Next Article in Journal
Advances and Future Challenges in Recombinant Adenoviral Vectored H5N1 Influenza Vaccines
Previous Article in Journal
Comparative Genomics of Korean Infectious Bronchitis Viruses (IBVs) and an Animal Model to Evaluate Pathogenicity of IBVs to the Reproductive Organs
Previous Article in Special Issue
Renal Alterations in Feline Immunodeficiency Virus (FIV)-Infected Cats: A Natural Model of Lentivirus-Induced Renal Disease Changes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Aspects of Feline Retroviruses: A Review

Medizinische Kleintierklinik, LMU University of Munich, Germany, Veterinaerstrasse 13, 80539 Munich, Germany
Viruses 2012, 4(11), 2684-2710; https://doi.org/10.3390/v4112684
Submission received: 12 October 2012 / Revised: 24 October 2012 / Accepted: 26 October 2012 / Published: 31 October 2012
(This article belongs to the Special Issue Feline Retroviruses)

Abstract

:
Feline leukemia virus (FeLV) and feline immunodeficiency virus (FIV) are retroviruses with global impact on the health of domestic cats. The two viruses differ in their potential to cause disease. FeLV is more pathogenic, and was long considered to be responsible for more clinical syndromes than any other agent in cats. FeLV can cause tumors (mainly lymphoma), bone marrow suppression syndromes (mainly anemia), and lead to secondary infectious diseases caused by suppressive effects of the virus on bone marrow and the immune system. Today, FeLV is less commonly diagnosed than in the previous 20 years; prevalence has been decreasing in most countries. However, FeLV importance may be underestimated as it has been shown that regressively infected cats (that are negative in routinely used FeLV tests) also can develop clinical signs. FIV can cause an acquired immunodeficiency syndrome that increases the risk of opportunistic infections, neurological diseases, and tumors. In most naturally infected cats, however, FIV itself does not cause severe clinical signs, and FIV-infected cats may live many years without any health problems. This article provides a review of clinical syndromes in progressively and regressively FeLV-infected cats as well as in FIV-infected cats.

1. Introduction

Feline leukemia virus (FeLV) and feline immunodeficiency virus (FIV) belong to the most common infectious diseases in cats. Both are retroviruses, but FeLV is a γ-retrovirus, while FIV is classified as a lentivirus. Although FeLV and FIV are closely related, they differ in their potential to cause disease. In the United States, prevalence of both infections is about 2% in healthy cats and up to about 30% in high-risk or sick cats [1,2]. Risk factors for infection include male gender, adulthood, and outdoor access [3,4]. Retroviral tests can diagnose only infection, not clinical disease.
FeLV is more pathogenic than FIV. For a long time, FeLV was considered to account for most disease-related deaths, and to be responsible for more clinical syndromes than any other single agent in cats. It was proposed that approximately one third of all tumor-related deaths in cats were caused by FeLV, and an even greater number of cats died of FeLV-related anemia and secondary infections caused by suppressive effects of the virus on bone marrow and the immune system. Today, these statements have to be revised, as in recent years the prevalence and consequently the importance of FeLV as a pathogen in cats have been decreasing. Still, if present in closed households with other viruses, such as feline coronavirus (FCoV), or FIV, FeLV infection has the greatest impact on survival [5]. The death rate of progressively FeLV-infected cats in multi-cat households has been estimated at approximately 50% in two years and 80% in three years [6,7], but is much lower today, at least for cats that are well taken care of and that are kept strictly indoors in single-cat households. A survey in the United States compared the survival of more than 1000 FeLV-infected cats to more than 8000 age- and sex-matched uninfected control cats and found that in FeLV-infected cats median survival was 2.4 years compared to 6.0 years for control cats [2]. Despite the fact that progressive FeLV infection is associated with a decrease in life expectancy, many owners elect to provide treatment for their cats, and with proper care, FeLV-infected cats might live for many years with good quality of life.
Although FIV can cause an acquired immunodeficiency syndrome in cats (“feline AIDS”) comparable to human immunodeficiency virus (HIV) infection in humans, with increased risk for opportunistic infections, neurologic diseases, and tumors, in most naturally infected cats, FIV does not cause a severe clinical syndrome. With proper care, FIV-infected cats can live many years and, in fact, can die at older age from causes unrelated to their FIV infection. In a follow-up study in naturally FIV-infected cats, the rate of progression was variable, with death occurring in about 18% of infected cats within the first two years of observation (about five years after the estimated time of infection). An additional 18% developed increasingly severe disease, but more than 50% remained clinically asymptomatic during the two years [8]. FIV infection has little impact on a cat population and does not reduce the number of cats in a household [2]. Thus, overall survival time is not shorter than in uninfected cats, and quality of life is usually fairly high over an extended period of time.

2. Stages of Infection

Both infections are chronic in nature and develop through different disease stages. Characteristically for both infections, there is a long asymptomatic phase, in which cats do not show clinical signs.
FeLV infection has different stages. Recently, novel diagnostic tools, including very sensitive PCR methods providing new data on the course of FeLV infection, have questioned the traditional understanding of FeLV pathogenesis. Cats believed to be immune to FeLV after infection were found to remain provirus-positive. Antigen-negative, provirus-positive cats are frequently detected and their clinical relevance and role in FeLV epidemiology is still not fully understood. Antigen-negative, provirus-positive cats are considered FeLV carriers. Following reactivation, they can act as an infection source. As FeLV provirus is integrated into the cat’s genome, it is unlikely to be fully cleared over time. Antigen-negative, provirus-positive cats do not shed the virus, but reactivation with reoccurring virus shedding is possible [9,10]. Based on this information, a new classification has been proposed, in which the stages of FeLV infection are defined as abortive infection (comparable to the former “regressor cats”), regressive infection (comparable to the former “transient viremia” followed by “latent infection”), progressive infection (comparable to the former “persistent viremia”), and focal or atypical infection (Table 1) [11,12,13,14].
Abortive infection. After infection, the virus starts initially to replicate in the local lymphoid tissue in the oropharyngeal area. In some immunocompetent cats (formerly called “regressor cats)”, viral replication may be terminated by an effective humoral and cell-mediated immune response; these cats never become viremic. They have high levels of neutralizing antibodies. Neither FeLV antigen nor viral RNA or proviral DNA can be detected in the blood at any time. Abortive infection is likely caused when a cat is exposed to low doses of FeLV [15]. It is still unknown, how often this situation really occurs in nature, because studies using very sensitive PCR methods have found that in many of the formerly considered “regressor cats”, the virus actually can still be found later in these cats when investigating tissue samples. Thus, it appears likely that none or only very few cats can completely clear FeLV infection from all cells.
Regressive infection develops following an effective immune response. In regressive infection, virus replication and viremia are contained prior to or shortly after bone marrow infection. After initial infection, replicating FeLV spreads systemically through infected mononuclear cells (lymphocytes and monocytes). During this stage, cats have positive results on tests that detect free antigen in plasma (e.g., ELISA). They shed virus, mainly with saliva. In cats with regressive infection, this viremia, however, is terminated within weeks or months (therefore formerly called “transient viremia”). In some cats, viremia may persist longer than three weeks. After about three weeks of viremia, bone marrow cells become infected, and infected hematopoietic precursor cells develop into infected granulocytes and platelets that circulate in the body. Even if bone marrow cells become infected, a certain percentage of cats is able to clear viremia. However, they cannot completely eliminate the virus from the body, even if they terminate viremia because the information for virus replication (proviral DNA) is present in bone marrow stem cells. This condition has been called “latent infection” (and is now a part of the regressive infection). The molecular basis of latency is the integration of a copy of the viral genome (provirus) into cellular chromosomal DNA. Although proviral DNA remains present within the cellular genome, no virus is actively produced. Thus, cats with regressive infection have negative results in all tests that detect FeLV antigen. During cell division, proviral DNA is replicated and the information given to the daughter cells. Thus, complete cell lineages may contain FeLV proviral DNA. However, proviral DNA is not translated into proteins, and no infectious virus particles are produced. Therefore, regressively infected cats do not shed FeLV and are not infectious to others. Sensitive PCR methods can detect provirus in the blood of cats with regressive infection that are antigen-negative. In a study in Switzerland it was shown that in addition to the antigen-positive, provirus-positive cats, about 10% of the cat population that were negative for antigen were positive for proviral DNA in the blood [16]. Regressive infection can be reactivated because the information for producing complete viral particles is present and can potentially be reused when antibody production decreases (e.g., after immunosuppression).
In cats with progressive infection, FeLV infection is not contained early in the infection. Thus, extensive virus replication occurs, first in the lymphoid tissues, followed by the bone marrow and mucosal and glandular epithelial tissues. Progressively infected cats remain persistently viremic. They are infectious to other cats for the remainder of their life. This condition has been called “persistent viremia” and is now classified as progressive infection. Cats with progressive infection develop FeLV-associated diseases, and most of them will die within a few years. Regressive and progressive infections can be distinguished by repeated testing for viral antigen in peripheral blood; regressively infected cats will turn negative at latest 16 weeks after infection, while progressively infected cats will remain positive. Initially both, regressive and progressive infections are accompanied by persistence of FeLV proviral DNA in the blood detected by PCR but later are associated with different FeLV loads when measured by quantitative PCR; regressive infection is associated with low, progressive infection with high virus load [11,17].
Focal infections or atypical infections have been reported in up to 10% of experimentally infected cats. Focal infections or atypical infections may also be observed in natural infections, but are probably rare in the field. Focal infections are characterized by a persistent atypical local viral replication (e.g., in mammary glands, bladder, eyes). This replication can lead to intermittent or low-grade production of antigen, and therefore, these cats can have weakly positive or discordant results in antigen tests, or positive and negative results may alternate [14].
Table 1. Stages of feline leukemia virus (FeLV)infection
Table 1. Stages of feline leukemia virus (FeLV)infection
Stages of FeLV infection FeLV p27 antigen in blood Virus blood culture Viral RNA in blood Viral DNA in blood Viral tissue culture Viral shedding FeLV-associated disease
Progressive Positive Positive Positive Positive Positive Positive Likely
Regressive Negative Negative Negative Positive Negative Negative Unlikely
Abortive Negative Negative Negative Negative Negative Negative Unlikely
Focal Variable Variable Variable Variable Positive Variable Unlikely
Experimental FIV infection also progresses through several stages, similar to HIV infection in people, including an acute phase, a clinically asymptomatic phase of variable duration, and a terminal phase sometimes called “feline acquired immunodeficiency syndrome” (“AIDS”) [18,19]. However, there is no clear distinction between these stages in naturally FIV-infected cats, and not all stages are apparent; therefore, the usefulness of this staging in natural FIV infection has been questioned. Moreover, even cats in moribund condition with severe immunosuppression and secondary infections may fully recover with appropriate care and return to an asymptomatic stage. Thus, different from HIV-infected people, cats classified as being in the “AIDS phase” (high virus load, severe clinical signs due to secondary infection) can recover and be asymptomatic again, and their virus loads can even decrease dramatically.

3. Clinical Signs

Clinical signs in both retrovirus infections are variable. After a long asymptomatic phase, cats can develop tumors, hematopoietic disorders, neurologic disorders, immunodeficiency, immune-mediated diseases, and stomatitis. The pathomechanism of these disorders is different in both retrovirus infections (Table 2).
Although FeLV was named after a tumor that first garnered its attention, most infected cats are presented to the veterinarian not for tumors but for anemia or immunosuppression. Clinical signs associated with FeLV infection can be classified as tumors, immunosuppression, hematologic disorders, immune-mediated diseases, and other syndromes (including neuropathy, reproductive disorders, fading kitten syndrome). Of 8642 FeLV-infected cats presented to North American Veterinary Teaching Hospitals, various co-infections (including FIV infection, feline infectious peritonitis (FIP), upper respiratory infection, hemotropic mycoplasmosis, and stomatitis) were the most frequent findings (15%), followed by anemia (11%), lymphoma (6%), leukopenia or thrombocytopenia (5%), and leukemia or myeloproliferative diseases (4%) [20]. The outcome of FeLV infection and the clinical course are determined by a combination of viral and host factors. Some of the differences in outcome can be traced to properties of the virus itself, such as the subgroup that determines differences in the clinical picture (e.g., FeLV-B is primarily associated with tumors, FeLV-C is primarily associated with non-regenerative anemia). A study aiming to define dominant host immune effects or mechanisms responsible for the outcome of infection by using longitudinal changes in FeLV-specific cytotoxic T-lymphocytes (CTL) found that high levels of circulating FeLV-specific effector CTLs appear before virus-neutralizing antibodies in cats that have recovered from exposure to FeLV. In contrast, progressive infection with persistent viremia has been associated with a silencing of virus-specific humoral and cell-mediated immunity host effector mechanisms [21]. Probably the most important host factor that determines the clinical outcome of cats infected with FeLV is the age of the cat at the time of infection [22]. Neonatal kittens develop marked thymic atrophy after infection (“fading kitten syndrome”), resulting in severe immunosuppression, wasting, and early death. As cats mature, they acquire a progressive resistance. When older cats become infected, they tend to have abortive or regressive infections or, if developing progressive infection, have at least milder signs and a more protracted period of apparent good health [7].
Clinical signs in naturally FIV-infected cats usually reflect secondary diseases, such as infections and neoplasia, to which FIV-infected cats are considered more susceptible. FIV itself may cause some clinical features (e.g., neurologic signs) resulting from abnormal function or inflammation of affected organs. In experimental infection, an initial stage is sometimes noticed usually with transient and mild clinical signs, including fever, lethargy, signs of enteritis, stomatitis, dermatitis, conjunctivitis, respiratory tract disease, and generalized lymph node enlargement [23]. The acute phase may last several days to a few weeks, after which cats will enter a period in which they appear clinically healthy. This phase is usually not noticed by the owners in naturally infected cats. The duration of the following asymptomatic phase varies, but usually lasts many years. Factors that influence the duration of the asymptomatic phase include the pathogenicity of the infecting isolate (also depending on the FIV subtype), exposure to secondary pathogens, and the age of the cat at the time of infection [24,25]. In the last, symptomatic phase (“AIDS phase”) of infection, the clinical signs are a reflection of opportunistic infections, neoplasia, myelosuppression, and neurologic disease.

3.1. Tumors

While FeLV-infected cats are 62-times more likely to develop lymphoma or leukemia than non-infected cats and FeLV plays a direct role in tumorgenesis, FIV-infected cats have about a five-fold increased risk of tumor development, and the role of FIV is usually indirect. Lymphomas are the most common tumors in FeLV- and FIV-infected cats. While FeLV-infected cats have most commonly T-cell lymphomas, lymphomas in FIV are mostly of B-cell origin [26,27].
FeLV is a major oncogene that causes different tumors in cats, most commonly lymphoma and leukemia, less often other hematopoietic tumors and rarely other malignancies (including neuroblastoma, osteochondroma, and others). The association between FeLV and lymphomas has been clearly established in several ways. First, these malignancies can be induced in kittens by experimental FeLV infection [28,29,30]. Second, cats naturally infected with FeLV have a higher risk of developing lymphoma than uninfected cats [29,31]. Third, most cats with lymphoma were—at least in earlier times when prevalence of FeLV was still higher—FeLV-positive in tests that detected infectious virus or FeLV antigens. Previously, up to 80% of feline lymphomas and leukemias were reported to be FeLV-related [32,33,34,35,36,37,38]. However, since the 1980s a reduction in the prevalence of viremia has been noted in cats with lymphoma [39,40,41]. The decrease in prevalence of FeLV infection in cats with lymphoma or leukemia also indicates a shift in tumor causation in recent years. Whereas 59% of all cats with lymphoma or leukemia were FeLV antigen-positive in one German study from 1980 to 1995, only 20% of the cats were FeLV antigen-positive in the years 1996 to 1999 in the same University Teaching Hospital [41]. In a recent study in the Netherlands, only four of 71 cats with lymphoma were FeLV-positive, although 22 of these cats had mediastinal lymphoma, which previously was strongly associated with FeLV infection [42]. A greater prevalence of lymphoma in older-age cats in now observed. The major reason for the decreasing association of FeLV with lymphoma is the decreasing prevalence of FeLV infection in the overall cat population as a result of FeLV vaccination as well as testing and elimination programs. However, prevalence of lymphomas caused by FeLV may be higher than indicated by conventional antigen testing of blood [43]. Cats from FeLV cluster households had a 40-fold higher rate of development of FeLV-negative lymphoma than did those from the general population. FeLV-negative lymphomas have also occurred in laboratory cats known to have been infected previously with FeLV [44]. FeLV proviral DNA was detected in lymphomas of cats that tested negative for FeLV antigen [43], also suggesting that the virus may be associated with a larger proportion of lymphomas than previously thought. FeLV has been shown to incorporate cellular genes; several such transducted genes also present in regressively infected cells have been implicated in viral oncogenesis [44,45,46]. It is still unclear, how common regressive FeLV infection is responsible for FeLV-associated tumors in the field as study results have been controversial. Proviral DNA was detected in formalin-fixed, paraffin-embedded tumor tissue in 7/11 FeLV-negative cats with lymphoma [43]. However, other groups found evidence of provirus in only 1/22 [45] and in 0/50 FeLV antigen-negative lymphomas [47].
The most important mechanism by which FeLV causes malignancy is by insertion of the FeLV genome into the cellular genome near a cellular oncogene (most commonly myc), resulting in activation and over-expression of that gene. These effects lead to uncontrolled proliferation of these cells (clone). A malignancy results in the absence of an appropriate immune response. FeLV may also incorporate the oncogene to form a recombinant virus (e.g., FeLV-B, FeSV) containing cellular oncogene sequences that are then rearranged and activated. When they enter a new cell, these recombinant viruses are oncogenic. In a study of 119 cats with lymphomas, transduction or insertion of the myc locus had occurred in 38 cats (32%) [48]. Thus, FeLV-induced neoplasms are caused, at least in part, by somatically acquired insertional mutagenesis in which the integrated provirus may activate a proto-oncogene or disrupt a tumor suppressor gene. A recent study suggested that the U3-LTR region of FeLV transactivates cancer-related signaling pathways through production of a non-coding 104 base RNA transcript that activates NF kappaB [49]. Twelve common integration sites for FeLV associated with lymphoma development have been identified in six loci: c-myc, flvi-1, flvi-2 (contains bmi-1), fit-1, pim-1, and flit-1. Oncogenic association of the loci is based on the fact that c-myc is known as a proto-oncogene, bmi-1 and pim-1 have been recognized as myc-collaborators, fit-1 appears to be closely linked to myb, and flit-1 insertion was shown to be associated with over-expression of cellular genes, e.g., activin-A receptor type II-like 1 (ACVRL1). [50]. Flit-1 seems to have an important role in the development of lymphomas and appears to represent a common novel FeLV proviral integration domain that may influence lymphomagenesis by insertional mutagenesis. Among 35 FeLV-related tumors, 5/25 thymic lymphomas demonstrated proviral insertion within flit-1 locus, whereas 0/4 alimentary lymphomas, 5/5 multicentric lymphomas, and 1/1 T-lymphoid leukemia examined had rearrangements in this region. Expression of ACVRL1 mRNA was detected in the two thymic lymphomas with flit-1 rearrangement, whereas normal thymuses and seven lymphoid tumors without flit-1 rearrangement had no detectable ACVRL1 mRNA expression [51].
Fibrosarcomas that are associated with FeLV are caused by FeSV, a recombinant virus that develops de novo in FeLV-A-infected cats by recombination of the FeLV-A genome with cellular oncogenes. Through a process of genetic recombination, FeSV acquires one of several oncogenes, such as fes, fms, or fgr. As a result, FeSV is an acutely transforming (tumor-causing) virus, leading to a polyclonal malignancy with multifocal tumors arising simultaneously after a short incubation period. With the decrease in FeLV prevalence, FeSV also has become less common. FeSV-induced fibrosarcomas are multicentric and usually occur in young cats. Strains of FeSV identified from naturally occurring tumors are defective and unable to replicate without the presence of FeLV-A as a helper virus that supplies proteins (such as those coded by the env gene) to FeSV. Fibrosarcomas caused by FeSV tend to grow rapidly, often with multiple cutaneous or subcutaneous nodules that are locally invasive and metastasize to the lung and other sites. Solitary fibrosarcomas in older cats are not caused by FeSV. These tumors are slower growing, locally invasive, slower metastasizing, and only occasionally curable by excision combined with radiation and/or gene therapy. They usually are classified as feline injection site sarcomas (FISS) caused by the granulomatous inflammatory reaction at the injection site, commonly occurring after inoculation of adjuvant-containing vaccines. It has been demonstrated that neither FeSV nor FeLV play any role in the development of FISS [52].
A few other tumors have been found in FeLV-infected cats; some of them might have an association with FeLV, others likely have just been observed by chance simultaneously in an infected cat. Iris melanomas, for example, are not associated with FeLV infections, although in one study three of 18 eyes tested positive for FeLV/FeSV proviral DNA [53]. In a more recent study, however, immunohistochemical staining and PCR did not find FeLV or FeSV in the ocular tissues of any cat with this disorder [54]. Multiple osteochondromas (cartilaginous exostoses on flat bones of unknown pathogenesis) have been described in FeLV-infected cats. Although histologically benign, they may cause significant morbidity if they occur in an area such as a vertebra and put pressure on the spinal cord or nerve roots [55,56]. In spontaneous feline olfactory neuroblastomas (aggressive, histologically inhomogenous tumors of the tasting and smelling epithelium of nose and pharynx with high metastasis rates), budding FeLV particles were found in the tumors and lymph node metastases, and FeLV DNA was detected in tumor tissue [57]. The exact role of FeLV in the genesis of these tumors is uncertain. Cutaneous horns are a benign hyperplasia of keratinocytes that have been described in FeLV-infected cats [58], but the role of FeLV is also unclear.
FIV-infected cats are about five times more likely to develop lymphoma or leukemia than non-infected cats [26,27]. Lymphomas (mostly B-cell lymphomas) [26,27,59,60], leukemias, but also several other tumors have been described in association with FIV infection [26,61,62,63,64,65,66], including squamous cell carcinoma, fibrosarcoma, and mast cell tumor. FIV provirus, however, is only occasionally detected in tumor cells [67,68,69,70], suggesting a more indirect role in lymphoma formation, such as decreased cell-mediated immune surveillance or chronic B-cell hyperplasia [68,71]. However, clonally integrated FIV DNA was found in lymphoma cells from one cat that had been experimentally infected six years earlier, indicating the possibility of an occasional direct oncogenic role of FIV [67,70,72]. The prevalence of FIV infection in one cohort of cats with lymphoma was 50% [60], much higher than the FIV prevalence in the population of cats without lymphomas, which is also supportive of a cause and effect relationship. FIV could alternatively increase tumor incidence by decreasing tumor immunosurveillance mechanisms. It also could promote tumor development through the immunostimulatory effects of replicating in lymphocytes.

3.2. Myelosuppression

Myelosupression and other hematopoietic disorders can occur in both, FeLV and FIV infection. It is, however, much more common and more severe in FeLV-infected cats.
Hematologic changes described in association with FeLV include anemia (non-regenerative or regenerative), persistent, transient, or cyclic neutropenia, platelet abnormalities (thrombocytopenia and platelet function abnormalities), aplastic anemia (pancytopenia), and panleukopenia-like syndrome. For the majority of pathogenic mechanisms in which FeLV causes bone marrow suppression, active virus replication is required. However, it has been demonstrated that in some FeLV antigen-negative cats, regressive FeLV infection without viremia may be responsible for bone marrow suppression. In a recent study including 37 cats with myelosuppression that tested FeLV antigen-negative in peripheral blood, 2/37 cats (5%) were found regressively infected with FeLV by bone marrow PCR (both had non-regenerative anemia) [73]. In these regressively infected cats, FeLV provirus may interrupt or inactivate cellular genes in the infected cells, or regulatory features of viral DNA may alter expression of neighboring genes. Additionally, cell function of provirus-containing myelomonocytic progenitor and stromal fibroblasts that provide bone marrow microenvironment may be altered. Alternatively, FeLV provirus may cause bone marrow disorders by inducing the expression of antigens on the cell surface, resulting in an immune-mediated destruction of the cell. Anemia is a major non-neoplastic complication that occurs in a majority of FeLV-infected cats [4]. Anemia in FeLV-infected cats may have various causes. Approximately 10% of FeLV-associated anemias are regenerative [74], most FeLV-associated anemias, however, are non-regenerative and are caused by the bone marrow suppressive effect of the virus resulting from primary infection of hematopoietic stem cells and infection of stroma cells that constitute the supporting environment for hematopoietic cells. In vitro exposure of normal feline bone marrow to some strains of FeLV caused suppression of erythrogenesis [6]. In addition to the direct effect of the virus on erythropoiesis, other factors can cause non-regenerative anemia in FeLV-infected cats (e.g., anemia of chronic inflammation promoted by high concentration of cytokines). FeLV infection can cause decreased platelet counts. It also can be responsible for platelet function deficits, and the lifespan of platelets is shortened in some FeLV-infected cats. Thrombocytopenia (resulting in bleeding disorders) can occur secondary to decreased platelet production from FeLV-induced bone marrow suppression or leukemic infiltration. Platelets harbor FeLV, and megakaryocytes are frequent targets of progressive FeLV infection. Immune-mediated thrombocytopenia, which rarely occurs as a single disease entity in cats, often accompanies immune-mediated hemolytic anemia (IMHA) in cats with underlying FeLV infection. FeLV infection also can cause decreased neutrophil or lymphocyte counts. Neutropenia is common in FeLV-infected cats [75] and generally occurs alone or in conjunction with other cytopenias. In some cases, myeloid hypoplasia of all granulocytic stages is observed, suggesting infection on neutrophil precursors. In some neutropenic FeLV-infected cats, an arrest in bone marrow maturation can occur at the myelocyte and metamyelocyte stages. It has been hypothesized that an immune-mediated mechanism is responsible in cases in which neutrophil counts recover with glucocorticoid treatment (“glucocorticoid-responsive neutropenia”).
Hematopoietic neoplasia (“myeloprolifertaive disorders”), including leukemia, can also cause bone marrow suppression syndromes by crowding out. Myelodysplastic syndrome (MDS), characterized by peripheral blood cytopenias and dysplastic changes in the bone marrow, is a pre-stage of acute myeloic leukemia. It was found that changes of the LTR region of the FeLV genome (presence of three tandem direct 47-bp repeats in the upstream region of the enhancer (URE)) are strongly associated with the induction of MDS [76]. Myelofibrosis, another cause of bone marrow suppression, is a condition characterized by abnormal proliferation of fibroblasts resulting from chronic stimulation of the bone marrow, such as chronic bone marrow activity from hyperplastic or neoplastic regeneration caused by FeLV. In severe cases, the entire endosteum within the medullary cavity can be obliterated.
Feline panleukopenia-like syndrome (FPLS), also known as FeLV-associated enteritis (FAE) or myeloblastopenia, consists of severe leukopenia (< 3000 cells/μl) with enteritis and destruction of intestinal crypt epithelium that mimics feline panleukopenia caused by feline panleukopenia virus (FPV) infection. However, FPV antigen has been demonstrated by IFA in intestinal sections of cats that died from this syndrome after being experimentally infected with FeLV [77]. FPV was also demonstrated by electron microscopy despite negative FPV antigen tests. It appears that this syndrome might actually not be caused by FeLV itself, as previously thought, but by co-infection with FPV. The syndrome also has been referred to as FAE in cats with progressive FeLV infection because the clinical signs observed are usually gastrointestinal, including hemorrhagic diarrhea, vomiting, oral ulceration or gingivitis, anorexia, and weight loss [78,79]. It is still unclear whether all theses syndromes have the same origin and are simply caused by co-infection with FPV (and even modified life FPV vaccines have been discussed) or if they are caused by FeLV itself [77].
Although cytopenias caused by bone marrow suppression are a common finding in FeLV infection, these are rather uncommon in FIV-infected cats. During the acute phase of infection, FIV-infected cats can exhibit mild neutropenia, which resolves as the cat progresses to the asymptomatic phase of infection. Clinically ill FIV-infected cats in a later phase of infection may have a variety of cytopenias, with lymphopenia being most common. Lymphopenia is caused by direct replication of the virus in CD4+ lymphocytes. Anemia and neutropenia (usually mild) may also be seen [4,51], although these abnormalities may be as much a reflection of concurrent disease as direct effects of FIV itself. A recent study in a high number (3784) of client-owned field cats compared hematologic parameters in FIV-infected, FeLV-infected and uninfected cats [4]. Anemia and thrombocytopenia were not significantly more common in FIV-infected versus uninfected cats. Only neutropenia was significantly more often present, in about 25% of FIV-infected cats. Soluble factors have been shown to inhibit bone marrow function in FIV-infected cats, and bone marrow infection has been associated with decreased ability to support hematopoietic potential in vitro or has been proposed as a mechanism underlying the development of cytopenias [51].

3.3. Neurologic Dysfunction

Neurologic dysfunction may be present in FeLV- and in FIV-infected cats and is one of the few syndromes directly caused by the retrovirus. However, mechanisms of neurologic dysfunction are different with both viruses.
In FeLV-infected cats, most neurologic signs are caused by lymphoma and lymphocytic infiltrations in brain or spinal cord leading to compression, but in some cases, no tumor is detectable with diagnostic imaging methods or at necropsy. In these cats, FeLV-induced neurotoxicity is suspected. Anisocoria, mydriasis, central blindness, or Horner’s syndrome have been described in FeLV-infected cats without morphologic changes. In some regions (such as the southeastern United States), urinary incontinence caused by neuropathies in FeLV-infected cats has been described [80]. Direct neurotoxic effects of FeLV have been discussed as pathogenetic mechanisms. FeLV envelope glycoproteins may be able to produce increased free intracellular calcium leading to neuronal death (this has also been described in HIV-infected humans). A polypeptide of the FeLV envelope was found to cause dose-dependent neurotoxicity associated with alterations in intracellular calcium ion concentration, neuronal survival, and neurite outgrowth. The polypeptide from a FeLV-C strain was significantly more neurotoxic than the same peptide derived from a FeLV-A strain [81,82]. Neurologic signs in 16 cats with progressive FeLV infection consisted of abnormal vocalization, hyperesthesia, and paresis progressing to paralysis. Some cats developed anisocoria or urinary incontinence during the course of their illness. Others had concurrent FeLV-related problems such as myelodysplastic disease. The clinical course of affected cats involved gradually progressive neurologic dysfunction. Microscopically, white-matter degeneration with dilation of myelin sheaths and swollen axons was identified in the spinal cord and brain stem of affected animals [80]. Immunohistochemical staining of affected tissues revealed consistent expression of FeLV p27 antigens in neurons, endothelial cells, and glial cells, and proviral DNA was amplified from multiple sections of the spinal cord [80]. These findings suggest that in some FeLV-infected cats, the virus may directly affect CNS cells cytopathically.
Neurologic signs also have been described in both natural and experimental FIV infections [83,84,85,86,87,88]. About 5% of symptomatic FIV-infected cats have a neurological disease as a predominant clinical feature. Neurologic disorders in FIV infection seem to be strain-dependent [89]. Both central and peripheral neurologic manifestations have been described, comparable to the changes in HIV-infected human beings. Dementia in human patients with AIDS is often characterized by a slight decline in cognitive ability or behavior, changes that may be too subtle to be recognized in cats. Neurological abnormalities seen in naturally infected cats tend to be more behavioral than motor. Psychotic behavior, twitching movements of the face and tongue, compulsive roaming, dementia, loss of bladder and rectal control, and disturbed sleep patterns have been observed. Other signs described include nystagmus, ataxia, seizures, and intention tremors [90,91,92]. Abnormal forebrain electrical activity and abnormal visual and auditory-evoked potentials have also been documented in cats that appeared otherwise normal [24,66,93,94]. Although the majority of FIV-infected cats do not show clinically overt neurologic signs, a much higher proportion of infected cats have microscopic CNS lesions. Brain lesions may occur in the absence of massive infection, and abnormal neurologic function has been documented in FIV-infected cats with only mild to moderate histologic evidence of inflammation [8]. Pathologic findings include the presence of perivascular infiltrates of mononuclear cells, diffuse gliosis, glial nodules, and white matter pallor. These lesions are usually located in the caudate nucleus, midbrain, and rostral brain stem [8]. Mostly, abnormal neurologic function is the result of a direct effect of the virus on CNS cells. Neurologic signs upon FIV infection are highly strain-dependent. The virus infects the brain early, with virus-induced CNS lesions sometimes developing within two months of experimental infection [8]. Microglia and astrocytes are infected by FIV, but the virus does not infect neurons. However, neuronal death has been associated with FIV infection; in particular, forebrain signs are often a result of direct neuronal injury from the virus. The exact mechanism of neuronal damage by FIV is unclear but may include neuronal apoptosis, effects on the neuron supportive functions of astrocytes, toxic products released from infected microglia, or cytokines produced in response to viral infection. In vitro studies support the hypothesis that FIV infection may impair normal metabolism in CNS cells, particularly astrocytes [8]. Documented abnormalities of astrocyte function include altered intercellular communication, abnormal glutathione reductase activity that could render cells more susceptible to oxidative injury, and alterations in mitochondrial membrane potential that disrupt the energy-producing capacities of the cell [95]. Astrocytes are by far the most common cell type of the brain and are important in maintaining CNS neuronal vascular microenvironment. One of the most important functions of astrocytes is to regulate the level of extracellular glutamate, a major excitatory neurotransmitter that accumulates as a consequence of neuronal activity. Excessive extracellular glutamate often results in neuronal toxicity and death. FIV infection of feline astrocytes can significantly inhibit their glutamate-scavenging ability, potentially resulting in neuronal damage [95,96]. Sometimes, neurologic signs may also be caused by opportunistic infections such as toxoplasmosis, cryptococcosis, or FIP.

3.4. Immunodeficiency and Secondary Infections

The most clinically important consequence of both retrovirus infections is immunosuppression. Immunosuppression can lead to secondary infectious diseases accounting for most clinical signs, but also can lead to decreased tumor surveillance mechanisms causing an increased risk of tumor development. It is important to realize that many of these secondary diseases in FeLV- and FIV-infected cats are treatable. The mechanisms that cause the immunosuppression are different for the two infections.
Many FeLV-infected cats have concurrent bacterial, viral, protozoal, and fungal infections, but few controlled studies exist proving that these cats have a higher rate of infection than FeLV-negative cats. Thus, although FeLV certainly can suppress immune function, it should not be assumed that all concurrent infections are a direct consequence of FeLV infection. Progressively FeLV-infected cats develop immunosuppression similar to that in HIV-infected people. The exact mechanisms of how the virus destroys the immune system are poorly understood, as is why different animals have such varying degrees of immunosuppression. Immunosuppression has been associated with non-integrated viral DNA from replication-defective viral variants [97]. These pathogenic immunosuppressive variants, such as FeLV-T, require a membrane-spanning receptor molecule (Pit1) and a second co-receptor protein (FeLIX) to infect T lymphocytes [98]. The latter protein is an endogenously expressed protein encoded by an endogenous provirus arising from FeLV-A, which is similar to the FeLV receptor-binding protein of FeLV-B [99].
FeLV-infected cats may develop thymic atrophy and depletion of lymph node paracortical zones following infection. Lymphopenia and neutropenia are common. In addition, neutrophils of viremic cats have decreased chemotactic and phagocytic function compared with those of normal cats. In some cats, lymphopenia may be characterized by preferential loss of CD4+ helper T cells, resulting in an inverted CD4/CD8 ratio (as typically seen in FIV infection) [100,101], but more commonly, substantial losses of helper cells and cytotoxic suppressor cells (CD8+ cells) occur [101]. Many immune function tests of naturally FeLV-infected cats are abnormal, including decreased response to T-cell mitogens, prolonged allograft reaction, reduced immunoglobulin production, depressed neutrophil function, and complement depletion. IL-2 and IL-4 are decreased in some cats [7,102], but FeLV does not appear to suppress IL-1 production from infected macrophages. IFN-γ may be deficient or increased. Increased TNF-α has been observed in serum of infected cats and in infected cells in culture. Each cytokine plays a vital role in the generation of a normal immune response, and the excess production of certain cytokines, such as TNF-α, can also cause illness. T-cells of FeLV-infected produce significantly lower levels of B-cell stimulatory factors than do those of normal cats (this defect becomes progressively more severe over time) [72], but when B-cells of FeLV-infected cats are stimulated in vitro by uninfected T-cells, their function remains normal. Primary and secondary humoral antibody responses to specific antigens are decreased and may occur delayed in FeLV-infected cats. In vaccination studies, FeLV-infected cats were not able to mount an adequate immune response to vaccines, such as rabies. Therefore, protection in a FeLV-infected cat after vaccination is not complete and not comparable to that in a healthy cat;thus, more frequent vaccinations (e.g., every six months) have to be considered.
In FIV-infected cats, immunosuppression usually occurs in later stages of the infection, and leads to predisposition for secondary infections. In a survey study of 826 naturally FIV-infected cats examined at North American Veterinary Teaching Hospitals, the most common disease syndromes were stomatitis, neoplasia (especially lymphoma and cutaneous squamous cell carcinoma), ocular disease (uveitis and chorioretinitis), anemia and leukopenia, opportunistic infections, renal insufficiency, lower urinary tract disease, and endocrinopathies, such as hyperthyroidism and diabetes mellitus [78]. Some of these problems, however, are most likely associated rather with the older age at which these cats presented (e.g., endocrinopathies, renal insufficiency) than with their FIV infection. Infections with many different “opportunistic” pathogens of viral, bacterial, protozoal, and fungal origin have been reported in FIV-infected cats. Few studies, however, have compared the prevalence of most of these infections in FIV-infected and non-infected cats, and thus, their relevancy as true secondary invaders is unclear.
The most important immunologic abnormality shown in experimental [104,105,106] as well as in natural [107,108] infection is a decrease in the number and relative proportion of CD4+ cells in the peripheral blood as well as in most primary lymphoid tissues [109]. Loss of CD4+ cells leads to inversion of the CD4/CD8 ratio. In addition, an increase in the proportion of CD8+ cells also contributes to the inversion [104,108,110], in particular a population referred to as “CD8+ alpha-hi, beta-low cells” [111,112,113], a subset of CD8+ cells that may contribute to suppression of viremia in FIV-infected cats. Causes of CD4+ cell loss include decreased production secondary to bone marrow or thymic infection, lysis of infected cells induced by FIV itself (cytopathic effects), destruction of virus-infected cells by the immune system, or death by apoptosis (cell death that follows receipt of a membrane signal initiating a series of programmed intracellular events) [114,115,116,117,118,119,120,121,122,123,124,125,126]. The degree of apoptosis correlates inversely with the CD4+ numbers and the CD4/CD8 ratio [127]. FIV env proteins are capable of inducing apoptosis in mononuclear cells by a mechanism that requires CXCR4 binding [128]. Ultimately, loss of CD4+ cells impairs immune responses, because CD4+ cells have critical roles in promoting and maintaining both humoral and cell-mediated immunity. A certain subset of CD4+ cells, the “Treg” (for T-regulatory cells), also seems to play an important role, and Treg cells with suppressive activity have been documented during early [129] and chronic FIV infection [130]. In FIV-infected cats, increased activity of Treg cells could thus play a role in suppressing immune responses to foreign antigens or pathogens. In addition, Treg cells are themselves targets for FIV infection [129,131], and may serve as a FIV reservoir during the latent stage of infection and be capable of stimulating virus production [132]. In addition, other immunologic abnormalities can be found. Lymphocytes may lose the ability to proliferate in response to stimulation with mitogens or antigens, and priming of lymphocytes by immunogens may be impaired [105,133,134,135,136,137,138,139]. Lymphocyte function may be reduced by altered expression of cell surface molecules, such as CD4, major histocompatibility complex II antigens, or cytokines and cytokine receptors [140,141,142,143,144], or through over-expression of abnormal molecules, such as receptors [145], leading to disrupted production of cytokines or receptor function. Impaired neutrophil adhesion and emigration in response to bacterial products have been described in FIV-infected cats [146,147,148]. Natural killer cell activity may be diminished [149] or increased [150], in acutely or asymptomatically infected cats, respectively. Changes in cytokine pattern include increased production of IFN-γ, TNF-α, IL-4, IL-6, IL-10, and IL-12 [151,152,153,154], but also differences in cytokine ratios (e.g., IL-10/IL-12 ratio) [155,156].

3.5. Immune-mediated Diseases

In addition to a dysregulation of the immune system leading to immunosuppression, retrovirus-infected cats can also develop immune-mediated diseases caused by an overactive immune response. The most commonly seen immune-mediated response is a hypergammaglobulinemia which is caused by an excessive antibody response against the chronic persistent infection. The produced antibodies are not neutralizing and thus, may lead to antigen antibody complex formation. These immune complexes can deposit, usually in narrow capillary beds, leading to glomerulonephritis, polyarthritis, uveitis, and vasculitis. Secondary immune-mediated diseases are more commonly seen in FIV- than in FeLV-infected cats. When comparing plasma electrophoretograms, FeLV-infected cats do not show hypergammaglobulinemia and hyperproteinemia significantly more often than non-infected cats, whereas in FIV-infection, hypergammaglobulinemia and hyperproteinemia occur significantly more commonly [4,157].
Nevertheless, immune-mediated diseases have been described in FeLV-infected cats as well. While humoral immunity to specific stimulation decreases during the course of FeLV infection, nonspecific increases of IgG and IgM have been noted. The loss of T-cell activity in combination with the formation of antigen antibody complexes promotes immune dysregulation [158]. Immune-mediated diseases described in FeLV-infected cats include IMHA [159], glomerulonephritis [160], uveitis with immune complex deposition in iris and ciliary body [161], as well as polyarthritis [58]. Chronic progressive polyarthritis can be triggered by FeLV; in about 20% of cats with polyarthritis, FeLV seems to be an associated agent [58]. Measurement of FeLV antigen has shown that cats with glomerulonephritis have more circulating viral proteins than do other FeLV-infected cats. Antigens that can lead to antigen antibody complex formation include not only whole virus particles, but also free gp70, p27, or p15E proteins [162,163].
Immune-mediated diseases observed in FIV-infected cats are caused by an excessive immune response leading to hypergammaglobulinemia [4,104,164]. Hypergammaglobulinemia reflects polyclonal B-cell stimulation and is a direct consequence of FIV infection, because experimentally FIV-infected specific pathogen-free (SPF) healthy cats also develop hypergammaglobulinemia [164]. Increased IgG as well as circulating immune complexes have been detected in FIV-infected cats [165].

3.6. Stomatitis

Chronic ulcero-proliferative gingivostomatitis is very common in retrovirus-infected cats, especially in those with FIV infection. In cats naturally infected with FIV, it is the most common syndrome (affecting up to 50%). It characteristically originates in the fauces and spreads rostrally, especially along the maxillary teeth. Histologically, the mucosa is invaded by plasma cells and lymphocytes, accompanied by variable degrees of neutrophilic and eosinophilic inflammation. Lesions are often painful, and tooth loss is common. Severe stomatitis can lead to anorexia and emaciation. The cause of this syndrome is unclear, but the histologic findings suggest an immune response to chronic antigenic stimulation or immune dysregulation. Circulating lymphocytes of cats with stomatitis have increased expression of inflammatory cytokines [103], further implicating immune activation in the pathogenesis of this condition. This type of stomatitis is not always correlated with FeLV or FIV infection [166], and is usually not seen in SPF cats experimentally infected with FeLV or FIV, suggesting that exposure to other infectious agents also plays a role [167]. Concurrent feline calicivirus (FCV) infection is often identified in the oral cavity of these cats, and experimental and naturally occurring co-infection of FIV and FCV infection results in more severe disease [168,169].

4. Conclusions

FeLV can cause severe clinical syndromes, and progressive FeLV infection is associated with a decrease in life expectancy. Still, many owners still elect to provide therapy for their FeLV-infected cats, and with proper treatment, FeLV-infected cats, especially in indoor-only households, may live for many years with good quality of life. Diseases secondary to immunosuppression account for a large portion of the syndromes seen in FeLV-infected cats, and it is important to realize that many of these secondary diseases are treatable. In most naturally infected cats, FIV does not cause a severe clinical syndrome. Most clinical signs in FIV-infected cats reflect secondary diseases, such as infections and neoplasia, to which FIV-infected cats are more susceptible. With proper care, FIV-infected cats can live many years and, in fact, commonly die at an old age from causes unrelated to their FIV infection. While long-term studies describing clinical outcomes of naturally occurring FeLV and FIV infection are lacking, modalities for treatment of secondary infections or other co-incident diseases are available, and by treating these symptomatically, the life expectancy and quality of life of FeLV- and FIV-infected animals can be significantly improved.
Table 2. Comparison of clinical signs and their main pathomechanism in feline leukemia virus-(FeLV-) infected and feline immunodeficiency virus-(FIV-) infected cats
Table 2. Comparison of clinical signs and their main pathomechanism in feline leukemia virus-(FeLV-) infected and feline immunodeficiency virus-(FIV-) infected cats
Clinical syndrome FeLV FIV
Tumors 62-times as likely as in non-infected cats, direct role of FeLV, mainly T-cell lymphoma 5-times as likely as in non-infected cats, indirect role of FIV, mainly B-cell lymphoma
Bone marrow suppression common, anemia, thrombocytopenia, neutropenia, pancytopenia, primary infection of bone marrow precursor cells and stroma cells rare, mainly neutropenia, soluble factors inhibiting bone marrow function
Neurologic disorders rare, direct influence of the virus, lymphoma and neurotoxic effects (of FeLV envelope glycoprotein) rare, direct influence of the virus (specific FIV strains), impairment of astrocyte function
Immunodeficiency common, several mechanisms, e.g., replication of virus in all bone marrow cells (including neutrophils), changes in cytokine pattern common, several mechanisms, e.g., decrease in CD4+ cells, changes in cytokine pattern
Immune-mediated diseases rare, e.g., immune-mediated hemolytic anemia sometimes, hyperglobulinemia common with immune complex deposition leading to e.g., glomerulonephritis and uveitis
Stomatitis common, multi-factorial disease very common, multi-factorial disease

Acknowledgments

Many doctoral students have contributed to various parts of the author’s publications cited in this paper and are acknowledged for all their hard work.

Conflict of Interest

The authors declare no conflict of interest.

References and Notes

  1. O'Connor, T.P., Jr.; Tonelli, Q.J.; Scarlett, J.M. Report of the national felv/fiv awareness project. J. Am. Vet. Med. Assoc. 1991, 199, 1348–1353. [Google Scholar]
  2. Levy, J.K.; Scott, H.M.; Lachtara, J.L.; Crawford, P.C. Seroprevalence of feline leukemia virus and feline immunodeficiency virus infection among cats in north america and risk factors for seropositivity. J Am Vet Med Assoc 2006, 228, 371–376. [Google Scholar] [CrossRef]
  3. Hoover, E.A.; Mullins, J.I. Feline leukemia-virus infection and diseases. Journal of the American Veterinary Medical Association 1991, 199, 1287–1297. [Google Scholar]
  4. Gleich, S.; Hartmann, K. Hematology and serum biochemistry of feline immunodeficiency virus-infected and feline leukemia virus-infected cats. J Vet Intern Med 2009, 23, 552–558. [Google Scholar] [CrossRef]
  5. Addie, D.D.; Dennis, J.M.; Toth, S.; Callanan, J.J.; Reid, S.; Jarrett, O. Long-term impact on a closed household of pet cats of natural infection with feline coronavirus, feline leukaemia virus and feline immunodeficiency virus. Vet Rec 2000, 146, 419–424. [Google Scholar] [CrossRef]
  6. Cotter, S.M. Feline viral neoplasia. In Infectious diseases of the dog and catGreene, C.E., Ed.; 2nd; WB Saunders: Philadelphia, 1998. [Google Scholar]
  7. Levy, J.K. Felv and non-neoplastic felv-related disease. In Textbook of veterinary internal medicine; Ettinger, S.J., Feldman, E.C., Eds.; WB Saunders: Philadelphia, 2000. [Google Scholar]
  8. Barr, A.C. Fiv and fiv-related diseases. In Textbook of veterinary internal medicine, 5th; Ettinger, S.J., Feldman, E.C., Eds.; WB Saunders: Philadelphia, 2000; pp. 433–438. [Google Scholar]
  9. Torres, A.N.; O'Halloran, K.P.; Larson, L.J.; Schultz, R.D.; Hoover, E.A. Development and application of a quantitative real-time pcr assay to detect feline leukemia virus rna. Vet Immunol Immunopathol 2008, 123, 81–89. [Google Scholar] [CrossRef]
  10. Cattori, V.; Tandon, R.; Pepin, A.; Lutz, H.; Hofmann-Lehmann, R. Rapid detection of feline leukemia virus provirus integration into feline genomic DNA. Mol Cell Probes 2006, 20, 172–181. [Google Scholar] [CrossRef]
  11. Torres, A.N.; Mathiason, C.K.; Hoover, E.A. Re-examination of feline leukemia virus: Host relationships using real-time pcr. Virology 2005, 332, 272–283. [Google Scholar] [CrossRef]
  12. Hofmann-Lehmann, R.; Cattori, V.; Tandon, R.; Boretti, F.S.; Meli, M.L.; Riond, B.; Pepin, A.C.; Willi, B.; Ossent, P.; Lutz, H. Vaccination against the feline leukaemia virus: Outcome and response categories and long-term follow-up. Vaccine 2007, 25, 5531–5539. [Google Scholar] [CrossRef]
  13. Hofmann-Lehmann, R.; Cattori, V.; Tandon, R.; Boretti, F.S.; Meli, M.L.; Riond, B.; Lutz, H. How molecular methods change our views of felv infection and vaccination. Vet Immunol Immunopathol 2008, 123, 119–123. [Google Scholar] [CrossRef]
  14. Levy, J.; Crawford, C.; Hartmann, K.; Hofmann-Lehmann, R.; Little, S.; Sundahl, E.; Thayer, V. 2008 american association of feline practitioners' feline retrovirus management guidelines. J Feline Med Surg 2008, 10, 300–316. [Google Scholar] [CrossRef]
  15. Major, A.; Cattori, V.; Boenzli, E.; Riond, B.; Ossent, P.; Meli, M.L.; Hofmann-Lehmann, R.; Lutz, H. Exposure of cats to low doses of felv: Seroconversion as the sole parameter of infection. Vet Res 2010, 41, 17. [Google Scholar] [CrossRef]
  16. Hofmann-Lehmann, R.; Huder, J.B.; Gruber, S.; Boretti, F.; Sigrist, B.; Lutz, H. Feline leukaemia provirus load during the course of experimental infection and in naturally infected cats. J Gen Virol 2001, 82, 1589–1596. [Google Scholar]
  17. Pepin, A.C.; Tandon, R.; Cattori, V.; Niederer, E.; Riond, B.; Willi, B.; Lutz, H.; Hofmann-Lehmann, R. Cellular segregation of feline leukemia provirus and viral rna in leukocyte subsets of long-term experimentally infected cats. Virus Res 2007, 127, 9–16. [Google Scholar] [CrossRef]
  18. English, R.V. Feline immunodeficiency virus. In Kirk's current veterinary therapy; Bonagura, J.D., Ed.; WB Saunders: Philadelphia, 1995. [Google Scholar]
  19. Goto, Y.; Nishimura, Y.; Mizuno, T.; Endo, Y.; Baba, K.; Momoi, Y.; Watari, T.; Hasegawa, A.; Tsujimoto, H. Quantification of viral ribonucleic acid in plasma of cats naturally infected with feline immunodeficiency virus. Am J Vet Res 2000, 61, 1609–1613. [Google Scholar] [CrossRef]
  20. Cotter, S.M. Management of healthy feline leukemia virus-positive cats. J Am Vet Med Assoc 1991, 199, 1470–1473. [Google Scholar]
  21. Flynn, J.N.; Dunham, S.P.; Watson, V.; Jarrett, O. Longitudinal analysis of feline leukemia virus-specific cytotoxic t lymphocytes: Correlation with recovery from infection. J Virol 2002, 76, 2306–2315. [Google Scholar] [CrossRef]
  22. Hoover, E.A.; Olsen, R.G.; Hardy, W.D., Jr.; Schaller, J.P.; Mathes, L.E. Feline leukemia virus infection: Age-related variation in response of cats to experimental infection. J Natl Cancer Inst 1976, 57, 365–369. [Google Scholar]
  23. Obert, L.A.; Hoover, E.A. Relationship of lymphoid lesions to disease course in mucosal feline immunodeficiency virus type c infection. Vet Pathol 2000, 37, 386–401. [Google Scholar] [CrossRef]
  24. Podell, M.; Hayes, K.; Oglesbee, M.; Mathes, L. Progressive encephalopathy associated with cd4/cd8 inversion in adult fiv-infected cats. J Acquir Immune Defic Syndr Hum Retrovirol 1997, 15, 332–340. [Google Scholar] [CrossRef]
  25. Pedersen, N.C.; Leutenegger, C.M.; Woo, J.; Higgins, J. Virulence differences between two field isolates of feline immunodeficiency virus (fiv-apetaluma and fiv-cpgammar) in young adult specific pathogen free cats. Vet Immunol Immunopathol 2001, 79, 53–67. [Google Scholar] [CrossRef]
  26. Poli, A.; Abramo, F.; Baldinotti, F.; Pistello, M.; Da Prato, L.; Bendinelli, M. Malignant lymphoma associated with experimentally induced feline immunodeficiency virus infection. J Comp Pathol 1994, 110, 319–328. [Google Scholar] [CrossRef]
  27. Callanan, J.J.; Jones, B.A.; Irvine, J.; Willett, B.J.; McCandlish, I.A.; Jarrett, O. Histologic classification and immunophenotype of lymphosarcomas in cats with naturally and experimentally acquired feline immunodeficiency virus infections. Vet Pathol 1996, 33, 264–272. [Google Scholar]
  28. Rickard, C.G.; Post, J.E.; Noronha, F.; Barr, L.M. A transmissible virus-induced lymphocytic leukemia of the cat. J Natl Cancer Inst 1969, 42, 987–1014. [Google Scholar]
  29. Hardy, W.D., Jr.; Hirshaut, Y.; Hess, P. Detection of the feline leukemia virus and other mammalian oncornaviruses by immunofluorescence. Bibl Haematol 1973, 39, 778–799. [Google Scholar]
  30. Jarrett, O.; Laird, H.M.; Hay, D. Determinants of the host range of feline leukaemia viruses. J Gen Virol 1973, 20, 169–175. [Google Scholar]
  31. Essex, M.; Cotter, S.M.; Hardy, W.D., Jr.; Hess, P.; Jarrett, W.; Jarrett, O.; Mackey, L.; Laird, H.; Perryman, L.; Olsen, R.G.; et al. Feline oncornavirus-associated cell membrane antigen. Iv. Antibody titers in cats with naturally occurring leukemia, lymphoma, and other diseases. J Natl Cancer Inst 1975, 55, 463–467. [Google Scholar]
  32. Cotter, S.M.; Hardy, W.D., Jr.; Essex, M. Association of feline leukemia virus with lymphosarcoma and other disorders in the cat. J Am Vet Med Assoc 1975, 166, 449–454. [Google Scholar]
  33. Francis, D.P.; Essex, M.; Hardy, W.D., Jr. Excretion of feline leukaemia virus by naturally infected pet cats. Nature 1977, 269, 252–254. [Google Scholar] [CrossRef]
  34. Francis, D.P.; Cotter, S.M.; Hardy, W.D., Jr.; Essex, M. Comparison of virus-positive and virus-negative cases of feline leukemia and lymphoma. Cancer Res 1979, 39, 3866–3870. [Google Scholar]
  35. Hardy, W.D., Jr.; McClelland, A.J.; Zuckerman, E.E.; Snyder, H.W., Jr.; MacEwen, E.G.; Francis, D.; Essex, M. Development of virus non-producer lymphosarcomas in pet cats exposed to felv. Nature 1980, 288, 90–92. [Google Scholar] [CrossRef]
  36. Reinacher, M. Infektionen mit dem felinen leukämie-virus (felv). Kleintierprax 1987, 32, 65–72. [Google Scholar]
  37. Shelton, G.H.; Grant, C.K.; Cotter, S.M.; Gardner, M.B.; Hardy, W.D., Jr.; DiGiacomo, R.F. Feline immunodeficiency virus and feline leukemia virus infections and their relationships to lymphoid malignancies in cats: A retrospective study (1968-1988). J Acquir Immune Defic Syndr 1990, 3, 623–630. [Google Scholar]
  38. Harrus, S.; Klement, E.; Aroch, I.; Stein, T.; Bark, H.; Lavy, E.; Mazaki-Tovi, M.; Baneth, G. Retrospective study of 46 cases of feline haemobartonellosis in israel and their relationships with felv and fiv infections. Vet Rec 2002, 151, 82–85. [Google Scholar]
  39. Mauldin, G.E.; Mooney, S.C.; Meleo, R.E. Chemotherapy in 132 cats with lymphoma: 1988-1994, Proceedings of the Veterinary Cancer Society; 1995; pp. 35–36.
  40. Moore, A.S.; Cotter, S.M.; Frimberger, A.E.; Wood, C.A.; Rand, W.M.; L'Heureux, D.A. A comparison of doxorubicin and cop for maintenance of remission in cats with lymphoma. J Vet Intern Med 1996, 10, 372–375. [Google Scholar] [CrossRef]
  41. Hartmann, K.; Gerle, K.; Leutenegger, C. Feline leukemia virus - most important oncogene in cats? In Abstracts from the 4th International Feline Retrovirus Research Symposium, Glasgow, Scotland, UK, 1998.
  42. Teske, E.; van Straten, G.; van Noort, R.; Rutteman, G.R. Chemotherapy with cyclophosphamide, vincristine, and prednisolone (cop) in cats with malignant lymphoma: New results with an old protocol. J Vet Intern Med 2002, 16, 179–186. [Google Scholar] [CrossRef]
  43. Jackson, M.L.; Haines, D.M.; Meric, S.M.; Misra, V. Feline leukemia virus detection by immunohistochemistry and polymerase chain reaction in formalin-fixed, paraffin-embedded tumor tissue from cats with lymphosarcoma. Can J Vet Res 1993, 57, 269–276. [Google Scholar]
  44. Rohn, J.L.; Linenberger, M.L.; Hoover, E.A.; Overbaugh, J. Evolution of feline leukemia virus variant genomes with insertions, deletions, and defective envelope genes in infected cats with tumors. J Virol 1994, 68, 2458–2467. [Google Scholar]
  45. Sheets, R.L.; Pandey, R.; Jen, W.C.; Roy-Burman, P. Recombinant feline leukemia virus genes detected in naturally occurring feline lymphosarcomas. J Virol 1993, 67, 3118–3125. [Google Scholar]
  46. Rezanka, L.J.; Rojko, J.L.; Neil, J.C. Feline leukemia virus: Pathogenesis of neoplastic disease. Cancer investigation 1992, 10, 371–389. [Google Scholar] [CrossRef]
  47. Stützer, B.; Simon, K.; Lutz, H.; Majzoub, M.; Hermanns, W.; Hirschberger, J.; Sauter-Louis, C.; Hartmann, K. Incidence of persistent viraemia and latent feline leukaemia virus infection in cats with lymphoma. J Feline Med Surg 2011, 13, 81–87. [Google Scholar] [CrossRef]
  48. Tsatsanis, C.; Fulton, R.; Nishigaki, K.; Tsujimoto, H.; Levy, L.; Terry, A.; Spandidos, D.; Onions, D.; Neil, J.C. Genetic determinants of feline leukemia virus-induced lymphoid tumors: Patterns of proviral insertion and gene rearrangement. J Virol 1994, 68, 8296–8303. [Google Scholar]
  49. Forman, L.W.; Pal-Ghosh, R.; Spanjaard, R.A.; Faller, D.V.; Ghosh, S.K. Identification of ltr-specific small non-coding rna in felv infected cells. FEBS Lett 2009, 583, 1386–1390. [Google Scholar] [CrossRef]
  50. Fujino, Y.; Ohno, K.; Tsujimoto, H. Molecular pathogenesis of feline leukemia virus-induced malignancies: Insertional mutagenesis. Vet Immunol Immunopathol 2008, 123, 138–143. [Google Scholar] [CrossRef]
  51. Fujino, Y.; Liao, C.P.; Zhao, Y.S.; Pan, J.; Mathes, L.E.; Hayes, K.A.; Ohno, K.; Tsujimoto, H.; Roy-Burman, P. Identification of a novel common proviral integration site, flit-1, in feline leukemia virus induced thymic lymphoma. Virology 2009, 386, 16–22. [Google Scholar] [CrossRef]
  52. Ellis, J.A.; Jackson, M.L.; Bartsch, R.C.; McGill, L.G.; Martin, K.M.; Trask, B.R.; Haines, D.M. Use of immunohistochemistry and polymerase chain reaction for detection of oncornaviruses in formalin-fixed, paraffin-embedded fibrosarcomas from cats. J Am Vet Med Assoc 1996, 209, 767–771. [Google Scholar]
  53. Stiles, J.; Bienzle, D.; Render, J.A.; Buyukmihci, N.C.; Johnson, E.C. Use of nested polymerase chain reaction (pcr) for detection of retroviruses from formalin-fixed, paraffin-embedded uveal melanomas in cats. Vet Ophthalmol 1999, 2, 113–116. [Google Scholar]
  54. Cullen, C.L.; Haines, D.M.; Jackson, M.L.; Grahn, B.H. Lack of detection of feline leukemia and feline sarcoma viruses in diffuse iris melanomas of cats by immunohistochemistry and polymerase chain reaction. J Vet Diagn Invest 2002, 14, 340–343. [Google Scholar]
  55. Pool, R.R.; Carrig, C.B. Multiple cartilagenous exostoses in a cat. Vet Pathol 1972, 9, 350–359. [Google Scholar]
  56. Lott-Stolz, G. [short original report. Osteochondromatosis in the cat]. Schweiz Arch Tierheilkd 1988, 130, 635–638. [Google Scholar]
  57. Schrenzel, M.D.; Higgins, R.J.; Hinrichs, S.H.; Smith, M.O.; Torten, M. Type c retroviral expression in spontaneous feline olfactory neuroblastomas. Acta Neuropathol 1990, 80, 547–553. [Google Scholar]
  58. Pedersen, N.C. Feline leukemia virus infection. Feline husbandry, disease and management in the multiple cat environment 1991, 210–220. [Google Scholar]
  59. Terry, A.; Callanan, J.J.; Fulton, R.; Jarrett, O.; Neil, J.C. Molecular analysis of tumours from feline immunodeficiency virus (fiv)-infected cats: An indirect role for fiv? Int J Cancer 1995, 61, 227–232. [Google Scholar] [CrossRef]
  60. Gabor, L.J.; Love, D.N.; Malik, R.; Canfield, P.J. Feline immunodeficiency virus status of australian cats with lymphosarcoma. Aust Vet J 2001, 79, 540–545. [Google Scholar] [CrossRef]
  61. Fleming, E.J.; McCaw, D.L.; Smith, J.A.; Buening, G.M.; Johnson, C. Clinical, hematologic, and survival data from cats infected with feline immunodeficiency virus: 42 cases (1983-1988). J Am Vet Med Assoc 1991, 199, 913–916. [Google Scholar]
  62. Hutson, C.A.; Rideout, B.A.; Pedersen, N.C. Neoplasia associated with feline immunodeficiency virus infection in cats of southern california. J Am Vet Med Assoc 1991, 199, 1357–1362. [Google Scholar]
  63. Buracco, P.; Guglielmino, R.; Abate, O.; Bocchini, V.; Cornaglia, E.; Denicola, D.B.; Cilli, M.; Ponzio, P. Large granular lymphoma in an fiv-positive and felv-negative cat. Journal of Small Animal Practice 1992, 33, 279–284. [Google Scholar] [CrossRef]
  64. Callanan, J.J.; McCandlish, I.A.; O'Neil, B.; Lawrence, C.E.; Rigby, M.; Pacitti, A.M.; Jarrett, O. Lymphosarcoma in experimentally induced feline immunodeficiency virus infection [corrected]. Vet Rec 1992, 130, 293–295. [Google Scholar]
  65. Court, E.A.; Watson, A.D.; Peaston, A.E. Retrospective study of 60 cases of feline lymphosarcoma. Aust Vet J 1997, 75, 424–427. [Google Scholar]
  66. Barr, M.C.; Huitron-Resendiz, S.; Selway, D.R.; Henriksen, S.J.; Phillips, T.R. Exogenous glucocorticoids alter parameters of early feline immunodeficiency virus infection. J Infect Dis 2000, 181, 576–586. [Google Scholar] [CrossRef]
  67. Beatty, J.A.; Callanan, J.J.; Terry, A.; Jarrett, O.; Neil, J.C. Molecular and immunophenotypical characterization of a feline immunodeficiency virus (fiv)-associated lymphoma: A direct role for fiv in b-lymphocyte transformation? J Virol 1998, 72, 767–771. [Google Scholar]
  68. Beatty, J.A.; Lawrence, C.E.; Callanan, J.J.; Grant, C.K.; Gault, E.A.; Neil, J.C.; Jarrett, O. Feline immunodeficiency virus (fiv)-associated lymphoma: A potential role for immune dysfunction in tumourigenesis. Vet Immunol Immunopathol 1998, 65, 309–322. [Google Scholar]
  69. Wang, J.; Kyaw-Tanner, M.; Lee, C. Characterisation of lymphosarcomas in australian cats using polymerase chain reaction and immunohistochemical examination. Aust Vet J 2001, 79, 41–46. [Google Scholar] [CrossRef]
  70. Beatty, J.; Terry, A.; MacDonald, J.; Gault, E.; Cevario, S.; O'Brien, S.J.; Cameron, E.; Neil, J.C. Feline immunodeficiency virus integration in b-cell lymphoma identifies a candidate tumor suppressor gene on human chromosome 15q15. Cancer Res 2002, 62, 7175–7180. [Google Scholar]
  71. Endo, Y.; Cho, K.W.; Nishigaki, K.; Momoi, Y.; Nishimura, Y.; Mizuno, T.; Goto, Y.; Watari, T.; Tsujimoto, H.; Hasegawa, A. Molecular characteristics of malignant lymphomas in cats naturally infected with feline immunodeficiency virus. Vet Immunol Immunopathol 1997, 57, 153–167. [Google Scholar] [CrossRef]
  72. Diehl, L.J.; Hoover, E.A. Early and progressive helper t-cell dysfunction in feline leukemia virus-induced immunodeficiency. J Acquir Immune Defic Syndr 1992, 5, 1188–1194. [Google Scholar]
  73. Stützer, B.; Muller, F.; Majzoub, M.; Lutz, H.; Greene, C.E.; Hermanns, W.; Hartmann, K. Role of latent feline leukemia virus infection in nonregenerative cytopenias of cats. J Vet Intern Med 2010, 24, 192–197. [Google Scholar] [CrossRef]
  74. Shelton, G.H.; Linenberger, M.L. Hematologic abnormalities associated with retroviral infections in the cat. Semin Vet Med Surg (Small Anim) 1995, 10, 220–233. [Google Scholar]
  75. Brown, M.R.; Rogers, K.S. Neutropenia in dogs and cats: A retrospective study of 261 cases. J Am Anim Hosp Assoc 2001, 37, 131–139. [Google Scholar]
  76. Hisasue, M.; Nagashima, N.; Nishigaki, K.; Fukuzawa, I.; Ura, S.; Katae, H.; Tsuchiya, R.; Yamada, T.; Hasegawa, A.; Tsujimoto, H. Myelodysplastic syndromes and acute myeloid leukemia in cats infected with feline leukemia virus clone33 containing a unique long terminal repeat. Int J Cancer 2009, 124, 1133–1141. [Google Scholar] [CrossRef]
  77. Lutz, H.; Castelli, I.; Ehrensperger, F.; Pospischil, A.; Rosskopf, M.; Siegl, G.; Grob, M.; Martinod, S. Panleukopenia-like syndrome of felv caused by co-infection with felv and feline panleukopenia virus. Vet Immunol Immunopathol 1995, 46, 21–33. [Google Scholar] [CrossRef]
  78. Kipar, A.; Kremendahl, J.; Grant, C.K.; von Bothmer, I.; Reinacher, M. Expression of viral proteins in feline leukemia virus-associated enteritis. Vet Pathol 2000, 37, 129–136. [Google Scholar]
  79. Kipar, A.; Kremendahl, J.; Jackson, M.L.; Reinacher, M. Comparative examination of cats with feline leukemia virus-associated enteritis and other relevant forms of feline enteritis. Vet Pathol 2001, 38, 359–371. [Google Scholar] [CrossRef]
  80. Carmichael, K.P.; Bienzle, D.; McDonnell, J.J. Feline leukemia virus-associated myelopathy in cats. Vet Pathol 2002, 39, 536–545. [Google Scholar]
  81. Fails, A.D.; Mitchell, T.W.; Rojko, J.L.; Whalen, L.R. An oligopeptide of the feline leukemia virus envelope glycoprotein is associated with morphological changes and calcium dysregulation in neuronal growth cones. J Neurovirol 1997, 3, 179–191. [Google Scholar] [CrossRef]
  82. Mitchell, T.W.; Rojko, J.L.; Hartke, J.R.; Mihajlov, A.R.; Kasameyer, G.A.; Gasper, P.W.; Whalen, L.R. Felv envelope protein (gp70) variable region 5 causes alterations in calcium homeostasis and toxicity of neurons. J Acquir Immune Defic Syndr Hum Retrovirol 1997, 14, 307–320. [Google Scholar]
  83. Dow, S.W.; Poss, M.L.; Hoover, E.A. Feline immunodeficiency virus: A neurotropic lentivirus. J Acquir Immune Defic Syndr 1990, 3, 658–668. [Google Scholar]
  84. Dow, S.W.; Dreitz, M.J.; Hoover, E.A. Feline immunodeficiency virus neurotropism: Evidence that astrocytes and microglia are the primary target cells. Vet Immunol Immunopathol 1992, 35, 23–35. [Google Scholar] [CrossRef]
  85. Podell, M.; Oglesbee, M.; Mathes, L.; Krakowka, S.; Olmstead, R.; Lafrado, L. Aids-associated encephalopathy with experimental feline immunodeficiency virus infection. J Acquir Immune Defic Syndr 1993, 6, 758–771. [Google Scholar]
  86. English, R.V.; Nelson, P.; Johnson, C.M.; Nasisse, M.; Tompkins, W.A.; Tompkins, M.B. Development of clinical disease in cats experimentally infected with feline immunodeficiency virus. J Infect Dis 1994, 170, 543–552. [Google Scholar] [CrossRef]
  87. Phillips, T.R.; Prospero-Garcia, O.; Puaoi, D.L.; Lerner, D.L.; Fox, H.S.; Olmsted, R.A.; Bloom, F.E.; Henriksen, S.J.; Elder, J.H. Neurological abnormalities associated with feline immunodeficiency virus infection. J Gen Virol 1994, 75 ( Pt 5), 979–987. [Google Scholar]
  88. Abramo, F.; Bo, S.; Canese, M.G.; Poli, A. Regional distribution of lesions in the central nervous system of cats infected with feline immunodeficiency virus. AIDS Res Hum Retroviruses 1995, 11, 1247–1253. [Google Scholar] [CrossRef]
  89. Power, C.; Buist, R.; Johnston, J.B.; Del Bigio, M.R.; Ni, W.; Dawood, M.R.; Peeling, J. Neurovirulence in feline immunodeficiency virus-infected neonatal cats is viral strain specific and dependent on systemic immune suppression. J Virol 1998, 72, 9109–9115. [Google Scholar]
  90. Prospero-Garcia, O.; Herold, N.; Phillips, T.R.; Elder, J.H.; Bloom, F.E.; Henriksen, S.J. Sleep patterns are disturbed in cats infected with feline immunodeficiency virus. Proc Natl Acad Sci U S A 1994, 91, 12947–12951. [Google Scholar]
  91. Gunn-Moore, D.A.; Pearson, G.R.; Harbour, D.A.; Whiting, C.V. Encephalitis associated with giant cells in a cat with naturally occurring feline immunodeficiency virus infection demonstrated by in situ hybridization. Vet Pathol 1996, 33, 699–703. [Google Scholar]
  92. Steigerwald, E.S.; Sarter, M.; March, P.; Podell, M. Effects of feline immunodeficiency virus on cognition and behavioral function in cats. J Acquir Immune Defic Syndr Hum Retrovirol 1999, 20, 411–419. [Google Scholar] [CrossRef]
  93. Podell, M.; Maruyama, K.; Smith, M.; Hayes, K.A.; Buck, W.R.; Ruehlmann, D.S.; Mathes, L.E. Frontal lobe neuronal injury correlates to altered function in fiv-infected cats. J Acquir Immune Defic Syndr 1999, 22, 10–18. [Google Scholar]
  94. Phipps, A.J.; Hayes, K.A.; Buck, W.R.; Podell, M.; Mathes, L.E. Neurophysiologic and immunologic abnormalities associated with feline immunodeficiency virus molecular clone fiv-ppr DNA inoculation. J Acquir Immune Defic Syndr 2000, 23, 8–16. [Google Scholar]
  95. Sellon, R.K. Feline immunodeficiency virus infection. In Infectious diseases of the dog and cat, 2nd; Greene, C.E., Ed.; WB Saunders: Philadelphia, 1998; pp. 84–96. [Google Scholar]
  96. Sellon, R.; Hartmann, K. Feline immunodeficiency virus infection. In Infectious diseases of the dog and cat; Greene, C.E., Ed.; WB Saunders: St. Louis, 2006; pp. 131–142. [Google Scholar]
  97. Overbaugh, J.; Donahue, P.R.; Quackenbush, S.L.; Hoover, E.A.; Mullins, J.I. Molecular cloning of a feline leukemia virus that induces fatal immunodeficiency disease in cats. Science 1988, 239, 906–910. [Google Scholar]
  98. Lauring, A.S.; Cheng, H.H.; Eiden, M.V.; Overbaugh, J. Genetic and biochemical analyses of receptor and cofactor determinants for t-cell-tropic feline leukemia virus infection. J Virol 2002, 76, 8069–8078. [Google Scholar]
  99. Barnett, A.L.; Wensel, D.L.; Li, W.; Fass, D.; Cunningham, J.M. Structure and mechanism of a coreceptor for infection by a pathogenic feline retrovirus. J Virol 2003, 77, 2717–2729. [Google Scholar]
  100. Quackenbush, S.L.; Donahue, P.R.; Dean, G.A.; Myles, M.H.; Ackley, C.D.; Cooper, M.D.; Mullins, J.I.; Hoover, E.A. Lymphocyte subset alterations and viral determinants of immunodeficiency disease induction by the feline leukemia virus felv-faids. J Virol 1990, 64, 5465–5474. [Google Scholar]
  101. Hoffmann-Fezer, G.; Mortelbauer, W.; Hartmann, K.; Mysliwietz, J.; Thefeld, S.; Beer, B.; Thum, I.; Kraft, W. Comparison of t-cell subpopulations in cats naturally infected with feline leukaemia virus or feline immunodeficiency virus. Res Vet Sci 1996, 61, 222–226. [Google Scholar] [CrossRef]
  102. Linenberger, M.L.; Deng, T. The effects of feline retroviruses on cytokine expression. Vet Immunol Immunopathol 1999, 72, 343–368. [Google Scholar]
  103. Levy, J.K. Cvt update: Feline immunodeficiency virus. In Kirk's current veterinary therapy xiii small animal practice; Bonagura, J.D., Ed.; WB Saunders: Philadelphia, 2000; pp. 284–288. [Google Scholar]
  104. Ackley, C.D.; Yamamoto, J.K.; Levy, N.; Pedersen, N.C.; Cooper, M.D. Immunologic abnormalities in pathogen-free cats experimentally infected with feline immunodeficiency virus. J Virol 1990, 64, 5652–5655. [Google Scholar]
  105. Barlough, J.E.; Ackley, C.D.; George, J.W.; Levy, N.; Acevedo, R.; Moore, P.F.; Rideout, B.A.; Cooper, M.D.; Pedersen, N.C. Acquired immune dysfunction in cats with experimentally induced feline immunodeficiency virus infection: Comparison of short-term and long-term infections. J Acquir Immune Defic Syndr 1991, 4, 219–227. [Google Scholar]
  106. Tompkins, M.B.; Nelson, P.D.; English, R.V.; Novotney, C. Early events in the immunopathogenesis of feline retrovirus infections. J Am Vet Med Assoc 1991, 199, 1311–1315. [Google Scholar]
  107. Novotney, C.; English, R.V.; Housman, J.; Davidson, M.G.; Nasisse, M.P.; Jeng, C.R.; Davis, W.C.; Tompkins, M.B. Lymphocyte population changes in cats naturally infected with feline immunodeficiency virus. AIDS 1990, 4, 1213–1218. [Google Scholar]
  108. Hoffmann-Fezer, G.; Thum, J.; Ackley, C.; Herbold, M.; Mysliwietz, J.; Thefeld, S.; Hartmann, K.; Kraft, W. Decline in cd4+ cell numbers in cats with naturally acquired feline immunodeficiency virus infection. J Virol 1992, 66, 1484–1488. [Google Scholar]
  109. Bull, M.E.; Kennedy-Stoskopf, S.; Levine, J.F.; Loomis, M.; Gebhard, D.G.; Tompkins, W.A. Evaluation of t lymphocytes in captive african lions (panthera leo) infected with feline immunodeficiency virus. Am J Vet Res 2003, 64, 1293–1300. [Google Scholar] [CrossRef]
  110. Willett, B.J.; Hosie, M.J.; Callanan, J.J.; Neil, J.C.; Jarrett, O. Infection with feline immunodeficiency virus is followed by the rapid expansion of a cd8+ lymphocyte subset. Immunology 1993, 78, 1–6. [Google Scholar]
  111. Shimojima, M.; Miyazawa, T.; Kohmoto, M.; Ikeda, Y.; Nishimura, Y.; Maeda, K.; Tohya, Y.; Mikami, T. Expansion of cd8alpha+beta- cells in cats infected with feline immunodeficiency virus. J Gen Virol 1998, 79 ( Pt 1), 91–94. [Google Scholar]
  112. Shimojima, M.; Nishimura, Y.; Miyazawa, T.; Tohya, Y.; Akashi, H. Phenotypic changes in cd8+ peripheral blood lymphocytes in cats infected with feline immunodeficiency virus. Microbes Infect 2003, 5, 1171–1176. [Google Scholar]
  113. Phadke, A.P.; de la Concha-Bermejillo, A.; Wolf, A.M.; Andersen, P.R.; Baladandayuthapani, V.; Collisson, E.W. Pathogenesis of a texas feline immunodeficiency virus isolate: An emerging subtype of clade b. Vet Microbiol 2006, 115, 64–76. [Google Scholar] [CrossRef]
  114. Bishop, S.A.; Gruffydd-Jones, T.J.; Harbour, D.A.; Stokes, C.R. Programmed cell death (apoptosis) as a mechanism of cell death in peripheral blood mononuclear cells from cats infected with feline immunodeficiency virus (fiv). Clin Exp Immunol 1993, 93, 65–71. [Google Scholar]
  115. Ohno, K.; Nakano, T.; Matsumoto, Y.; Watari, T.; Goitsuka, R.; Nakayama, H.; Tsujimoto, H.; Hasegawa, A. Apoptosis induced by tumor necrosis factor in cells chronically infected with feline immunodeficiency virus. J Virol 1993, 67, 2429–2433. [Google Scholar]
  116. Ohno, K.; Okamoto, Y.; Miyazawa, T.; Mikami, T.; Watari, T.; Goitsuka, R.; Tsujimoto, H.; Hasegawa, A. Induction of apoptosis in a t lymphoblastoid cell line infected with feline immunodeficiency virus. Arch Virol 1994, 135, 153–158. [Google Scholar]
  117. Johnson, C.M.; Benson, N.A.; Papadi, G.P. Apoptosis and cd4+ lymphocyte depletion following feline immunodeficiency virus infection of a t-lymphocyte cell line. Vet Pathol 1996, 33, 195–203. [Google Scholar]
  118. Guiot, A.L.; Rigal, D.; Chappuis, G. Spontaneous programmed cell death (pcd) process of lymphocytes of fiv-infected cats: Pharmacological modulation in vitro. Int J Immunopharmacol 1997, 19, 167–179. [Google Scholar] [CrossRef]
  119. Mizuno, T.; Momoi, Y.; Endo, Y.; Nishimura, Y.; Goto, Y.; Ohno, K.; Watari, T.; Tsujimoto, H.; Hasegawa, A. Apoptosis enhanced by soluble factor produced in feline immunodeficiency virus infection. J Vet Med Sci 1997, 59, 1049–1051. [Google Scholar]
  120. Mortola, E.; Endo, Y.; Mizuno, T.; Ohno, K.; Watari, T.; Tsujimoto, H.; Hasegawa, A. Effect of interleukin-12 and interleukin-10 on the virus replication and apoptosis in t-cells infected with feline immunodeficiency virus. J Vet Med Sci 1998, 60, 1181–1185. [Google Scholar]
  121. Mortola, E.; Okuda, M.; Ohno, K.; Watari, T.; Tsujimoto, H.; Hasegawa, A. Inhibition of apoptosis and virus replication in feline immunodeficiency virus-infected cells by n-acetylcysteine and ascorbic acid. J Vet Med Sci 1998, 60, 1187–1193. [Google Scholar]
  122. Piedimonte, G.; Crinelli, R.; Della Salda, L.; Corsi, D.; Pennisi, M.G.; Kramer, L.; Casabianca, A.; Sarli, G.; Bendinelli, M.; Marcato, P.S.; et al. Protein degradation and apoptotic death in lymphocytes during fiv infection: Activation of the ubiquitin-proteasome proteolytic system. Exp Cell Res 1999, 248, 381–390. [Google Scholar] [CrossRef]
  123. Mizuno, T.; Goto, Y.; Baba, K.; Masuda, K.; Ohno, K.; Tsujimoto, H. Tnf-alpha-induced cell death in feline immunodeficiency virus-infected cells is mediated by the caspase cascade. Virology 2001, 287, 446–455. [Google Scholar]
  124. Tompkins, M.B.; Bull, M.E.; Dow, J.L.; Ball, J.M.; Collisson, E.W.; Winslow, B.J.; Phadke, A.P.; Vahlenkamp, T.W.; Tompkins, W.A. Feline immunodeficiency virus infection is characterized by b7+ctla4+ t cell apoptosis. J Infect Dis 2002, 185, 1077–1093. [Google Scholar] [CrossRef]
  125. Mizuno, T.; Goto, Y.; Baba, K.; Momoi, Y.; Endo, Y.; Nishimura, Y.; Masuda, K.; Ohno, K.; Tsujimoto, H. Quantitative analysis of fas and fas ligand mrnas in a feline t-lymphoid cell line after infection with feline immunodeficiency virus and primary peripheral blood mononuclear cells obtained from cats infected with the virus. Vet Immunol Immunopathol 2003, 93, 117–123. [Google Scholar]
  126. Guiot, A.L.; Rigal, D.; Chappuis, G. Spontaneous programmed cell death (pcd) process of lymphocytes of fiv-infected cats: Cellular targets and modulation. Vet Immunol Immunopathol 1997, 58, 93–106. [Google Scholar] [CrossRef]
  127. Holznagel, E.; Hofmann-Lehmann, R.; Leutenegger, C.M.; Allenspach, K.; Huettner, S.; Forster, U.; Niederer, E.; Joller, H.; Willett, B.J.; Hummel, U.; et al. The role of in vitro-induced lymphocyte apoptosis in feline immunodeficiency virus infection: Correlation with different markers of disease progression. J Virol 1998, 72, 9025–9033. [Google Scholar]
  128. Garg, H.; Joshi, A.; Tompkins, W.A. Feline immunodeficiency virus envelope glycoprotein mediates apoptosis in activated pbmc by a mechanism dependent on gp41 function. Virology 2004, 330, 424–436. [Google Scholar]
  129. Mexas, A.M.; Fogle, J.E.; Tompkins, W.A.; Tompkins, M.B. Cd4+cd25+ regulatory t cells are infected and activated during acute fiv infection. Vet Immunol Immunopathol 2008, 126, 263–272. [Google Scholar] [CrossRef]
  130. Petty, C.S.; Tompkins, M.B.; Tompkins, W.A. Transforming growth factor-beta/transforming growth factor-betarii signaling may regulate cd4+cd25+ t-regulatory cell homeostasis and suppressor function in feline aids lentivirus infection. J Acquir Immune Defic Syndr 2008, 47, 148–160. [Google Scholar] [CrossRef]
  131. Joshi, A.; Garg, H.; Tompkins, M.B.; Tompkins, W.A. Different thresholds of t cell activation regulate fiv infection of cd4+cd25+ and cd4+cd25- cells. Virology 2005, 335, 212–221. [Google Scholar]
  132. Joshi, A.; Vahlenkamp, T.W.; Garg, H.; Tompkins, W.A.; Tompkins, M.B. Preferential replication of fiv in activated cd4(+)cd25(+)t cells independent of cellular proliferation. Virology 2004, 321, 307–322. [Google Scholar] [CrossRef]
  133. Hara, Y.; Ishida, T.; Ejima, H.; Tagawa, M.; Motoyoshi, S.; Tomoda, I.; Shimizu, M.; Shichinohe, K. Decrease in mitogen-induced lymphocyte proliferative responses in cats infected with feline immunodeficiency virus. Nippon Juigaku Zasshi 1990, 52, 573–579. [Google Scholar]
  134. Hosie, M.J.; Jarrett, O. Serological responses of cats to feline immunodeficiency virus. AIDS 1990, 4, 215–220. [Google Scholar] [CrossRef]
  135. Taniguchi, A.; Ishida, T.; Konno, A.; Washizu, T.; Tomoda, I. Altered mitogen response of peripheral blood lymphocytes in different stages of feline immunodeficiency virus infection. Nippon Juigaku Zasshi 1990, 52, 513–518. [Google Scholar]
  136. Taniguchi, A.; Ishida, T.; Washizu, T.; Tomoda, I. Humoral immune response to t cell dependent and independent antigens in cats infected with feline immunodeficiency virus. J Vet Med Sci 1991, 53, 333–335. [Google Scholar] [CrossRef]
  137. Torten, M.; Franchini, M.; Barlough, J.E.; George, J.W.; Mozes, E.; Lutz, H.; Pedersen, N.C. Progressive immune dysfunction in cats experimentally infected with feline immunodeficiency virus. J Virol 1991, 65, 2225–2230. [Google Scholar]
  138. Bishop, S.A.; Williams, N.A.; Gruffydd-Jones, T.J.; Harbour, D.A.; Stokes, C.R. An early defect in primary and secondary t cell responses in asymptomatic cats during acute feline immunodeficiency virus (fiv) infection. Clin Exp Immunol 1992, 90, 491–496. [Google Scholar]
  139. Bishop, S.A.; Williams, N.A.; Gruffydd-Jones, T.J.; Harbour, D.A.; Stokes, C.R. Impaired t-cell priming and proliferation in cats infected with feline immunodeficiency virus. AIDS 1992, 6, 287–293. [Google Scholar] [CrossRef]
  140. Willett, B.J.; Hosie, M.J.; Dunsford, T.H.; Neil, J.C.; Jarrett, O. Productive infection of t-helper lymphocytes with feline immunodeficiency virus is accompanied by reduced expression of cd4. AIDS 1991, 5, 1469–1475. [Google Scholar]
  141. Ohno, K.; Watari, T.; Goitsuka, R.; Tsujimoto, H.; Hasegawa, A. Altered surface antigen expression on peripheral blood mononuclear cells in cats infected with feline immunodeficiency virus. J Vet Med Sci 1992, 54, 517–522. [Google Scholar] [CrossRef]
  142. Rideout, B.A.; Moore, P.F.; Pedersen, N.C. Persistent upregulation of mhc class ii antigen expression on t-lymphocytes from cats experimentally infected with feline immunodeficiency virus. Vet Immunol Immunopathol 1992, 35, 71–81. [Google Scholar] [CrossRef]
  143. Choi, I.S.; Yoo, H.S.; Collisson, E.W. Evaluation of expression patterns of feline cd28 and ctla-4 in feline immunodeficiency virus (fiv)-infected and fiv antigen-induced pbmc. J Vet Sci 2000, 1, 97–103. [Google Scholar]
  144. Mizuno, T.; Goto, Y.; Baba, K.; Masuda, K.; Ohno, K.; Tsujimoto, H. Molecular cloning of feline tumour necrosis factor receptor type i (tnfr i) and expression of tnfr i and tnfr ii in lymphoid cells in cats. Eur J Immunogenet 2003, 30, 107–113. [Google Scholar] [CrossRef]
  145. Nishimura, Y.; Shimojima, M.; Sato, E.; Izumiya, Y.; Tohya, Y.; Mikami, T.; Miyazawa, T. Downmodulation of cd3epsilon expression in cd8alpha+beta- t cells of feline immunodeficiency virus-infected cats. J Gen Virol 2004, 85, 2585–2589. [Google Scholar] [CrossRef]
  146. Hanlon, M.A.; Marr, J.M.; Hayes, K.A.; Mathes, L.E.; Stromberg, P.C.; Ringler, S.; Krakowka, S.; Lafrado, L.J. Loss of neutrophil and natural killer cell function following feline immunodeficiency virus infection. Viral Immunol 1993, 6, 119–124. [Google Scholar] [CrossRef]
  147. Kubes, P.; Heit, B.; van Marle, G.; Johnston, J.B.; Knight, D.; Khan, A.; Power, C. In vivo impairment of neutrophil recruitment during lentivirus infection. J Immunol 2003, 171, 4801–4808. [Google Scholar]
  148. Heit, B.; Jones, G.; Knight, D.; Antony, J.M.; Gill, M.J.; Brown, C.; Power, C.; Kubes, P. Hiv and other lentiviral infections cause defects in neutrophil chemotaxis, recruitment, and cell structure: Immunorestorative effects of granulocyte-macrophage colony-stimulating factor. J Immunol 2006, 177, 6405–6414. [Google Scholar]
  149. Zaccaro, L.; Falcone, M.L.; Silva, S.; Bigalli, L.; Cecchettini, A.; Giorgi, F.; Malvaldi, G.; Bendinelli, M. Defective natural killer cell cytotoxic activity in feline immunodeficiency virus-infected cats. AIDS Res Hum Retroviruses 1995, 11, 747–752. [Google Scholar]
  150. Zhao, Y.; Gebhard, D.; English, R.; Sellon, R.; Tompkins, M.; Tompkins, W. Enhanced expression of novel cd57+cd8+ lak cells from cats infected with feline immunodeficiency virus. J Leukoc Biol 1995, 58, 423–431. [Google Scholar]
  151. Lerner, D.L.; Grant, C.K.; de Parseval, A.; Elder, J.H. Fiv infection of il-2-dependent and -independent feline lymphocyte lines: Host cells range distinctions and specific cytokine upregulation. Vet Immunol Immunopathol 1998, 65, 277–297. [Google Scholar] [CrossRef]
  152. Liang, Y.; Hudson, L.C.; Levy, J.K.; Ritchey, J.W.; Tompkins, W.A.; Tompkins, M.B. T cells overexpressing interferon-gamma and interleukin-10 are found in both the thymus and secondary lymphoid tissues of feline immunodeficiency virus-infected cats. J Infect Dis 2000, 181, 564–575. [Google Scholar]
  153. Orandle, M.S.; Crawford, P.C.; Levy, J.K.; Udoji, R.; Papadi, G.P.; Ciccarone, T.; Mergia, A.; Johnson, C.M. Cd8+ thymic lymphocytes express reduced levels of cd8beta and increased interferon gamma in cats perinatally infected with the jsy3 molecular clone of feline immunodeficiency virus. AIDS Res Hum Retroviruses 2000, 16, 1559–1571. [Google Scholar] [CrossRef]
  154. Ritchey, J.W.; Levy, J.K.; Bliss, S.K.; Tompkins, W.A.; Tompkins, M.B. Constitutive expression of types 1 and 2 cytokines by alveolar macrophages from feline immunodeficiency virus-infected cats. Vet Immunol Immunopathol 2001, 79, 83–100. [Google Scholar]
  155. Dean, G.A.; Bernales, J.A.; Pedersen, N.C. Effect of feline immunodeficiency virus on cytokine response to listeria monocytogenes in vivo. Vet Immunol Immunopathol 1998, 65, 125–138. [Google Scholar]
  156. Lehman, T.L.; O'Halloran, K.P.; Fallon, S.A.; Habermann, L.M.; Campbell, J.A.; Nordone, S.; Dean, G.A.; Hoover, E.A.; Avery, P.R. Altered bone marrow dendritic cell cytokine production to toll-like receptor and cd40 ligation during chronic feline immunodeficiency virus infection. Immunology 2009, 126, 405–412. [Google Scholar] [CrossRef]
  157. Miro, G.; Domenech, A.; Escolar, E.; Collado, V.M.; Tejerizo, G.; De Las Heras, A.; Gomez-Lucia, E. Plasma electrophoretogram in feline immunodeficiency virus (fiv) and/or feline leukaemia virus (felv) infections. J Vet Med A Physiol Pathol Clin Med 2007, 54, 203–209. [Google Scholar] [CrossRef]
  158. Pedersen, N.C. Feline leukemia virus infection. In Feline infectious diseases; Pedersen, N.C., Ed.; American Veterinary Publications: Santa Barbara, CA, 1988. [Google Scholar]
  159. Kohn, B.; Weingart, C.; Eckmann, V.; Ottenjann, M.; Leibold, W. Primary immune-mediated hemolytic anemia in 19 cats: Diagnosis, therapy, and outcome (1998-2004). J Vet Intern Med 2006, 20, 159–166. [Google Scholar]
  160. Anderson, L.J.; Jarrett, W.F. Membranous glomerulonephritis associated with leukaemia in cats. Res Vet Sci 1971, 12, 179–180. [Google Scholar]
  161. Brightman, A.H. , 2nd; Ogilvie, G.K.; Tompkins, M. Ocular disease in felv-positive cats: 11 cases (1981-1986). J Am Vet Med Assoc 1991, 198, 1049–1051. [Google Scholar]
  162. Day, N.K.; O'Reilly-Felice, C.; Hardy, W.D. , Jr.; Good, R.A.; Witkin, S.S. Circulating immune complexes associated with naturally occurring lymphosarcoma in pet cats. J Immunol 1980, 125, 2363–2366. [Google Scholar]
  163. Tuomari, D.L.; Olsen, R.G.; Singh, V.K.; Kraut, E.H. Detection of circulating immune complexes by a clq/protein a-elisa during the preneoplastic stages of feline leukemia virus infection. Vet Immunol Immunopathol 1984, 7, 227–238. [Google Scholar] [CrossRef]
  164. Flynn, J.N.; Cannon, C.A.; Lawrence, C.E.; Jarrett, O. Polyclonal b-cell activation in cats infected with feline immunodeficiency virus. Immunology 1994, 81, 626–630. [Google Scholar]
  165. Matsumoto, H.; Takemura, N.; Sako, T.; Koyama, H.; Motoyoshi, S.; Inada, Y. Serum concentration of circulating immune complexes in cats infected with feline immunodeficiency virus detected by immune adherence hemagglutination method. J Vet Med Sci 1997, 59, 395–396. [Google Scholar]
  166. Quimby, J.M.; Elston, T.; Hawley, J.; Brewer, M.; Miller, A.; Lappin, M.R. Evaluation of the association of bartonella species, feline herpesvirus 1, feline calicivirus, feline leukemia virus and feline immunodeficiency virus with chronic feline gingivostomatits. J Feline Med Surg 2008, 10, 66–72. [Google Scholar] [CrossRef]
  167. Levy, J.K. Feline immunodeficiencyvirus. In Current veterinary therapy xiii; Bonagura, J.D., Ed.; WB Saunders: Philadelphia, 2000. [Google Scholar]
  168. Tenorio, A.P.; Franti, C.E.; Madewell, B.R.; Pedersen, N.C. Chronic oral infections of cats and their relationship to persistent oral carriage of feline calici-, immunodeficiency, or leukemia viruses. Vet Immunol Immunopathol 1991, 29, 1–14. [Google Scholar] [CrossRef]
  169. Reubel, G.H.; George, J.W.; Higgins, J.; Pedersen, N.C. Effect of chronic feline immunodeficiency virus infection on experimental feline calicivirus-induced disease. Vet Microbiol 1994, 39, 335–351. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Hartmann, K. Clinical Aspects of Feline Retroviruses: A Review. Viruses 2012, 4, 2684-2710. https://doi.org/10.3390/v4112684

AMA Style

Hartmann K. Clinical Aspects of Feline Retroviruses: A Review. Viruses. 2012; 4(11):2684-2710. https://doi.org/10.3390/v4112684

Chicago/Turabian Style

Hartmann, Katrin. 2012. "Clinical Aspects of Feline Retroviruses: A Review" Viruses 4, no. 11: 2684-2710. https://doi.org/10.3390/v4112684

Article Metrics

Back to TopTop