation following irradiation, since this allows us to trace the distribution of labeled immune cells entering any organ. Studies using bone marrow chimeras to examine the brain distribution of bone marrow-derived cells have been reported since the late 80s’ (Davoust et al., 2008). These studies have consistently shown that bone marrow-derived cells under non-inflammatory conditions are primarily located in the leptomeninges, choroid plexus and perivascular spaces (Anandasabapathy et al., 2011, Chinnery et al., 2010, Hess et al., 2004, Hickey and Kimura, 1988, Soulas et al., 2009, Vallieres and Sawchenko, 2003). Bone marrow-derived cells also enter the circumventricular organs (CVOs), which contain a high density of capillaries lacking blood brain barrier (BBB) (Vallieres and Sawchenko, 2003). These bone marrow-derived cells preserve hematopoietic identity or express markers of the myeloid lineage. However, there has been some controversy over whether bone marrow-derived cells infiltrate the brain parenchyma protected by the complete BBB under non-inflammatory conditions. Some investigators have stated that, after bone marrow transplantation (BMT) by intravenous (IV) injection of donor cells, donor-derived cells were widely distributed throughout the brain parenchyma, and differentiated into microglia with a highly ramified morphology (Asheuer et al., 2004, Simard and Rivest, 2004). Others have claimed that the donors’ bone marrow-derived cells were found chiefly in association with blood vessels, and only rarely in the parenchyma of the brain (Hickey and Kimura, 1988, Soulas et al., 2009, Vallieres and Sawchenko, 2003). A recent study indicated that postnatal hematopoietic progenitors did not significantly contribute to microglia renewal or homeostasis in the adult brain (Ginhoux et al., 2010).

Meanwhile, a novel BMT procedure, namely intra-bone marrow (IBM)-BMT has been developed. In this procedure BMCs are collected from the marrow of the donors’ long bones by perfusion, and whole BMCs are injected directly into the bone marrow cavity of the recipients instead of being injected IV (Ikehara, 2003, Ikehara, 2011). The IBM procedure facilitates the efficacious and speedy transfer of not only hematopoietic stem cells (HSCs) but also mesenchymal stem cells (MSCs) from donor into recipient. When IBM-BMT is performed at the same time as the transplantation of heterotopic organs, it facilitates the induction of persistent donor-specific tolerance without rejection and reduces the incidence of graft versus host disease to far below the incidence level when the IV-BMT method is used (Esumi et al., 2003, Guo et al., 2008, Kaneda et al., 2005, Okazaki et al., 2008, Taira et al., 2005). IBM-BMT has also been successful in treating a wide variety of diseases in animal models, including autoimmune disease (Kushida et al., 2001), hematotologic malignant neoplasms (Suzuki et al., 2005), diabetes mellitus (Taira et al., 2005), emphysema (Adachi et al., 2006), senile osteoporosis (Takada et al., 2006) and dementia (Li et al., 2009).

In the present study, we investigated the distribution and time-dependent changes in the density of bone marrow-derived cells as well as their differentiation in the brain parenchyma in a non-inflammatory condition in bone marrow chimeric mice produced by the IBM procedure, and then compared the findings with those by the IV procedure. Based on the pattern of marrow-derived cell distribution, we focused on the anatomical, histological and cytokine expression features of the choroid plexus stroma, the attachments of choroid plexus and adjacent brain parenchyma.

Section snippets

Animals

Male C57BL/6N mice at 8–16 weeks of age (B6 mice) were used as recipients, and male C57BL/6-Tg (CAG-EGFP) mice at 5–14 weeks of age that expressed enhanced green fluorescent protein (GFP; GFP-B6 mice) were used as donors for syngeneic transplantation. B6 mice at 8–12 weeks of age were also used as non-treated mice. Mice were purchased from SLC (Shizuoka, Japan) and maintained in the animal facility of Kansai Medical University under specific pathogen-free conditions. All mice were handled in

Distribution of bone marrow-derived donor cells in the leptomeninges, choroid plexus, perivascular spaces and circumventricular organs

GFP-immunopositive cells derived from donor bone marrow (marrow-derived cells) appeared in the leptomeninges (Fig. 1A) and choroid plexus stroma (Fig. 1B) 2 weeks after BMT, and in the brain perivascular spaces (Fig. 1C) 1 month after BMT in the chimera prepared using both the IV and IBM procedures. In the leptomeninges, the marrow-derived cells were round or spindle-shaped. The spindle-shaped marrow-derived cells were often found along the blood vessel walls and in the subpial spaces along the

Selective localization of ramified marrow-derived cells in the brain adjacent to the attachments of choroid plexus

The present study indicated that marrow-derived myeloid lineage cells entered the specific discrete regions of the adult brain parenchyma to differentiate into cells with ramified morphology in a non-inflammatory condition following syngeneic BMT. Ramified marrow-derived cells were distributed over larger numbers of discrete brain regions, and resided there for longer, in chimeric mice by IBM-BMT than IV-BMT (Fig. 9). Most of the discrete regions were adjacent to the attachments of choroid

Acknowledgments

This study was supported by Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (JSPS, Contract Grant Nos.: 22790392 to SHI and 21590458 24650190 to AS) and by Otsuka Pharmaceutical Company, Ltd. This study is a part of the Research on Allergic Disease and Immunology Committee from Health and Labour Sciences Research Grants of the Ministry of Health, Labour and Welfare.

References (76)

  • C. Hernandez-Lopez et al.

    Stromal cell-derived factor 1/CXCR4 signaling is critical for early human T-cell development

    Blood

    (2002)
  • D.C. Hess et al.

    Hematopoietic origin of microglial and perivascular cells in brain

    Exp. Neurol.

    (2004)
  • C. Hundhausen et al.

    The disintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion

    Blood

    (2003)
  • S. Ikehara

    A novel strategy for allogeneic stem cell transplantation: perfusion method plus intra-bone marrow injection of stem cells

    Exp. Hematol.

    (2003)
  • S. Ikehara

    A novel BMT technique for treatment of various currently intractable diseases

    Best Pract. Res. Clin. Haematol.

    (2011)
  • T. Imai et al.

    Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion

    Cell

    (1997)
  • R.W. Johnson et al.

    Aging, neuroinflammation, and behavior

  • H. Kaneda et al.

    Long-term observation after simultaneous lung and intra-bone marrow-bone marrow transplantation

    J. Heart Lung Transplant.

    (2005)
  • I. Karkkainen et al.

    Metalloprotease-disintegrin (ADAM) genes are widely and differentially expressed in the adult CNS

    Mol. Cell Neurosci.

    (2000)
  • S. Kent et al.

    Sickness behavior as a new target for drug development

    Trends Pharmacol. Sci.

    (1992)
  • N. Kumagai et al.

    Involvement of pro-inflammatory cytokines and microglia in an age-associated neurodegeneration model, the SAMP10 mouse

    Brain Res.

    (2007)
  • T. Kushida et al.

    Intra-bone marrow injection of allogeneic bone marrow cells: a powerful new strategy for treatment of intractable autoimmune diseases in MRL/lpr mice

    Blood

    (2001)
  • M. Li et al.

    Amelioration of cognitive ability in senescence-accelerated mouse prone 8 (SAMP8) by intra-bone marrow-bone marrow transplantation

    Neurosci. Lett.

    (2009)
  • A. Nishiyori et al.

    Localization of fractalkine and CX3CR1 mRNAs in rat brain: does fractalkine play a role in signaling from neuron to microglia?

    FEBS Lett.

    (1998)
  • M. Noda et al.

    CXCL12-CXCR4 chemokine signaling is essential for NK-cell development in adult mice

    Blood

    (2011)
  • C. Onore et al.

    The role of immune dysfunction in the pathophysiology of autism

    Brain Behav. Immun.

    (2012)
  • J.J. Oppenheim et al.

    There is more than one interleukin 1

    Immunol. Today

    (1986)
  • P.H. Patterson

    Immune involvement in schizophrenia and autism: etiology, pathology and animal models

    Behav. Brain Res.

    (2009)
  • N.J. Rothwell

    Functions and mechanisms of interleukin 1 in the brain

    Trends Pharmacol. Sci.

    (1991)
  • C. Soulas et al.

    Genetically modified CD34+ hematopoietic stem cells contribute to turnover of brain perivascular macrophages in long-term repopulated primates

    Am. J. Pathol.

    (2009)
  • T. Sugiyama et al.

    Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches

    Immunity

    (2006)
  • K. Tokoyoda et al.

    Cellular niches controlling B lymphocyte behavior within bone marrow during development

    Immunity

    (2004)
  • R. Yirmiya et al.

    Immune modulation of learning, memory, neural plasticity and neurogenesis

    Brain Behav. Immun.

    (2011)
  • R. Yirmiya et al.

    Brain interleukin-1 is involved in spatial memory and passive avoidance conditioning

    Neurobiol. Learn. Mem.

    (2002)
  • Y. Adachi et al.

    Treatment and transfer of emphysema by a new bone marrow transplantation method from normal mice to Tsk mice and vice versa

    Stem Cells

    (2006)
  • N. Anandasabapathy et al.

    Flt3L controls the development of radiosensitive dendritic cells in the meninges and choroid plexus of the steady-state mouse brain

    J. Exp. Med.

    (2011)
  • M. Asheuer et al.

    Human CD34+ cells differentiate into microglia and express recombinant therapeutic protein

    Proc. Natl. Acad. Sci. USA

    (2004)
  • J.F. Bazan et al.

    A new class of membrane-bound chemokine with a CX3C motif

    Nature

    (1997)
  • Cited by (23)

    • Delayed microglial activation associated with the resolution of neuroinflammation in a mouse model of sublethal endotoxemia-induced systemic inflammation

      2022, Toxicology Reports
      Citation Excerpt :

      Astrocytes have been shown to be the major brain parenchymal cells that are targeted by choroid plexus-derived cytokines. As the astrocytes contain cytokine receptors on the endfeet, which are located at the subpial and perivascular regions, in addition to possessing elongated cytoplasmic, these processes are involved in the choroid plexus interactions [13]. After stimulation of the astrocytes, late cytokines such as CCL11, CXCL10 and granulocyte-colony stimulating factor (G-CSF) are subsequently produced, thereby changing the microenvironment of the brain parenchyma [14,15].

    • Increased cytokine expression in the choroid plexus stroma and epithelium in response to endotoxin-induced systemic inflammation in mice

      2021, Toxicology Reports
      Citation Excerpt :

      Based on the distribution of bone marrow-derived cells in the intracranial space, it has been revealed that the leptomeninges, choroid plexus stroma, attachments of choroid plexus, brain parenchymal perivascular space, circumventricular organs and astrocytic endfeet are the major histological architecture that gives a site for cell-cell interaction between immune and brain parenchymal cells [1]. It should be noted that the density of bone marrow-derived cells (mostly with myeloid differentiation) is highest in the choroid plexus stroma, followed by the leptomeninges [1]. Perivascular, leptomeningeal and choroid plexus macrophages transmit immune responses to the brain parenchyma at brain boundaries [2].

    • Pediatric acute lymphoblastic leukemia—Conquering the CNS across the choroid plexus

      2018, Leukemia Research
      Citation Excerpt :

      Although a recently published in vitro study on the cellular interaction of the BBB with lymphoblasts failed to detect an altered expression of CXCL12 in endothelial cells [33], the role of a paracrine secretion of CXCL12 within the choroid plexus during CNS invasion of ALL across the BCSFB is yet elusive. Of note, CXCL12 was recently shown to be released within the choroid plexus stroma in a mouse model capable to guide bone marrow-derived immune cells to enter CNS structures adjacent to the choroid plexus [36], and may correspondingly promote ALL cell invasion to the brain. Given this local expression of CXCL12 and results from other groups discussed above, our in vitro finding indicate a permissive role of CXCL12/CXCR4 signaling on the CNS invasion of (at least) T-ALL across the blood-cerebrospinal fluid barrier.

    • Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences

      2017, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      Other studies using BM-chimera models suggest that repeated stress exposure increased trafficking of peripheral monocytes into the brain parenchyma, promoting neuroinflammation (Ataka et al., 2013, Brevet et al., 2010). Moreover, reports indicate that BM-derived monocytes that enter the brain can induce anxiety- or depressive-like responses (D'Mello et al., 2009, Hasegawa-Ishii et al., 2013, Sawada et al., 2014). However, LysM-GFP mice showed that peripheral monocytes were localized in brain vasculature following RSD (Wohleb, 2013).

    View all citing articles on Scopus
    View full text