• Non ci sono risultati.

e ur o l og ical Dis o rd e rs Human T - L y mph ot r o pic Virus - Ass o cia te dN 17

N/A
N/A
Protected

Academic year: 2022

Condividi "e ur o l og ical Dis o rd e rs Human T - L y mph ot r o pic Virus - Ass o cia te dN 17"

Copied!
18
0
0

Testo completo

(1)

Human T-Lymphotropic Virus-Associated Neurological Disorders

Pathogenesis, Immunology, and Clinical Management

Michael D. Lairmore, Bindhu Michael, Lee Silverman, and Amrithraj Nair

1. INTRODUCTION

Human T-cell lymphotropic virus type 1 (HTLV-1), the first human retrovirus discovered, has a tropism for T lymphocytes and has been established as the causative agent of adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4 T-cell malignancy (1–4). HTLV-2 was subse- quently isolated and found to share approximately 60% nucleotide sequence homology with HTLV-1 (5). In the mid-1980s, HTLV-1 began to be associated through epidemiologic studies as a contributing factor in chronic neurodegenerative disorders. These clinical syndromes were first reported in Japan and Martinique (French West Indies), both regions that were endemic for HTLV-1 infection (6,7). Through important international cooperative studies, both syndromes were revealed to be identical in clinical presentation and are now known collectively as HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-2 has been sporadically associated with lymphoproliferative diseases and recently has been implicated in isolated case reports of neurological disease (8,9).

Currently, a total of 10 to 20 million people worldwide are estimated to be infected with HTLV-1, and HTLV-1 is considered endemic in southwestern Japan, the Caribbean basin, Central and West Africa, the Middle East, Melanesia, and South America (10). It is also a serious public health problem among the at-risk groups in Europe and North America and therefore is a target of intervention policies, including blood-donor screening (10–16). Although the majority of the infected persons (~95%) remain lifelong asymptomatic carriers of the virus, 2 to 3% of those infected develop ATLL, and another 2 to 3% develop chronic inflammatory diseases, such as HAM/TSP (11,17,18). Natural transmission of HTLV-1 and HTLV-2 occurs through cell-associ- ated transmission by three primary routes: orally via breast milk from infected mothers to chil- dren, via sexual contact (predominantly from male to female), and by exposure to infected blood or whole-cell blood products (e.g., transfusion, use of contaminated needles) (9,10,19).

Although the molecular events of virus replication are beginning to be unraveled and knowl- edge about HTLV-1- and HTLV-2-associated diseases has increased, many questions regarding the pathogenesis of neurologic diseases associated these retroviruses remain. For example, it is unclear why some HTLV-1-infected subjects develop ATLL or HAM/TSP, whereas the majority remains

319

From: Current Clinical Neurology: Inflammatory Disorders of the Nervous System:

Pathogenesis, Immunology, and Clinical Management

Edited by: A. Minagar and J. S. Alexander © Humana Press Inc., Totowa, NJ

(2)

asymptomatic. What factors contribute to the pathogenesis of these two widely divergent clinical disorders? In HAM/TSP patients, why is there preferential damage to the thoracic spinal cord?

What is the mechanism of viral persistence in the face of a strong immune response? What is the nature of the cell-mediated immune response to HTLV-1 infection, and is this response protective or could it lead to lymphocyte-mediated lesion development if it is uncontrolled? In this chapter, we focus on the pathogenesis of HAM/TSP in terms of its clinical features, pathologic lesions, patient susceptibility factors, and immune responses and review the proposed mechanisms for the development of this chronic neurological disorder.

2. CLINICAL FEATURES OF HAM/TSP

In evaluation of the clinical presentation of HTLV-associated neurological disorders, it is important to understand that HTLV-1 has been associated not only with HAM/TSP but also with several inflammatory diseases, such as alveolitis, polymyositis, arthritis, uveitis, and peripheral myopathy (11,18). HAM/TSP is strongly associated with HTLV-1 infection and is characterized as a progressive chronic myelopathy, with preferential damage of the thoracic spinal cord (11,20–23). In affected patients, the myelopathy presents as paraparesis with spasticity in the lower extremities, hypereflexia, muscle weakness, and sphincter disorders, including dysfunction of the urinary bladder and intestines (Table 1). Less frequently, it may precede or give rise to a cerebellar syndrome with ataxia and intention tremor (24). Some HAM/TSP patients exhibit a predominance of sympathetic nervous system dysfunction (25). HAM/TSP patients may exhibit prolonged central motor conduction time (CMCT) in all the limbs and central sensory conduction time in lower limbs, with CMCT in upper limbs correlating with the clinical severity of HAM/TSP (26,27). Magnetic resonance imaging (MRI) of HAM/TSP patients reveals the pres- ence of multiple white-matter lesions in both the spinal cord and the brain, involving perivascular demyelination and axonal degeneration (28–31). In addition, recent studies show that the carriers and patients with HAM/TSP exhibit mild cognitive impairments in verbal and visual memory, attention, and visuomotor abilities (32).

Table 1

Summary of HTLV-1- and HTLV-2-Associated Inflammatory Neurological Diseases Associated

Viral neurological Typical presenting Host factors associated

etiology diseases symptoms Treatments with disease

HTLV-1 HAM/TSP Spasticity in the lower Corticosteroids, Certain patient HLA extremities, hypereflexia, plasmapheresis, haplotypes muscle weakness, interferon, and (HLA-A*02 and sphincter disorders antiretroviral Cw*08) lead to a (urinary bladder and drugs. lower risk of HAM/TSP, intestines), and less Experimentally, whereas HLA-B*5401 frequently, cerebellar BPHA inhibitors and DRB1*0101 syndrome with ataxia of MMP-2 and are associated and intention tremor MMP-9 with an elevated

risk of HAM/TSP HTLV-2 Isolated cases Spastic ataxia Corticosteroids Unknown

of spastic and

pararesis symptomatic

and ataxia support

HTLV, human T-lymphotropic virus; HAM/TSP, HTLV-1-associated myelopathy/tropical spastic paraparesis;

BPHA, N-biphenyl sulphonyl-phenylalanine hydroxamic acid; MMP, matrix metalloproteinase.

(3)

3. SPINAL CORD AND BRAIN PATHOLOGY ASSOCIATED WITH HAM/TSP

In patients with HAM/TSP, the primary site of lesion development is the spinal cord; lesions are particularly severe in the middle to lower thoracic regions (11,33,34). This lesion distribution is consistent with neurological symptoms, such as spastic paraparesis of the lower limbs. Brain lesions less commonly may occur simultaneously with spinal cord involvement and may be detected using MRI, which reveals abnormal-intensity lesions in the brain (white matter) of patients with HAM/TSP (28,29,31,35). Within the spinal cord, regions mainly affected are the so- called watershed zones, suggesting a nonrandom distribution (36). These include degenerative lesions in the lateral corticospinal tract and spinocerebellar or spinothalamic tract of the lateral col- umn. Damage to the anterior and posterior columns is less extensive and more variable than that of the lateral column, in correlation with the clinical findings. In addition, there is also perivascular and parenchymal cellular infiltration, consisting of mainly T cells (both CD4

+

and CD8

+

T cells), macrophages, astrocytes, and glial cells (33). Similar perivascular inflammatory infiltration is also present in the brain, particularly in the deep white matter and the marginal area of the cortex and white matter (33). Interestingly, the nature of perivascular-infiltrate and cytokine expression varies depending on the duration of the disease. Patients with short duration of illness, extending from 2.5 to 4.5 yr, exhibit distribution of CD4

+

cells, CD8

+

cells, and macrophages with the presence of proinflammatory cytokines, such as interleukin (IL)-1 β, tumor necrosis factor (TNF)-α, and inter- feron (IFN)- γ (37,38), whereas in those with more prolonged duration of illness, extending from 8 to 10 yr, there are greater numbers of CD8

+

cells compared to CD4

+

cells, with fewer proinflam- matory cytokines (33,39). The distribution of macrophages and microglia varies depending on the duration of illness (40). Intriguingly, patients with shorter duration of spinal cord lesions and with a predominance of CD4

+

cells appear to contain more HTLV-1 pX and pol DNA than do those with longer duration of illness who exhibit fewer CD4

+

cells (41). HTLV-1 tax RNA is also found in infiltrating CD4

+

T lymphocytes in active central nervous system (CNS) lesions in patients with HAM/TSP (42), particularly within astrocytes, but not in perivascular infiltrating cells (43). These reports collectively suggest that viral proteins may elicit the early inflammatory lesions associated with HAM/TSP, but subsequent lesions may develop as a result of secondary injury caused by cytokines and other inflammatory mediators from infiltrating leukocytes.

Within the myelopathy, widespread loss of myelin and axons has been observed (33,44).

β-Amyloid precursor protein (APP), an early axonal damage marker, is more intensively expressed in areas of active-inflammatory lesions than in those of inactive-chronic lesions, suggest- ing that axonal damage is closely associated with inflammation in active-chronic lesions (34). Addi- tionally, the presence of APP

+

axons without relation to inflammatory cells in inactive-chronic lesions suggests that soluble neurotoxic factors might induce the axonal changes (34). This same study reported that myelinated fibers in the anterior and posterior spinal roots in lower thoracic to lumbar levels had occasional APP

+

axons, suggesting that spinal nerve roots can be affected in HAM/TSP, especially in lower thoracic to lumbar levels.

4. ANIMAL MODELS OF HAM/TSP

A variety of animal models of HTLV-1 infection have been reported, although the virus consis-

tently infects only rabbits (45,46), some nonhuman primates (47,48) and, to a lesser extent, rats

(49,50). Viral transmission in mice using typical methods of infection produces inconsistent

infections and limited virus expression in tissues (51–55). Nonhuman primates have been infected

with HTLV-1, and certain species have a natural infection with simian T-lymphotropic virus infec-

tion type 1 (56–62). The squirrel monkey has been successfully infected with HTLV-1 and offers

an attractive nonhuman primate model of HTLV-1 for vaccine testing (63–66). Rats have been

infected with HTLV-1 cell lines and offer a model of the neurological disease associated with the

(4)

viral infection (67–70). In addition, rats have been successfully used to test the role of cell-mediated immunity to the infection (71–73), although controversy exists regarding the reproducibility of the viral infection in rats (50).

The WKA strain of rat appears to be uniquely susceptible to an inflammatory neurological disease resembling HAM/TSP when is infected with HTLV-1-infected cell lines via inoculation (74). This strain appears susceptible to the infection with approximately 80% infection efficiency when rats are exposed at 16 wk of age with HTLV-1 cell lines derived from rat, human, or rabbit cells (74–76). In this model, affected rats develop spastic paraparesis of the hind legs with lesions confined primarily to the lateral and anterior funiculi of the spinal cord. Lesions include extensive symmetrical white- matter degeneration characterized by loss of myelin, axon damage, vacuolar degeneration, infiltra- tion with foamy macrophages, and astrocytic gliosis, most severe within the thoracic cord and continuing from the cervical to the lumbar area with degeneration of nerve roots and peripheral nerves (70,75). Electron microscopic studies of these lesions indicated symmetrical alterations mainly confined to the marginal areas of white matter with distribution in the anterior and the lateral columns. Additional ultrastructural features included separation of myelin lamellae and vacuolation of myelin sheaths with debris and the presence of both demyelinated and remyelinated axons, occa- sionally with tubuloreticular inclusions (74,75). Ultrastructural alterations in the peripheral nerve were similar to those of the spinal cord with the addition of apoptosis of oligodendrocytes and Schwann cells. Similar to the human infection virus, particles were absent in either the spinal cord or peripheral nerve lesions (75). Antibodies against HTLV-1 antigens were demonstrated in the plasma and cerebrospinal fluid (CSF) of these rats, whereas HTLV-1 provirus was less frequently detected from the peripheral blood mononuclear cells (PBMCs) and spinal cords of these rats; however, many other strains of rats do not develop inflammatory neurological lesions under the same conditions, suggesting that the development of the lesions in rats is under strict genetic restriction (69).

Although the clinical and neuropathological features of the WKA rat model of HAM/TSP generally mimic those of HAM/TSP in humans, the inflammatory cell infiltration is generally reduced or absent in the affected rat spinal cord lesions, unlike the disease in humans (77).

Using a variety of promoters and enhancers to drive gene expression, transgenic mouse models of HTLV-1-associated diseases have provided new tools to understand the role of tax and enve- lope proteins in the pathogenesis of HTLV-1-associated lesions. Nerenberg et al. (78,79) produced neurofibromas associated with peripheral nerves in tax transgenic mice by 8 mo of age. These mice also exhibit skeletal lesions similar to that in patients with neurofibromatosis who have a mutation in the NF-1 gene. Dysregulation of cytokines, in particular granulocyte-macrophage colony-stimulating factor (GM-CSF), may have resulted in the infiltration of neutrophils associ- ated with these lesions (80). Kitajima and Nerenberg (81) decreased the size of these neurofi- broma tumors with antisense inhibition of NF- κB, directly demonstrating the importance of this transcription factor in the development of the tumors. A limitation of this line using mice as a model of HTLV-1 is the lack of reports associating the virus with lesions of peripheral nerves.

Seeking to understand how the virus targets nervous system tissue, Coscoy and Ozden et al. (82) demonstrated that the HTLV-1 promoter directs the expression of lacZ markers in choroid plexus cells, ependymal cells and, occasionally, in neurons. Some lines of tax transgenic mice have increased nerve growth factor-, GM-CSF-, and IL-2-receptor expression in their tissues (83,84).

The wide array of inflammatory lesions associated with HTLV-1 have long been hypothesized to be mediated by autoimmune reactions triggered by the virus (78,85–87).

5. VIRAL AND HOST FACTORS THAT CONTRIBUTE TO HAM/TSP

For carriers in Japan, the lifetime risk of HAM/TSP is estimated to be 0.23%, based on multiple

risk factors (88). Significant progress has been made in the understanding of the risk factors that con-

tribute to the development of HAM/TSP. These studies collectively indicated that the disease is influ-

enced by a complex interplay between host factors, in particular the immune response of infected

(5)

subjects and viral properties that allow HTLV-1 and HTLV-2 to persist in the presence of a vigorous immune response.

5.1. Human T-Cell Lymphotropic Virus Proviral Load and Patient Gender Among the factors that have been linked to a higher risk of HAM/TSP is a high HTLV-1 proviral load (51,89–92). PBMCs of HAM/TSP patients have ~16-fold more proviral DNA than do asymptomatic carriers (93,94). Patients with proviral loads representing more than 1% of the total PBMC may have a significantly higher risk of HAM/TSP, suggesting that increased viral loads contribute to the development of lesions (11). In patients with HAM/TSP, the anti-HTLV-1 anti- body titer, which parallels the number of viral-infected PBMC, is a useful correlate to predict HAM/TSP in HTLV-1 carriers (89,91).

5.2. Host Genetic Factors

Host genetic factors have been demonstrated to clearly have a role in the development of HTLV-1-associated diseases, including HAM/TSP. Asymptomatic carriers from families of patients with HAM/TSP were found to have elevated proviral loads, compared to unrelated asymptomatic carriers (95), suggesting the role of inherited traits in the control of HTLV-1 proviral loads. Certain human leukocyte antigen (HLA) alleles have been associated with a lower risk of developing HAM/TSP, whereas other HLA alleles have been linked to a higher risk of developing ATLL (96–98).

HLA-A*02 and Cw*08 are associated with lower patient HTLV-1 proviral loads and a reduced risk of developing HAM/TSP, whereas HLA-B*5401 is associated with higher proviral loads and an ele- vated risk of developing HAM/TSP (96,97). Additionally, HLA-DRB1*0101 was linked to a higher risk of HAM/TSP (99), which was later demonstrated to be only in the absence of the protective effect linked to HLA-A*02 (96). Intriguingly, those individuals with major histocompatibility com- plex (MHC) class I molecules linked to a higher risk of ATLL, such as HLA-A*26, HLAB*4002, HLA-B*4006, and HLA-B*4801, have anchoring regions, unable to bind to 69 different HTLV-1 tax peptides and are thus predicted to be unable to produce efficient tax-specific cytotoxic T lympho- cytes (CTL) responses, characteristic of HAM/TSP (98). On the other hand, MHC class I molecules predisposing to persons to HAM/TSP likely have a strong affinity through anchoring regions for binding tax peptides and thus are able to generate a strong CTL response against HTLV-1-infected cells and also against resident CNS cells, leading to HAM/TSP. In support of this contention, patients with HAM/TSP exhibit elevated HTLV-1-specific CTL activity, whereas patients with ATLL have been found in some studies to have lower HTLV-1-specific CTL activity (100,101);

however, these findings and theories are not unequivocal. For example, the frequency of HLA-A*02 allele considered to be protective in HAM/TSP (96) is similar in some studies of asymptomatic car- riers and patients with HAM/TSP (98). Moreover, this allele is also found in patients with ATLL. In addition, stromal cell-derived factor 1 +801A 3′ untranslated region, and IL-15 191C alleles have been recently linked to a lower risk for developing HAM/TSP, whereas the promoter TNF-863A allele was associated with higher risk for developing HAM/TSP (102).

5.3. Strain Variations in HTLV-1

There have been several studies attempting to identify viral strains that are uniquely neu- ropathogenic or leukemogenic; however to date, unique viral strains associated with neurologic dis- ease have not been conclusively identified (89,103–107). Most of these studies focused on variation in the HTLV-1 tax gene. In addition to being recognized by HTLV-1-specific CD8

+

T cells, tax is a strong transactivator of many host genes, such as inflammatory cytokines and their receptors. The role of tax in the pathogenesis of HAM/TSP is not completely understood; however, it is thought that a variation of HTLV-1 tax could cause changes in host immune functions and disease progression.

Based on the high frequency of defective proviruses carrying only part of the tax gene in the CNS

of patients with HAM/TSP, tax was suggested to have a role in the development of the disease (108).

(6)

Sequence analysis of HTLV-1 tax in patients with HAM/TSP and asymptomatic carriers revealed that four nucleotide substitutions in the tax gene increase the risk of HAM/TSP (109). Another study of monozygotic twins and their infected mothers and brothers who also had HAM/TSP indicated that three infected individuals had the consensus tax sequence, whereas the asymptomatic twin car- ried sequence variations in five nucleotide positions, including four substitutions resulting in amino acid changes (110). Some patients with ATLL had cells containing HTLV-1 proviral mutants with large deletions of tax or premature stop codons in the 5´ half of tax (111). Renjifo et al. (112) also reported that amino acid substitutions in tax are associated with HAM/TSP, but Mahieux et al. (113) refuted this, reporting that HAM/TSP is simply linked to the cosmopolitan genotype.

Other viral proteins are also thought to contribute to the diversity in diseases caused by HTLV-1.

HTLV-1 accessory protein p12

I

, which is a product of the pX open reading frame I (ORF-I), is associated with immature forms of the MHC class I, interferes with the interaction of MHC class I with β

2

-microglobulin, decreases the surface expression of transfected MHC-I, directs its degra- dation in the proteasome, and is coexpressed in Hela-Tat cells (114,115). These results suggest that p12

I

might help the virus escape immune surveillance by downregulating MHC class I sur- face expression. Interestingly, proteasome destabilization of viral proteins is considered to be an intracellular defense mechanism against viral infection (116,117). Lysine is a known target for covalent binding of ubiquitin (118), and the metabolic instability of p12

I

is mediated in part by ubiquitylation at a single lysine residue at position 88 and subsequent proteasomal degradation, as well as by destabilizing residues at its amino terminus (119).

Interestingly, earlier analysis of p12

I

ORF in 21 HTLV-1 strains from different geographical areas (120) had demonstrated that the p12

I

amino acid sequence is highly conserved and that an arginine residue is found more frequently at position 88, whereas the less frequent lysine-carrying allele was found only in some HAM/TSP cases. Trovato et al. (119) extended this information by studying an additional 32 ex vivo samples from healthy carriers, patients with HAM/TSP or ATLL, and families in which both diseases occur, and confirmed that the lysine residue is found only in patients with TSP/HAM, regardless of geographical locations, suggesting that a selective pressure over p12

I

might occur in the host; however, lysine residue at position 88 in p12

I

was not found in all patients with HAM/TSP. It is hypothesized that the reduced stability of p12

I

in patients with HAM/TSP caused by this sequence variation may facilitate generation of a viral-specific CTL response, since degradation of p12

I

would alleviate the reduction of MHC class I molecules at the cell surface.

This lysine residue at position 88 of p12

I

did not appear to be a universal diagnostic marker for HTLV-1-associated neurological disease, since this phenotype was found not only in patients with HAM/TSP patients but also in asymptomatic HTLV-1 carriers who did not develop neurologic signs (121). A limitation of most studies is the analysis of samples from individuals who were born in the same geographic region, and thus, these findings might simply represent a particular HTLV-1 carrier population in which the selective pressure on the p12

I

sequence would not occur. Therefore, the significance of natural p12

I

alleles is unclear, and it is possible that the lysine at position 88 of p12

I

might have a significant effect on the biological effects of the protein in the host, including giv- ing a possible selective advantage in individuals with a certain MHC class I. Thus, to date the role of HTLV-1 sequence variation does not appear to be a significant factor in the pathogenesis of HAM/TSP, and most evidence indicates host immunologic factors are more important determinants of HTLV-1-associated neurological disease.

5.4. Route of Infection

HTLV-1 and HTLV-2 are highly cell-associated viruses and are transmitted by routes that favor

preservation of infected cells. Consequently, these viruses are transmitted by exposure to whole

blood or blood products that contain intact cells, intravenous drug use, sexual transmission (pre-

dominantly from male seminal fluid), and breastfeeding (90,122). Exposure to HTLV-1 via blood

transfusion has been associated with the development of HAM/TSP (123), whereas mucosal exposure

(7)

has been linked to the development of ATLL (122,124). In a rat model of HTLV-1 infection, oral inoculation of HTLV-1-infected cells resulted in immune unresponsiveness in spite of persistent HTLV-1 infection (125). Oral vs intravenous routes of infection in rats has revealed differences in levels of anti-HTLV-1 antibodies and proliferative T-cell response against the virus, indicating that oral exposure may result in a reduced antiviral immune response (73). From these data, investigators have hypothesized that the absence of an effective T-cell response in subjects exposed via mucosal or oral routes might lead to survival and subsequent transformation of the infected cell population, resulting in ATLL. On the other hand, intravenous inoculation may result in a vigorous immune response that, in the process of elimination of cells infected with HTLV-1, may promote inflammation in the CNS typical of HAM/TSP (33). Collectively, these studies suggest that the route of infection, in particular intravenous inoculation of HTLV-1-infected cells, influences the immune response gener- ated against the virus and contributes to the incidence of HAM/TSP in patients exposed by this route.

5.5. The Host Immune Response to HTLV-1

Patients with HAM/TSP exhibit 10 to 100-fold increases in proviral DNA in their peripheral blood, compared to asymptomatic, but infected, subjects (90,126). Furthermore, when an infected subject’s proviral load is more than 1% of PBMCs, there is higher risk of HAM/TSP (11), although HAM/TSP occurs in some patients with a low proviral DNA load, indicating that a high proviral DNA load by itself is not a conclusive predictor for the development of HAM/TSP (11).

Most studies support the hypothesis that the progression to HAM/TSP is influenced by an interac- tion of host factors, including the immune response, viral factors, and high proviral DNA load.

Typically, T-cell transformation is a feature of ATLL, whereas cellular destruction and inflamma- tion are a feature of HAM/TSP. These distinctive characteristics might account for the fact that there are very few patients with both ATLL and HAM/TSP. Similarly, these diseases differ in various immunologic parameters related to HTLV-1 infection. Although CD4

+

T cells, CD8

+

T cells, and activated macrophages are present within white-matter lesions of patients with HAM/ TSP (127), leukemic cells in ATLL are mostly CD4

+

T cells and occasionally are CD4/CD8 double-positive (17,18). Peripheral blood and the CSF of patients with HAM/TSP have high levels of proinflam- matory cytokines, such as IFN- γ, TNF-α, IL-1, and IL-6, suggesting the importance of these proinflammatory cytokines in HAM/TSP (128–131). Additionally, CSF of these patients contains activated lymphocytes, suggesting that activated lymphocytes and macrophages also have a role in the development of HAM/TSP (127).

Patients with HAM/TSP develop a vigorous expansion of CD8

+

T cells, compared to asymp- tomatic carriers (132) and HTLV-1 tax-specific CTLs are readily demonstrated in the CSF of these patients (133). In fact, patients with HAM/TSP have about 40 to 280-fold more tax-specific precur- sor CTLs than do asymptomatic carriers, consistent with an increase in virus-specific CD8

+

CTL response in these patients. Additionally, patients with HAM/TSP have high levels of anti-HTLV-1 antibodies in serum and CSF (134). Studies of peripheral blood and CSF from the same patient with HAM/TSP have revealed tax-specific CTLs to be about twofold higher in the CSF, suggesting the possible role of tax-specific CTLs in the cellular destruction and inflammation in the CNS of these patients (133). Activated CTLs are present in active inflammatory lesions in the spinal cords of patients with HAM/TSP (92,135). The mechanism of cell destruction by tax-specific CTLs is not elucidated; however, it is proposed that CTLs could directly kill not only HTLV-1-infected cells expressing tax but also CNS cells expressing a crossreactive cellular determinant and neighboring uninfected neuronal cells by apoptosis mediated via the toxic inflammatory cytokines (18).

6. VIRAL ENTRY INTO THE CENTRAL NERVOUS SYSTEM

HTLV-1-infected cells are present in the CNS of patients with HAM/TSP. These patients also

have higher HTLV-1 proviral loads in the CSF than in their PBMCs (136). The HTLV-1 integration

site in the cellular DNA of HTLV-1-infected lymphocytes was the same in the CSF and peripheral

(8)

blood of these patients, indicating that HTLV-1-infected cells in peripheral blood migrate to the CNS and accumulate in lesions in patients with HAM/TSP (137,138). The mechanism of selective migration of HTLV-1-infected cells to the CNS of these patients is not clear. CD4

+

T cells from patients with HAM/TSP have an enhanced ability to attach and transmigrate through reconstituted basement membranes in vitro and endothelial cells, compared with CD4

+

T cells from asymp- tomatic carriers (139,140), indicating that these cells likely have more ability to migrate into the CNS across the blood–brain barrier (BBB). Additionally, close contact between brain endothelial cells and HTLV-1-infected CD4

+

T cells causes increased production of TNF- α, likely increasing the paracellular permeability of the brain endothelial monolayer and transmigration of activated T cells and monocytes across the BBB. Romero et al. (141) demonstrated that viral particles were taken up into vesicles by brain endothelial cells and released from the basolateral surface, whereas HTLV-1-infected T cells fuse to brain endothelial cells. Based on these observations, mechanisms proposed for the entry of HTLV-1 into the CNS of patients with HAM/TSP include paracellular transmigration of virus-infected cells through the BBB, absorption and release of viral particles from vesicles within brain endothelial cells, and direct infection of brain endothelial cells (127).

Adhesion molecules are considered critical for this migration, correlating with the increased expression of vascular cell adhesion molecule-1 expression in spinal cord lesions (142) and very late antigen-4 (VLA-4), as well as monocyte chemoattractant protein-1 in perivascular inflamma- tory infiltrates and matrix metalloproteinase (MMP)-2 and MMP-9 in infiltrating mononuclear cells of HAM/TSP patients (143). In addition, α4β1 and α5β1 integrins appear to be overexpressed in HTLV-1-infected T cells of patients with HAM/TSP (144).

After entering the CNS, HTLV-1-infected CD4

+

and CD8

+

T cells are believed to participate in a number of events, resulting in infection of resident CNS cell populations, activation of astro- cytes and microglial cells, induction of proinflammatory cytokine and chemokine synthesis, recruitment of inflammatory infiltrates into the CNS, BBB disruption, dysregulation of oligoden- drocyte homeostasis, demyelination, and axonal degradation (145). Within the CNS, astrocytes and microglial cells are susceptible to HTLV-1 infection (43,146,147), presumably serving to pro- mote increased production of proinflammatory cytokines and chemokines and to recruit of antigen- specific CD4

+

and CD8

+

T cells from the circulation.

7. MECHANISM OF VIRAL PERSISTENCE, DESPITE A STRONG IMMUNE RESPONSE

Among the many characteristics of HTLV-1 that influence viral persistence, one important pecu-

liarity of this virus is its low rate of evolution and low sequence diversity, in contrast to human

immunodeficiency virus-1. A number of mechanisms have been postulated to explain how this

retrovirus maintains persistence yet elicits a prominent immune response in infected individuals. In

this regard, it is important to understand that the virus may be spread either by infectiously (primarily

from infected cells) or by mitotic division of infected cells (replication of the integrated provirus

during cell division) (148), although based on the relatively lower level of variability in sequence,

HTLV-1 is thought to be maintained mainly by mitotic transmission and proliferation of cells har-

boring the provirus, since this route is associated with a much lower rate of mutation (17) than infec-

tious transmission. In support of this hypothesis, clonal expansion of infected CD4

+

and CD8

+

T-cell

populations have been observed in patients with HAM/TSP (137,148,149). In contrast and perhaps

not mutually exclusive are observations that some patients form a persistent and vigorous CTL

response against HTLV-1 tax, and this accounts for reduced proviral loads and protection against

HAM/TSP; these studies suggest the existence of active viral gene expression in vivo (89). Based upon

positive responses to treatments with the nucleoside analogue lamivudine (3TC), a reverse transcrip-

tase inhibitor, Taylor et al. (150) has speculated that HTLV-1 must undergo reverse transcription and

maintain tax expression to elicit a high frequency of tax-specific CD8

+

T cells. Recent studies suggest

that an elevated proportion of HTLV-1-infected cells (10–80%) express tax within 12 h of infection

(9)

and thus stimulate a high frequency of tax-specific CTLs (17,89). These in turn are stimulated to divide rapidly but are made highly susceptible to tax-specific CTL-mediated lysis. The virus cannot complete the replication cycle because of limited availability of activated susceptible host cells and the rapid CTL-mediated lysis; thus, mitotic spread might be responsible for the maintenance of the proviral load, possibly accounting for the low rate of mutation and evolution (17). Furthermore, this sequence of events would also account for the immunodominance of tax as a target antigen for CTLs, since tax-expressing cells appear to be susceptible to CTL lysis before expressing other viral proteins. Since tax is essential for the maintenance of proviral load, generation of strong CTL response to tax possibly lowers proviral load and susceptibility to HAM/TSP (89). Thus, host factors may determine the strength or efficiency of CTL response to HTLV-1 and an individual’s proviral load and their risk of HAM/TSP. Equally likely in this balance is the tenet that CTLs might con- tribute to the inflammation in HAM/TSP (90,151). The concentration of antigen necessary to elicit IFN- γ secretion by a CD8

+

T cell is greater than for target cell lysis. Based upon this immunological principal, it has been suggested that a patients balance between an active CTL response and control of virus replication vs an overactive immune response with IFN- γ secretion that promotes inflammation is a key determinant of whether a patient develops HAM/TSP (89).

8. PROPOSED MECHANISMS OF PATHOGENESIS

Immunologic mechanisms are generally considered to have a significant role in the pathogenesis of HAM/TSP. This belief is strongly supported by studies indicating that patients with HAM/TSP have elevated cellular and humoral anti-HTLV-1 immune responses in comparison to asymptomatic carriers and seronegative controls. The virus predominantly targets CD4

+

T cells (152–155), which have increased adhesion activity to endothelial cells, promoting transmigrating activity through base- ment membranes (156) and secreting proinflammatory cytokines, such as IFN- γ and TNF-α (157).

Both the peripheral blood and CSF of patients with HAM/TSP contain HTLV-1-specific HLA class I-restricted CD8

+

CTLs (100), with activity mostly against pX gene products, especially the Tax 11–19 peptide (LLFGYPVYV) (158). With spinal cord lesions, the presence of CD8

+

T cells, presumably in response to CD4

+

T cells expressing HTLV-1 proteins, suggests a direct involvement of these cells in lesion development (37,41). These HTLV-1-specific CD8

+

CTLs most likely mediate target cell destruction through perforin-dependent mechanisms or the production of inflammatory cytokines and chemoattractants (159,160), although the mechanism of maintaining these CD8

+

T cells during the prolonged course of infection is not certain. Although IL-15 may have a role in the disease (161), it is also thought that viral antigens continuously drive these cells in vivo (91).

An autoimmune mechanism has also been proposed to be involved in the pathogenesis of HAM/TSP, as in inflammatory neurological diseases, such as multiple sclerosis (MS) and Guillain- Barre syndrome. In support of this hypothesis, researchers have demonstrated that infiltrating lymphocytes in the spinal cord lesions of patients with HAM/TSP have a unique T-cell receptor CDR3 motif, which is also found in brain lesions in patients with MS and experimental autoimmune encephalomyelitis (162). An HTLV-1-infected CD4

+

T-cell clone derived from the peripheral blood of a patient with HAM/TSP showed a proliferating response to crude protein extracted from seronegative spinal cord samples but not to proteins from lymph nodes (11), whereas the HTLV-1 tax-specific CD8

+

CTL clones recognized and lysed self-peptide-pulsed target cells in vitro (163).

Another hypothesis couples infection with autoimmune disease. Molecular mimicry has also

been implicated in the pathogenesis of diseases, such as diabetes, lupus, and MS, in which there is

little direct evidence linking causative agents to pathogenic immune reactions. Molecular

mimicry is characterized by an immune response to an environmental agent that crossreacts with

a host antigen, resulting in disease. The clinical presentation of HAM/TSP is often difficult to dis-

tinguish from MS. In this regard, serum immunoglobulin (IgG) from HAM/TSP patients reacted

to neurons in human CNS not infected with HTLV-1 but not to cells in the peripheral nervous sys-

tem or other organs, and more importantly, the reactivity could be abrogated by pretreatment with

(10)

recombinant tax protein (164). Interestingly, IgG from patients with HAM/TSP identified hetero- geneous nuclear ribonuclear protein-A1 as the autoantigen (164). It was hypothesized that axons are preferentially damaged by such autoantibodies, which inhibits neuronal firing.

9. TREATMENT AND PREVENTION

To date, effective treatments against the devastating neurological complications of HAM/TSP or preventive measures to stop the development of the disease in HTLV-1-infected patients have had limited success. Interventions with corticosteroids, plasmapheresis, IFN, and antiretroviral drugs have been used as treatments in patients with HAM/TSP with poor or inconsistent results, although virological and clinical improvement has been obtained patients with HAM/TSP who were treated with zidovudine and/or lamivudine (150). Inhibition of HTLV reverse transcriptase, along with the cytostatic effect of some nucleoside analogues, including zidovudine, is thought to reduce the virus’ replication, as well as the proviral load; however, the clinical consequences of this need further examination (165). Based on the finding that MMPs, specifically MMP-2 and MMP-9, have important roles in the breakdown of the BBB in the CNS of patients with HAM/TSP, N-biphenyl sulphonyl-phenylalanine hydroxamic acid, a selective inhibitor of MMP-2 and MMP-9, was recently used tested as an inhibitor of the migrating activity of CD4

+

T cells in patients with HAM/TSP and was found effective, suggesting its therapeutic potential against HAM/TSP (166). In the past, some cases of TSP have responded to steroids, such as prednisone and danazol. These are generally considered useful in the clinical management of patients, partic- ularly females, with HAM/TSP (167). Prednisolone, IFN- α, fosfomycin, high-dose vitamin C, blood purification therapy, heparin, sulfapyridine, thyrotropin-releasing hormone, erythromycin, and mizoribine have been found useful in improving motor disability in patients with HAM/TSP, although success rates vary (168,169). Rolipram, a phosphodiesterase type IV inhibitor was demonstrated to inhibit TNF- α production in HTLV-1-infected cells lines and peripheral blood cells of patients with HAM/TSP, to variable degrees (168,170). Overall, an effective and long-term therapeutic protocol or specific drug that treats HAM/TSP is lacking.

Multiple factors favor development of a vaccine against HTLV-1: the virus displays relatively low antigenic variability, natural immunity occurs in humans, and experimental vaccination with the envelope antigen has been successful in animal models (171–173). There have been sugges- tions that gene therapy to control virus replication may be useful against HAM/TSP (174), although, thus far, there is no successful gene therapy or vaccine for HAM/TSP, owing in part to the lack of animal models that can be used for testing. In addition, the low incidence and unpre- dictability of onset of clinical signs of the disease among those infected with the virus makes clinical trial accrual problematic.

10. CONCLUSION AND FUTURE DIRECTIONS

It is critical that future studies focus on therapeutic and prevention strategies of HTLV-1- associated diseases. Although the clinical symptoms of these diseases are well characterized, the pathogenesis is not clearly understood. Our understanding of the risk factors for HAM/TSP has increased tremendously, and many theories have been proposed concerning pathogenic mecha- nisms of the disease. Many of these theories on pathogenesis and immune response are contradictory, and additional studies are necessary to elucidate this apparent dichotomy in our understanding.

Nevertheless, the knowledge of risk factors and possible pathogenesis mechanisms should enable us, in the near future, to predict the risk of progression from asymptomatic carrier stage to HAM/TSP. Future studies directed toward understanding the role of cell-mediated immunity in HTLV-1 infection will likely lead to further clarification of the pathogenesis of HAM/TSP.

This knowledge, in turn, will provide valuable directions to develop efficient therapeutic and pre-

ventive strategies against HAM/TSP. Recent studies have provided new information about repli-

cation strategies used by the virus to regulate its gene expression and will undoubtedly help in the

(11)

effort to understand the role of viral-mediated events vs immune-mediated damage in HAM/TSP.

Additional studies of the transcriptional regulation of the long terminal repeat during cell activa- tion should facilitate the design of therapeutic strategies to prevent the initiation of viral gene expression within the peripheral blood and CNS. Studies of proviral DNA and viral gene expres- sion in the peripheral blood and CNS during clinical latency and HAM/TSP would also provide important information.

There has been great progress in the development of new animal models that exhibit many of the pathological features associated with HAM/TSP and ATLL, and future studies to refine these animal models would clarify the early events that take place after initial exposure of the host to HTLV-1 and the kinetics of viral gene expression in various cell types and stages of disease development.

Gene array studies to identify patterns of gene expression in various stages in asymptomatic carriers and patients with ATLL or HAM/TSP would provide valuable insight into the pathogene- sis of these diseases. The ultimate goal of these studies is to prevent the onset of the devastating neurological diseases associated with HTLV-1 infection.

REFERENCES

1. Poiesz BJ, Ruscetti FW, Gazdar AF, Bunn PA, Minna JD, Gallo RC. Detection and isolation of type C retrovirus par- ticles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc Natl Acad Sci USA 1980;77:7415–7419.

2. Hinuma Y, Nagata K, Hanaoka M, et al. Adult T Cell Leukemia: antigens in an ATL cell line and detection of anti- bodies to antigen in human sera. Proc Natl Acad Sci USA 1981;78:6476–6480.

3. Miyoshi I, Kubonishi I, Yoshimoto S, et al. Type C virus particles in a cord T-cell line derived by co-cultivating normal human cord leukocytes and human leukaemic T cells. Nature 1981;294:770–771.

4. Yoshida M, Miyoshi I, Hinuma Y. Isolation and characterization of retrovirus from cell lines of human T-cell leukemia and its implication in the disease. Proc Natl Acad Sci U S A 1982;79:2031–2035.

5. Kalyanaraman VS, Sarngadharan MG, Robert-Guroff M, Miyoshi I, Golde D, Gallo RC. A new subtype of human T-cell leukemia virus (HTLV-II) associated with a T-cell variant of hairy cell leukemia. Science 1982;218:571–573.

6. Osame M, Usuku K, Izumo S, et al. HTLV-I associated myelopathy, a new clinical entity. Lancet 1986;1(8488):1031–1032.

7. Gessain A, Barin F, Vernant J, et al. Antibodies to human T lymphotropic virus type 1 in patients with tropical spastic paresis. Lancet 1985;2:407–410.

8. Zehender G, Colasante C, Santambrogio S, De Maddalena C, Massetto B, Cavalli B et al. Increased risk of developing peripheral neuropathy in patients coinfected with HIV-1 and HTLV-2. J Acquir Immune Defic Syndr 2002;314:440–447.

9. Lowis GW, Sheremata WA, Minagar A. Epidemiologic features of HTLV-II: serologic and molecular evidence. Ann Epidemiol 2002;121:46–66.

10. Edlich RF, Arnette JA, Williams FM. Global epidemic of human T-cell lymphotropic virus type-I (HTLV-I). J Emerg Med 2000;181:109–119.

11. Nagai M, Osame M. Human T-cell lymphotropic virus type I and neurological diseases. J Neurovirol 2003;92:228–235.

12. Courouce AM, Pillonel J, Lemaire JM, Saura C. HTLV testing in blood transfusion. Vox Sang 1998;74(Suppl 2):

165–169.

13. Ferreira OC, Planelles V, Rosenblatt JD. Human T-cell leukemia viruses: epidemiology, biology, and pathogenesis.

Blood Rev 1997;112:91–104.

14. Murphy EL, Glynn SA, Fridey J, et al. Increased incidence of infectious diseases during prospective follow-up of human T-lymphotropic virus type II- and I-infected blood donors. Arch Intern Med 1999;159:1485–1491.

15. Murphy EL, Busch MP, Tong M, Cornett P, Vyas GN. A prospective study of the risk of transfusion-acquired viral infections. Transfus Med 1998;83:173–178.

16. Schreiber GB, Murphy EL, Horton JA, et al. Risk factors for human T-cell lymphotropic virus types I and II HTLV-I and -II. in blood donors: the Retrovirus Epidemiology Donor Study. NHLBI Retrovirus Epidemiology Donor Study.

J Acquir Immune Defic Syndr Hum Retrovirol 1997;143:263–271.

17. Mortreux F, Gabet AS, Wattel E. Molecular and cellular aspects of HTLV-1 associated leukemogenesis in vivo.

Leukemia 2003;171:26–38.

18. Barmak K, Harhaj E, Grant C, Alefantis T, Wigdahl B. Human T cell leukemia virus type I-induced disease: pathways to cancer and neurodegeneration. Virology 2003;3081:1–12.

19. Fujino T, Nagata Y. HTLV-I transmission from mother to child. J Reprod Immunol 2000;472:197–206.

20. Adedayo O, Grell G, Bellot P. Hospital admissions for human T-cell lymphotropic virus type-1 (HTLV-1) associated diseases in Dominica. Postgrad Med J 2003;79:341–344.

(12)

21. Kasahata N, Shiota J, Miyazawa Y, Nakano I, Murayama S. Acute human T-lymphotropic virus type 1-associated myelopathy: a clinicopathologic study. Arch Neurol 2003;606:873–876.

22. Furukawa Y, Kubota R, Eiraku N, et al. Human T-cell lymphotropic virus type I (HTLV-I)-related clinical and labo- ratory findings for HTLV-I-infected blood donors. J Acquir Immune Defic Syndr 2003;323:328–334.

23. Kiwaki T, Umehara F, Arimura Y, et al. The clinical and pathological features of peripheral neuropathy accompanied with HTLV-I associated myelopathy. J Neurol Sci 2003;206:17–21.

24. Castillo LC, Gracia F, Roman GC, Levine P, Reeves WC, Kaplan J. Spinocerebellar syndrome in patients infected with human T-lymphotropic virus types I and II (HTLV-I/HTLV-II): report of 3 cases from Panama. Acta Neurol Scand 2000;101:405–412.

25. Alamy AH, Menezes FB, Leite AC, Nascimento OM, Araujo AQ. Dysautonomia in human T-cell lymphotrophic virus type I-associated myelopathy/tropical spastic paraparesis. Ann Neurol 2001;50:681–685.

26. Matsuzaki T, Nakagawa M, Nagai M, et al. HTLV-I proviral load correlates with progression of motor disability in HAM/TSP: analysis of 239 HAM/TSP patients including 64 patients followed up for 10 years. J Neurovirol 2001;7:228–234.

27. Suga R, Tobimatsu S, Kira J, Kato M. Motor and somatosensory evoked potential findings in HTLV-I associated myelopathy. J Neurol Sci 1999;167:102–106.

28. Godoy AJ, Kira J, Hasuo K, Goto I. Characterization of cerebral white matter lesions of HTLV-I-associated myelopathy tropical spastic paraparesis in comparison with multiple sclerosis and collagen-vasculitis: a semiquanti- tative MRI study. J Neurol Sci 1995;133:102–111.

29. Fukushima T, Ikeda T, Uyama E, Uchino M, Okabe H, Ando M. Cognitive event-related potentials and brain mag- netic resonance imaging in HTLV-1 associated myelopathy (HAM). J Neurol Sci 1994;126:30–39.

30. Kira J, Goto I, Otsuka M, Ichiya Y. Chronic progressive spinocerebellar syndrome associated with antibodies to human T-lymphotropic virus type-I—Clinico-virological and magnetic resonance imaging studies. J Neurol Sci 1993;115:111–116.

31. Kira JI, Fujihara K, Itoyama Y, Goto I, Hasuo K. Leukoencephalopathy in HTLV-I-associated mylopathy/tropical spas- tic paraparesis: MRI analysis and a two year follow up study after corticosteroid therapy. J Neurol Sci 1991;106:41–49.

32. Silva MT, Mattos P, Alfano A, Araujo AQ. Neuropsychological assessment in HTLV-1 infection: a comparative study among TSP/HAM, asymptomatic carriers, and healthy controls. J Neurol Neurosurg Psychiatry 2003;

74:1085–1089.

33. Osame M. Pathological mechanisms of human T-cell lymphotropic virus type I-associated myelopathy (HAM/TSP).

J Neurovirol 2002;8:359–364.

34. Umehara F, Abe M, Koreeda Y, Izumo S, Osame M. Axonal damage revealed by accumulation of beta-amyloid pre- cursor protein in HTLV-I-associated myelopathy. J Neurol Sci 2000;176:95–101.

35. McKendall RR, Oas J, Lairmore MD. HTLV-I-associated myelopathy endemic in Texas-born residents and isolation of virus from CSF cells. Neurology 1991;41:831–836.

36. Izumo S, Umehara F, Osame M. HTLV-I-associated myelopathy. Neuropathology 2000;20(Suppl):S65–S68.

37. Umehara F, Izumo S, Ronquillo AT, Matsumuro K, Sato E, Osame M. Cytokine expression in the spinal cord lesions in HTLV-I-associated myelopathy. J Neuropathol Exp Neurol 1994;53:72–77.

38. Umehara F, Izumo S, Nakagawa M, et al. Immunocytochemical analysis of the cellular infiltrate in the spinal cord lesions in HTLV-I-Associated myelopathy. J Neuropathol Exp Neurol 1993;52:424–430.

39. Kubota R, Furukawa Y, Izumo S, Usuku K, Osame M. Degenerate specificity of HTLV-1-specific CD8+ T cells during viral replication in patients with HTLV-1-associated myelopathy (HAM/TSP). Blood 2003;101:3074–3081.

40. Abe M, Umehara F, Kubota R, Moritoyo T, Izumo S, Osame M. Activation of macrophages microglia with the calcium- binding proteins MRP14 and MRP8 is related to the lesional activities in the spinal cord of HTLV-I associated myelopathy. J Neurol 1999;246:358–364.

41. Kubota R, Umehara F, Izumo S, Ijichi S, Matsumuro K, Yashiki S et al. HTLV-I proviral DNA amount correlates with infiltrating CD4+ lymphocytes in the spinal cord from patients with HTLV-I-associated myelopathy. J Neuroim- munol 1994;53:23–29.

42. Moritoyo T, Reinhart TA, Moritoyo H, et al. Human T-lymphotropic virus type I-associated myelopathy and tax gene expression in CD4(+) T lymphocytes. Ann Neurol 1996;40:84–90.

43. Lehky TJ, Fox CH, Koenig S, et al. Detection of human T-lymphotropic virus type I (HTLV-I) tax RNA in the central nervous system of HTLV-I-associated myelopathy/tropical spastic paraparesis patients by in situ hybridization. Ann Neurol 1995;37:167–175.

44. Ohta M, Ohta K, Nishimura M, Saida T. Detection of myelin basic protein in cerebrospinal fluid and serum from patients with HTLV-1-associated myelopathy/tropical spastic paraparesis. Ann Clin Biochem 2002;39(Pt 6): 603–605.

45. Akagi T, Takeda I, Oka T, Ohtsuki Y, Yano S, Miyoshi I. Experimental infection of rabbits with human T-cell leukemia virus type 1. Jpn J Cancer Res 1985;76:86–94.

46. Lairmore MD, Roberts B, Frank D, Rovnak J, Weiser MG, Cockerell GL. Comparative biological responses of rabbits infected with human T-lymphotropic virus Type I isolates from patients with lymphoproliferative and neurodegener- ative disease. Int J Cancer 1992;50:124–130.

(13)

47. Murata N, Hakoda E, Machida H, et al. Prevention of human T cell lymphotropic virus type 1 infection in Japanese macaques by passive immunization. Leukemia 1996;10:1971–1974.

48. Nakamura H, Hayami M, Ohta Y, et al. Protection of cynomolgus monkeys against infection by human T-cell leukemia virus type-1 by immunization with viral env gene products produced in Escerichia coli. Int J Cancer 1987;40:403–407.

49. Suga T, Kameyama T, Shimotohno K, et al. Infection of rats with HTLV-1: a Small-Animal model for HTLV-1 carriers.

Int J Cancer 1991;49:764–769.

50. Ibrahim F, Fiette L, Gessain A, Buisson N, Dethe G, Bomford R. Infection of rats with human T-cell leukemia virus type-1: susceptibility of inbred strains, antibody response and provirus location. Int J Cancer 1994;58:446–451.

51. Nitta T, Tanaka M, Sun B, Hanai S, Miwa M. The genetic background as a determinant of human T-cell leukemia virus type 1 proviral load. Biochem Biophys Res Commun 2003;309:161–165.

52. Furuta RA, Sugiura K, Kawakita S, et al. Mouse model for the equilibration interaction between the host immune system and human T-cell leukemia virus type 1 gene expression. J Virol 2002;76:2703–2713.

53. Furuta RA, Sugiura K, Kawakita S, et al. Mouse model for the equilibration interaction between the host immune system and human T-cell leukemia virus type 1 gene expression. J Virol 2002;76:2703–2713.

54. Feng R, Kabayama A, Uchida K, Hoshino H, Miwa M. Cell-free entry of human T-cell leukemia virus type 1 to mouse cells. Jpn J Cancer Res 2001;92:410–416.

55. Fang JH, Kushida S, Feng RQ, et al. Transmission of human T-cell leukemia virus type 1 to mice. J Virol 1998;72:3952–3957.

56. Leendertz FH, Boesch C, Junglen S, Pauli G, Ellerbrook H. Characterization of a new simian T-lymphocyte virus type 1 (STLV-1) in a wild living chimpanzee (Pan troglodytes verus) from Ivory Coast: evidence of a new STLV-1 group? AIDS Res Hum Retroviruses 2003;19:255–258.

57. Niphuis H, Verschoor EJ, Bontjer I, Peeters M, Heeney JL. Reduced transmission and prevalence of simian T-cell lym- photropic virus in a closed breeding colony of chimpanzees (Pan troglodytes verus). J Gen Virol 2003;84(Pt 3):615–620.

58. Gabet AS, Gessain A, Wattel E. High simian T-cell leukemia virus type 1 proviral loads combined with genetic sta- bility as a result of cell-associated provirus replication in naturally infected, asymptomatic monkeys. Int J Cancer 2003;107:74–83.

59. Takemura T, Yamashita M, Shimada MK, et al. High prevalence of simian T-lymphotropic virus type L in wild ethiopian baboons. J Virol 2002;76:1642–1648.

60. Mahieux R, Chappey C, Georgescourbot MC, et al. Simian T-cell lymphotropic virus type 1 from Mandrillus sphinx as a simian counterpart of human T-cell lymphotropic virus type 1 subtype D. J Virol 1998;72:10,316–10,322.

61. Voevodin A, Samilchuk E, Schatzl H, Boeri E, Franchini G. Interspecies transmission of macaque simian T-cell leukemia/lymphoma virus type 1 in baboons resulted in an outbreak of malignant lymphoma. J Virol 1996;

70:1633–1639.

62. Gessain A, De TG. Geographic and molecular epidemiology of primate T lymphotropic retroviruses: HTLV-I, HTLV-II, STLV-I, STLV-PP, and PTLV-L. [Review]. Adv Vir Res 1996;47:377–426, 377–426.

63. Mortreux F, Kazanji M, Gabet AS, de Thoisy B, Wattel E. Two-step nature of human T-cell leukemia virus type 1 replication in experimentally infected squirrel monkeys (Saimiri sciureus). J Virol 2001;75:1083–1089.

64. Kazanji M, Tartaglia J, Franchini G, et al. Immunogenicity and protective efficacy of recombinant human T-cell leukemia/lymphoma virus type 1 NYVAC and naked DNA vaccine candidates in squirrel monkeys (Saimiri sci- ureus). J Virol 2001;75:5939–5948.

65. Kazanji M, Ureta-Vidal A, Ozden S, et al. Lymphoid organs as a major reservoir for human T-cell leukemia virus type 1 in experimentally infected squirrel monkeys (Saimiri sciureus): provirus expression, persistence, and humoral and cellular immune responses. J Virol 2000;74:4860–4867.

66. Kazanji M. HTLV type 1 infection in squirrel monkeys (Saimiri sciureus): a promising animal model for HTLV type 1 human infection. AIDS Res Hum Retroviruses 2000;16:1741–1746.

67. Hakata Y, Yamada M, Shida H. Rat CRM1 is responsible for the poor activity of human T–cell leukemia virus type 1 rex protein in rat cells. J Virol 2001;75:11515–11525.

68. Kasai T, Ikeda H, Tomaru U, et al. A rat model of human T lymphocyte virus type I (HTLV-I) infection: in situ detec- tion of HTLV-I provirus DNA in microglia/macrophages in affected spinal cords of rats with HTLV-1-induced chronic progressive myeloneuropathy. Acta Neuropathol 1999;97:107–112.

69. Sun B, Fang J, Yagami K, et al. Age-dependent paraparesis in WKA rats: evaluation of MHC k-haplotype and HTLV-1 infection. J Neurol Sci 1999;167:16–21.

70. Ishiguro N, Abe M, Seto K, et al. A rat model of human T lymphocyte virus type 1 (HTLV-1) infection. 1. Humoral antibody response, provirus integration, and YTLV-1-associated myelopathy/tropical spastic paraparesis-like myelopathy in seronegative HTLV-1 carrier rats. J Exp Med 1992;176:981–989.

71. Hasegawa A, Ohashi T, Hanabuchi S, et al. Expansion of human T-cell leukemia virus type 1 (HTLV-1) reservoir in orally infected rats: inverse correlation with HTLV-1-specific cellular immune response. J Virol 2003;77:

2956–2963.

72. Hanabuchi S, Ohashi T, Koya Y, et al. Development of human T-cell leukemia virus type 1-transformed tumors in rats following suppression of T-cell immunity by CD80 and CD86 blockade. J Virol 2000;74:428–435.

(14)

73. Kannagi M, Ohashi T, Hanabuchi S, et al. Immunological aspects of rat models of HTLV type 1-infected T lympho- proliferative disease. AIDS Res Hum Retroviruses 2000;16:1737–1740.

74. Ohya O, Tomaru U, Yamashita I, et al. HTLV-I induced myeloneuropathy in WKAH rats: apoptosis and local activa- tion of the HTLV-I pX and TNF-alpha genes implicated in the pathogenesis. Leukemia 1997;11(Suppl 3):255–7, 255–257.

75. Seto K, Abe M, Ohya O, et al. A rat model of HTLV-I infection: development of chronic progressive myeloneuropathy in seropositive WKAH rats and related apoptosis. Acta Neuropathol 1995;89:483–490.

76. Kushida S, Mizusawa H, Matsumura M, et al. High incidence of HAM/TSP-like symptoms in WKA rats after administration of human T-cell leukemia virus type 1-producing cells. J Virol 1994;68:7221–7226.

77. Yoshiki T. Chronic progressive myeloneuropathy in WKAH rats induced by HTLV-I infection as an animal model for HAM/TSP in humans. Intervirology 1995;38:229–237.

78. Nerenberg M, Xu X, Brown DA. Transgenic models of HTLV-I mediated disease and latency. Curr Top Microbiol Immunol 1996;206:175–196.

79. Nerenberg M, Hinrichs SH, Reynolds RK, Khoury G, Jay G. The tat gene of human T-lymphotropic virus type 1 induces mesenchymal tumors in transgenic mice. Science 1987;237:1324–1329.

80. Xu X, Heidenreich O, Kitajima I, et al. Constitutively activated JNK is associated with HTLV-1 mediated tumorige- nesis. Oncogene 1996;13:135–142.

81. Kitajima I, Shinohara T, Bilakovics J, Brown DA, Xu X, Nerenberg M. Ablation of transplanted HTLV-I tax-trans- formed tumors in mice by antisense inhibition of NF-kappa B. Science 1993;259:1523.

82. Coscoy L, Gonzalez-Dunia D, Chirinian-Swan S, Brahic M, Ozden S. Analysis of the expression directed by two HTLV-I promoters in transgenic mice. J Neurovirol 1996;2:336–344.

83. Green J, Begley C, Wagner D, Waldmann TA, Jay G. Trans-activation of granulocyte macrophage colony-stimulating factor and the interleukin 2 receptor in transgenic mice carrying the HTLV-I tax gene. Mol Cell Biol 1989;9:4731–4737.

84. Green JE. Trans activation of nerve growth factor in transgenic mice containing the human T-cell lymphotropic virus type I tax gene. Mol Cell Biol 1997;11:4635–4641.

85. Iwakura Y. Roles of IL-1 in the development of rheumatoid arthritis: consideration from mouse models. Cytokine Growth Factor Rev 2002;13:341–355.

86. Grossman WJ, Ratner L. Transgenic mouse models for HTLV-I infection. J Acquir Immune Defic Syndr Hum Retrovirol 1996;13(Suppl 1):S162–S169.

87. Ozden S, Coscoy L, Gonzalez-Dunia D. HTLV-I transgenic models: an overview. J Acquir Immune Defic Syndr Hum Retrovirol 1996;13(Suppl 1):S154–S161.

88. Kaplan JE, Osame M, Kubota H, et al. The risk of development of HTLV-I-associated myelopathy/tropical spastic paraparesis among persons infected with HTLV-I. J Acq Immun Defic Synd 1990;3:1096–1101.

89. Bangham CR. Human T-lymphotropic virus type 1 (HTLV-1): persistence and immune control. Int J Hematol 2003;78:297–303.

90. Bangham CR. The immune control and cell-to-cell spread of human T-lymphotropic virus type 1. J Gen Virol 2003;84(Pt 12):3177–3189.

91. Yamano Y, Nagai M, Brennan M, et al. Correlation of human T-cell lymphotropic virus type 1 (HTLV-1). mRNA with proviral DNA load, virus-specific CD8(+) T cells, and disease severity in HTLV-1-associated myelopathy (HAM/TSP). Blood 2002;99:88–94.

92. Kitze B, Usuku K. HTLV-1-mediated immunopathological CNS disease. Curr Top Microbiol Immunol 2002;265:197–211.

93. Hashimoto K, Higuchi I, Osame M, Izumo S. Quantitative in situ PCR assay of HTLV-1 infected cells in peripheral blood lymphocytes of patients with ATL, HAM/TSP and asymptomatic carriers. J Neurol Sci 1998;159:67–72.

94. Albrecht B, Collins ND, Newbound GC, Ratner L, Lairmore MD. Quantification of human T-cell lymphotropic virus type 1 proviral load by quantitative competitive polymerase chain reaction. J Virol Meth 1998;75:123–140.

95. Nagai M, Usuku K, Matsumoto W, et al. Analysis of HTLV-I proviral load in 202 HAM/TSP patients and 243 asymptomatic HTLV-I carriers: high proviral load strongly predisposes to HAM/TSP. J Neurovirol 1998;4:586–593.

96. Jeffery KJ, Usuku K, Hall SE, et al. HLA alleles determine human T-lymphotropic virus-I (HTLV-I) proviral load and the risk of HTLV-I-associated myelopathy. Proc Nat Acad Sci U S A 1999;96:3848–3853.

97. Jeffery KJ, Siddiqui AA, Bunce M, et al. The influence of HLA class I alleles and heterozygosity on the outcome of human T cell lymphotropic virus type I infection. J Immunol 2000;165:7278–7284.

98. Yashiki S, Fujiyoshi T, Arima N, et al. HLA-A*26, HLA-B*4002, HLA-B*4006, and HLA-B*4801 alleles predispose to adult T cell leukemia: the limited recognition of HTLV type 1 tax peptide anchor motifs and epitopes to generate anti-HTLV type 1 tax CD8(+) cytotoxic T lymphocytes. AIDS Res Hum Retroviruses 2001;17:1047–1061.

99. Usuku K, Nishizawa M, Matsuki K, et al. Association of a particular amino acid sequence of the HLA-DR β1 chain with HTLV-I-associated myelopathy. Eur J Immunol 1990;20:1603–1606.

(15)

100. Jacobson S, Shida H, Mcfarlin DE, Fauci AS, Koenig S. Circulating CD8+ cytotoxic T lymphocytes specific for HTLV-I pX in patients with HTLV-I associated neurological disease. Nature 1990;348:245–248.

101. Kannagi M, Shida H, Igarashi H, et al. Target epitope in the tax protein of HTLV-I recognized by class I MHC- restricted cytotoxic T cells. J Virol 1992;66:2928–2933.

102. Vine AM, Witkover AD, Lloyd AL, Jeffery KJ, Siddiqui A, Marshall SE et al. Polygenic control of human T lym- photropic virus type I (HTLV-I) provirus load and the risk of HTLV-I-associated myelopathy/tropical spastic para- paresis. J Infect Dis 2002;186:932–939.

103. Daenke S, Nightingale S, Cruickshank JK, Bangham CRM. Sequence variants of human T-cell lymphotropic virus type I from patients with tropical spastic paraparesis and adult T-cell leukemia do not distinguish neurological from leukemic isolates. J Virol 1990;64:1278–1282.

104. Evangelista A, Maroushek S, Minnigan H, et al. Nucleotide sequence analysis of a provirus derived from an individ- ual with tropical spastic paraparesis. Microb Pathog 1990;8:259–278.

105. Gould KG, Bangham CR. Virus variation, escape from cytotoxic T lymphocytes and human retroviral persistence.

Semin Cell Dev Biol 1998;9:321–328.

106. Mahieux R, Ibrahim F, Mauclere P, et al. Molecular epidemiology of 58 new African human T-cell leukemia virus type 1 (HTLV-1) strains: identification of a new and distinct HTLV-1 molecular subtype in central Africa and in pygmies.

J Virol 1997;71:1317–1333.

107. Saito M, Furukawa Y, Kubota R, et al. Frequent mutation in pX region of HTLV-1 is observed in HAM/TSP patients, but is not specifically associated with the central nervous system lesions. J Neurovirol 1995;1:286–294.

108. Kira JI. The presence of HTLV-I proviral DNA in the central nervous system of patients with HTLV-I-associated myelopathy/tropical spastic paraparesis. Mol Neurobiol 1994;8:139–145.

109. Furukawa Y, Yamashita M, Usuku K, Izumo S, Nakagawa M, Osame M. Phylogenetic subgroups of human T cell lymphotropic virus (HTLV) type I in the tax gene and their association with different risks for HTLV-I-associated myelopathy/tropical spastic paraparesis. J Infect Dis 2000;182(5):1343–1349.

110. Nakane S, Shirabe S, Moriuchi R, et al. Comparative molecular analysis of HTLV-I proviral DNA in HTLV-I infected members of a family with a discordant HTLV-I-associated myelopathy in monozygotic twins. J Neurovirol 2000;6:275–283.

111. Furukawa Y, Kubota R, Tara M, Izumo S, Osame M. Existence of escape mutant in HTLV-I tax during the development of adult T-cell leukemia. Blood 2001;97:987–993.

112. Renjifo B, Borrero I, Essex M. Tax mutation associated with tropical spastic paraparesis human T-cell leukemia virus type I-associated myelopathy. J Virol 1995;69:2611–2616.

113. Mahieux R, de The G, Gessain A. The tax mutation at nucleotide 7959 of human T-cell leukemia virus type 1 (HTLV-1) is not associated with tropical spastic paraparesis/HTLV-1-associated myelopathy but is linked to the cos- mopolitan molecular genotype. J Virol 1995;69:5925–5927.

114. Johnson JM, Mulloy JC, Ciminale V, Fullen J, Nicot C, Franchini G. The MHC class I heavy chain is a common target of the small proteins encoded by the 3´ end of HTLV type 1 and HTLV type 2. AIDS Res Hum Retroviruses 2000;16:1777–1781.

115. Johnson JM, Nicot C, Fullen J, et al. Free major histocompatibility complex class I heavy chain is preferentially tar- geted for degradation by human T-cell leukemia/lymphotropic virus type 1 p12(I) protein. J Virol 2001;75:

6086–6094.

116. Dantuma NP, Masucci MG. The ubiquitin/proteasome system in Epstein-Barr virus latency and associated malignancies. Semin Cancer Biol 2003;13:69–76.

117. Laney JD, Hochstrasser M. Substrate targeting in the ubiquitin system. Cell 1999;97:427–430.

118. Varshavsky A. The ubiquitin system. Trends Biochem Sci 1997;22:383–387.

119. Trovato R, Mulloy JC, Johnson JM, Takemoto S, de Oliveira MP, Franchini G. A lysine-to-arginine change found in natural alleles of the human T-cell lymphotropic/leukemia virus type 1 p12(I) protein greatly influences its stability.

J Virol 1999;73:6460–6467.

120. Franchini G. Molecular mechanisms of human T-cell leukemia/lymphotropic virus type I infection. Blood 1995;86:3619–3639.

121. Martins ML, Soares BC, Ribas JG, et al. Frequency of p12K and p12R alleles of HTLV type 1 in HAM/TSP patients and in asymptomatic HTLV type 1 carriers. AIDS Res Hum Retroviruses 2002;18:899–902.

122. Kakuda K, Ikematsu H, Chong WL, Hayashi J, Kashiwagi S. Molecular epidemiology of human T lymphotropic virus type 1 transmission in Okinawa, Japan. Am J Trop Med Hyg 2002;66:404–408.

123. Osame M, Janssen R, Kubota H, et al. Nationwide survey of HTLV-I-associated myelopathy in Japan: association with blood transfusion. Ann Neurol 1990;28:50–56.

124. Lin BT, Musset M, Szekely AM, et al. Human T-cell lymphotropic virus-1-positive T-cell leukemia/lymphoma in a child: report of a case and review of the literature. Arch Pathol Lab Med 1997;121:1282–1286.

125. Kato H, Koya Y, Ohashi T, et al. Oral administration of human T-cell leukemia virus type 1 induces immune unre- sponsiveness with persistent infection in adult rats. J Virol 1998;72:7289–7293.

Riferimenti

Documenti correlati

In order to define the subcellular localization of endogenous HBZ in HAM/TSP, PBMC of four HAM/TSP patients, namely PH1485, PH1593, PH1601 and PH1624 were studied

Figure 23: Expression and localization of endogenous HBZ in HTLV-1 chronically infected and in ATL tumor cells C5MJ HTLV-1 chronically infected, ATL-2s ATL tumor cell line and

Major histocompatibility complex class II transactivator CIITA is a viral restriction factor that targets human T-cell lymphotropic virus type 1 Tax-1 function and inhibits

Besides this well known function, we recently found that CIITA acts as an endogenous restriction factor against HTLV-1 (human T cell lymphotropic virus type 1) and HTLV-2

Two adjacent regions (1-252 and 253-410) of CIITA bound independently to Tax-1, but only region 1-252 mediated Tax-1 inhibition, in agree- ment with the fact that CIITA residues

Moreover the bind- ing of endogenous NF-YB with Tax-2 molecules was increased by the presence of CIITA, strongly suggesting that at least a trimeric NF-YB-CIITA-Tax-2 complex is

In this study, we showed that T lymphocytes proteome changes may be a valid measure both to predict the disease state in human subjects and to track progression

The levels of expression of three major classes of HTLV-2 mRNAs were measured at different time points in BJAB-Gu infected human B-cell line and in IL-2 stimulated