Elsevier

Immunology Letters

Volume 122, Issue 2, 21 February 2009, Pages 150-158
Immunology Letters

The drug monosodium luminol (GVT) preserves crypt-villus epithelial organization and allows survival of intestinal T cells in mice infected with the ts1 retrovirus

https://doi.org/10.1016/j.imlet.2008.12.012Get rights and content

Abstract

Of the cytopathic retroviruses that affect mammals, including HIV-1, many selectively infect CD4+ T cells and cause immunosuppressive syndromes. These diseases destroy both the thymus and the small and large intestines, after infecting and killing T-lineage cells in both tissues. A mutant of the murine leukemia retrovirus MoMuLV-TB, called ts1, causes this syndrome in susceptible strains of mice. In FVB/N strain mice that are infected at birth, thymic atrophy, CD4+ T cell loss, intestinal collapse, body wasting, and death occur by ∼30–40 days postinfection (dpi). Apoptosis of ts1-infected T-lineage cells, in the thymus, peripheral lymphoid system and intestines is caused by accumulation of the ts1 mutant viral envelope preprotein gPr80env, which is inefficiently cleaved into the mature viral proteins gp70 and PrP15E. We show here that ts1 infection in the small intestine is followed by loss of intestinal epithelial cell (IEC) thyroid-stimulating hormone (TSH) and cell cycling gradients (along the crypt-villus axes), accumulation of gPr80env in intestinal cells, apoptosis of developing T cells in the lamina propria (LP), and intestinal collapse by ∼30 dpi. In infected mice treated with the antioxidant drug monosodium luminol (GVT®), however, normal intestinal epithelial cell gradients are still in place at 30 dpi, and IECs covering both the crypts and villi contain large amounts of the antioxidant transcription factor Nrf2. In addition, no apoptotic cells are present, and accumulated gpr80env is absent from the tissue at this time. We conclude that GVT treatment can make ts1 a noncytopathic virus for intestinal lymphoid cells, as it does for thymocytes [25]. As in the thymus, GVT may protect the intestine by reducing oxidant stress in infected intestinal T cells, perhaps by prevention of gPr80env accumulation via Nrf2 upregulation in the IECs. These results identify GVT as a potential therapy for intestinal diseases or inflammatory conditions, including HIV-AIDS, in which oxidative stress is a triggering or exacerbating factor.

Introduction

The murine retrovirus MoMuLV causes thymic leukemias by 8–10 months of age in susceptible mice that are infected at birth [1]. A mutant of MoMuLV, called ts1, causes a fulminant disease with a much shorter timecourse, leading to death of infected animals by 30 days after infection (dpi) [2]. The ts1 mutant has a mutation in its env gene, so that productively infected cells contain an abnormal gPr80env preprotein [3], [4]. This abnormality interferes with the normal cleavage of the gPr80env to the mature viral envelope proteins gp70 and PrP15E. Without efficient cleavage, gPr80env accumulates in certain infected target cells, including astrocytes in the central nervous system (CNS), thymocytes and T cells [5], [6]. In glial cells of the central nervous system, this accumulation initiates an unfolded protein response, endoplasmic reticulum stress, mitochondrial stress, oxidative stress and apoptosis [7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18]. Similar responses occur in T-lineage cells of infected mice, which accumulate gpr80env and die as a consequence of oxidative stress-induced apoptosis [19].

The systemic consequences of ts1 infection are similar to HIV-AIDS at many levels [6], [8], [20]. Studies in this paper were designed to identify specific effects of ts1 infection on the mouse small intestine, whose symptomatology is similar to that induced by HIV-1, the primate retrovirus SIV, and the feline immunodeficiency virus FIV [21], [22], [23], [24]. The intestines are significant in retroviral diseases for another reason. Like the thymus, they can receive bone marrow T-cell progenitors and host their maturation into mature T cells (see below). This makes this tissue an important target for retroviral infection and cellular damage, because production of naïve T cells normally continues in these tissues throughout life, and because intestinal lymphoid cells in the LP govern the health and functions of the mucosal epithelium above. We therefore conducted this study in parallel with investigations of thymus tissues in ts1-infected mice [25].

Fig. 1 shows that mouse small intestines are composed of finger-like villi facing the gut lumen and of crypts buried in the connective tissue. Both regions are covered with a continuous layer of epithelium, with the pluripotent stem cells for the entire epithelium residing in the lower crypts [26], [27], [28]. These stem cells give rise to several cell types found on the villi: to absorptive enterocytes covering the villi (the predominant cell type in the mucosa), to mucus-producing goblet cells and enteroendocrine cells scattered over the villi between the enterocytes (specific cell types are not shown in the figure). Crypt stem cells also give rise to defensin-producing Paneth cells, which form a cup at the bottom of the crypts, and which do not move upward toward the villi [27], [28].

The progeny of the crypt epithelial stem cells are transit amplifying cells and differentiating cells, which move upward, as they divide, toward the crypt-villus junction [CVJ]. Once differentiated, these cells stop dividing, so that the epithelium covering the villi, above the CVJ, contains mostly quiescent enterocytes, goblet cells and enterendocrine cells nudging each other passively upward to the tips of the villi. When the IECs arrive at the tips of the villi, they undergo apoptosis and are shed, together with any T cells that have inserted themselves between the cells (top of Fig. 1). The CVJ thus is a sharp boundary that separates dividing from quiescent cells.

As noted above, the small and large intestines of mammals can host the differentiation of bone marrow T-precursor cells into mature T cells [29], [30], [31], [32]. The idea that the intestine might host T cell differentiation was put forward more than 10 years ago (reviewed in [32]). However, this claim was controversial until recent laser-capture confocal microscopy studies established clonal identity between early T cell progenitors in aggregates in the crypts, called cryptopatches (CPs) and mature T cells in the distal villi [33].

Fig. 1 shows T-cell progenitors differentiating in the lamina propria (LP) connective tissue beneath the CMJ, and mature T cells above it. This reflects the continuous arrival in the crypts of bone marrow stem cells, which are c-kit-positive (red). These begin differentiation to become mitotic T-cell progenitor progeny in the crypt LP (purple). The maturing and fully mature descendants of these intestine-born T cells are CD8-positive T cells that reside in the LP of the villi [29], [30], [31], [32]. CD4+ cells are also present, but these arrive from the blood as mature T cells that have already completed their differentiation in the thymus [34]. These are scattered throughout the crypt and villous LP. As in the thymus, therefore, the CVJ boundary separates distinct epithelia whose adjacent T cells are at distinct developmental stages.

In the normal thymus, the cortical and medullary epithelial cells express two distinct cytokeratin types [35]. Unlike the thymus however, the intestinal epithelium is a simple epithelium throughout; and all of its cells express CK8 only [36], as do cortical epithelial cells of the thymus. However, two other differences distinguish the epithelial cells of the intestinal crypts from those of the villi. One is these is the constitutive expression of TSH by crypt epithelial cells, and not by epithelial cells of the villi [37], [38], [39], [40]. The other is the presence of dividing epithelial cells exclusively in crypts, but not in the villi [26]. The presence and maintenance of these two standing gradients in the epithelial intestine reflects the presence of normally differentiating T-cell progenitors in the LP beneath, just as the presence of a healthy corticomedullary epithelial separation does for thymocytes [25]. Like thymocytes, intestinal T cells undergo sequential developmental checkpoints that depend upon their being at the right place, at the right time, in relation to the epithelium above [35].

In mice infected with ts1 (ts1-only), we show here that intestinal epithelial cell gradients are dysregulated, that extensive T cell apoptosis occurs, and that the intestines collapse by 30 dpi. In mice infected with ts1 and also treated with GVT (ts1-GVT), however, crypt-villus cytoarchitecture is intact at 30 dpi, intestinal epithelial cell gradients are in place, no T cell apoptosis is occurring, and the gPr80env protein is not accumulated by any cell type in the tissue. In the intact small intestines of GVT-protected mice, most epithelial cells are negative for gp70, and thus do not appear to be infected, but they do contain large amounts of the transcription factor Nrf2, which orchestrates protective antioxidant responses [41], [42]. These effects of ts1-GVT treatment are remarkably similar to those occurring in the thymi of ts1-GVT mice, whose epithelial cells also become loaded with Nrf2 [25].

Section snippets

Virus

The ts1 virus, a mutant of MoMuLV, was propagated in TB cells, a thymus-bone marrow cell line, and titered on 15F cells, as previously described [8].

Antibodies and reagents

GVT® (sodium α-luminol) was provided by Bach-Pharma, Inc., North Andover, MA. Goat anti-MoMuLV gp70 was from Microbiology Associates, Burlingame, CA (this antibody recognizes epitopes shared by the mature gp70 viral envelope protein and the precursor preprotein gPr80env). Anti-mouse Ki67 was from Vector Laboratories, Burlingame, CA.

Polyclonal

Crypt-villus organization and T cell numbers are abnormal in intestines from ts1-only mice, but not from those of ts1-GVT animals

Fig. 2 shows frozen sections of small intestines from 30 dpi uninfected, ts1-only, and ts1-GVT mice, stained with hematoxylin and eosin (H&E) for cytoarchitecture (A), with anti-gp70 to identify infected cells (B), and with anti-CD4 or anti-CD8 to identify cells in the LP of the villi (C). Hyperproliferation of the crypts is a common intestinal manifestation of infection by other retroviruses, including SIV in nonhuman primates, FIV in cats, and HIV-1 infection in humans [21], [22], [23], [24].

Discussion

Like ts1, other cytopathic retroviruses, including HIV-1, SIV and FIV, cause damage to the intestines and selective loss of CD4+ cells [6], [21], [22], [23], [24]. At present, there is still much to learn generally about intestinal responses to cytopathic retrovirus infection, but we believe that our work provides information relevant to HIV-1 enteropathy, and that it identifies GVT as a therapeutic agent that can prevent, maintain, or restore gut epithelial and T cell homeostasis in HIV-AIDS.

Acknowledgements

We thank Shawna Johnson and Rebecca Deen for assistance in preparing the manuscripts, Kent Claypool for providing invaluable help with flow cytometry analysis, and Nancy Otto and Jimi Lynn Brandon of the Science Park Histology Core for preparing excellent frozen sections and for advising us regarding immunohistochemical analysis. This work was supported by NIH grants NS43984 and MH71583 (P.K.Y.), by a Career Re-entry supplement to MH71583 for V.L.S., and by NIH grant MH077470 (V.L.S.). Other

References (52)

  • V.L. Scofield et al.

    Intestinal TSH production is localized in crypt enterocytes and in villus ‘hotblocks’ and is coupled to IL-7 production: evidence for involvement of TSH during acute enteric virus infection

    Immunol Lett

    (2005)
  • D. Montufar-Solis et al.

    Upregulation of ICOS on CD43+ CD4+ murine small intestinal intraepithelial lymphocytes during acute reovirus infection

    Biochem Biophys Res Commun

    (2006)
  • T. Nguyen et al.

    The pathways and molecular mechanisms regulating Nrf2 activation in response to chemical stress

    Free Radic Biol Med

    (2004)
  • A. Lau et al.

    Dual roles of Nrf2 in cancer

    Pharmacol Res

    (2008)
  • W. Schmidt et al.

    Rapid increase of mucosal CD4 T cells followed by clearance of intestinal cryptosporidiosis in an AIDS patient receiving highly active antiretroviral therapy

    Gastroenterology

    (2001)
  • B. Asjo et al.

    Moloney virus (M-MuLV) leukemogenesis: virus spread, antibody production and antigenic expression in neonatally virus-inoculated young mice

    Int J Cancer

    (1981)
  • P.F. Szurek et al.

    A Val-25-to-Ile substitution in the envelope precursor polyprotein, gPr80env, is responsible for the temperature sensitivity, inefficient processing of gPr80env, and neurovirulence of ts1, a mutant of Moloney murine leukemia virus TB

    J Virol

    (1990)
  • P.K. Wong et al.

    Alteration from T- to B-cell tropism reduces thymic atrophy and cytocidal effects in thymocytes but not neurovirulence induced by ts1, a mutant of Moloney murine leukemia virus TB

    Proc Natl Acad Sci USA

    (1991)
  • H.T. Kim et al.

    Enhanced proteolysis of IκBα and IκBβ proteins in astrocytes by Moloney murine leukemia virus (MoMuLV)-ts1 infection: a potential mechanism of NF-κB activation

    J Neurovirol

    (2001)
  • Y. Jiang et al.

    Retrovirus-induced oxidative stress with neuroimmunodegeneration is suppressed by antioxidant treatment with a refined monosodium a-luminol (Galavit)

    J Virol

    (2006)
  • H.T. Kim et al.

    Up-regulation of astrocyte cyclooxygenase-2, CCAAT/enhancer-binding protein-homology protein, glucose-related protein 78, eukaryotic initiation factor 2α, and c-Jun N-terminal kinase by a neurovirulent murine retrovirus

    J Neurovirol

    (2005)
  • N. Liu et al.

    The peroxisome proliferator phenylbutyric acid (PBA) protects astrocytes from ts1 MoMuLV-induced oxidative cell death

    J Neurovirol

    (2002)
  • N. Liu et al.

    Possible involvement of both endoplasmic reticulum- and mitochondria-dependent pathways in MoMuLV-ts1-induced apoptosis in astrocytes

    J Neurovirol

    (2004)
  • W. Qiang et al.

    Activation of transcription factor Nrf-2 and its downstream targets in response to Moloney murine leukemia virus ts1-induced thiol depletion and oxidative stress in astrocytes

    J Virol

    (2004)
  • W. Qiang et al.

    Astrocytes survive chronic infection and cytopathic effects of the ts1 mutant of the retrovirus Moloney murine leukemia virus by upregulation of antioxidant defenses

    J Virol

    (2006)
  • G.F. Lungu et al.

    Down-regulation of Jab1, HIF-1a, and VEGF by Moloney murine leukemia virus-ts1 infection: a possible cause of neurodegeneration

    J Neurovirol

    (2008)
  • Cited by (14)

    • Monosodium luminol reinstates redox homeostasis, improves cognition, mood and neurogenesis, and alleviates neuro- and systemic inflammation in a model of Gulf War Illness

      2020, Redox Biology
      Citation Excerpt :

      We chose to focus on redox imbalance as the underlying cause of brain dysfunction and chronic inflammation in GWI because GWI-related chemicals can trigger chronic oxidative stress in cells [32], and chronically elevated ROS levels can induce HMGB1 leakage, unrelenting inflammation and brain dysfunction [34,58]. Considering these, we chose MSL, a drug that has shown promise for re-establishing redox homeostasis in several disease models earlier [35–40]. Indeed, a higher dose of MSL reinstated redox balance in this study, which was evident from several observations.

    • Phenylbutyric acid suppresses protein accumulation-mediated ER stress in retrovirus-infected astrocytes and delays onset of paralysis in infected mice

      2010, Neurochemistry International
      Citation Excerpt :

      This hypothesis is supported by other studies showing that reactive oxygen species can be produced by the accumulation of misfolded proteins, and that antioxidants reduce ER stress and improve protein secretion, thereby reducing protein accumulation (Malhotra et al., 2008). We have also previously demonstrated that treatment of ts1-infected mice with the antioxidant compound monosodium luminol (GVT or MSL) extended their survival by decreasing gPr80env accumulation in T cells (Scofield et al., 2009a; Scofield et al., 2009b) and astrocytes (Kuang and Wong, unpublished data). This may be due to the fact that GVT, in addition to acting as a free radical scavenger, also upregulates the cellular antioxidant system with activation and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) (Reddy et al., 2010).

    • The drug monosodium luminol (GVT<sup>®</sup>) preserves thymic epithelial cell cytoarchitecture and allows thymocyte survival in mice infected with the T cell-tropic, cytopathic retrovirus ts1

      2009, Immunology Letters
      Citation Excerpt :

      The simplest way to interpret the systemic protective effect of GVT in ts1 infection, then, would start with the thymus. The survival and apparent homeostasis of its TEC and thymocyte cell compartments, under GVT treatment, could be the basis of what becomes either a fatal disease or a harmless infection in other tissues as well (including the intestine [97]). In the ts1-GVT thymus, we have shown here (1) that the TECs maintain normal characteristics in ts1-GVT mice; (2) that thymocytes of ts1-GVT mice survive despite continued infection; (3) that the thymocytes have lower than normal ROS levels; (4) that TEC Nrf2 levels are upregulated and stabilized; and (5) that these thymocytes do not contain accumulated gPr80env.

    View all citing articles on Scopus
    View full text