Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Leukemia ; 19(2): 176-82, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15592433

ABSTRACT

CD33 is expressed on the malignant blast cells in most cases of acute myeloid leukemia (AML) but not on normal hematopoietic pluripotent stem cells. Antibody-based therapies for AML have, therefore, focused on CD33 as a suitable tumor-associated target antigen. The most promising results have been obtained with gemtuzumab ozogamicin (GO, Mylotarg), a humanized IgG(4) anti-CD33 monoclonal antibody joined to a calicheamicin-gamma(1) derivative. Engagement of CD33 by GO results in immunoconjugate internalization and hydrolytic release of the toxic calicheamicin moiety, which, in turn, causes DNA damage and cell death. Since 2000, when GO was approved for clinical use, treatment trials and pilot studies have revealed potential expanded applications along with additional limitations. At the same time, correlative biological and in vitro functional studies have further characterized CD33 expression patterns in AML, the significance of CD33-antibody interactions, pathways involved in GO-induced cytotoxicity and potential drug resistance mechanisms. This review summarizes the recent data addressing mechanisms of GO action and discusses their relevance with regard to clinical applications and the limitations of using experimental model systems to mimic in vivo conditions. As the first drug conjugate approved for clinical use, GO serves as an important paradigm for other immunoconjugates against internalizing tumor antigens.


Subject(s)
Aminoglycosides/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Leukemia, Myeloid, Acute/drug therapy , Antibodies, Monoclonal, Humanized , Cell Survival/immunology , Drug Resistance, Neoplasm/immunology , Gemtuzumab , Humans , Sialic Acid Binding Ig-like Lectin 3
3.
Blood ; 98(4): 988-94, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11493443

ABSTRACT

Expression of multidrug resistance (MDR) features by acute myeloid leukemia (AML) cells predicts a poor response to many treatments. The MDR phenotype often correlates with expression of P-glycoprotein (Pgp), and Pgp antagonists such as cyclosporine (CSA) have been used as chemosensitizing agents in AML. Gemtuzumab ozogamicin, an immunoconjugate of an anti-CD33 antibody linked to calicheamicin, is effective monotherapy for CD33(+) relapsed AML. However, the contribution of Pgp to gemtuzumab ozogamicin resistance is poorly defined. In this study, blast cell samples from relapsed AML patients eligible for gemtuzumab ozogamicin clinical trials were assayed for Pgp surface expression and Pgp function using a dye efflux assay. In most cases, surface expression of Pgp correlated with Pgp function, as indicated by elevated dye efflux that was inhibited by CSA. Among samples from patients who either failed to clear marrow blasts or failed to achieve remission, 72% or 52%, respectively, exhibited CSA-sensitive dye efflux compared with 29% (P =.003) or 24% (P <.001) among samples from responders. In vitro gemtuzumab ozogamicin--induced apoptosis was also evaluated using an annexin V--based assay. Low levels of drug-induced apoptosis were associated with CSA-sensitive dye efflux, whereas higher levels correlated strongly with achievement of remission and marrow blast clearance. In vitro drug-induced apoptosis could be increased by CSA in 14 (29%) of 49 samples exhibiting low apoptosis in the absence of CSA. Together, these findings indicate that Pgp plays a role in clinical resistance to gemtuzumab ozogamicin and suggest that treatment trials combining gemtuzumab ozogamicin with MDR reversal agents are warranted. (Blood. 2001;98:988-994)


Subject(s)
Aminoglycosides , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Clinical Trials, Phase II as Topic , Drug Resistance, Multiple/genetics , Drug Resistance, Multiple/immunology , Leukemia, Myeloid/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Acute Disease , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Bone Marrow/pathology , Carbocyanines/pharmacokinetics , Cyclosporine/pharmacology , Drug Synergism , Fluorescent Dyes , Gemtuzumab , Humans , Immunotoxins/pharmacology , Leukemia, Myeloid/pathology , Leukocytes, Mononuclear/pathology , Phenotype , Regression Analysis , Remission Induction , Treatment Outcome , Tumor Cells, Cultured/drug effects
4.
Am J Hematol ; 64(4): 242-50, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10911375

ABSTRACT

Congenital dysfibrinogenemia is a rare cause of unexplained thrombosis. However, most individuals with dysfibrinogenemia are asymptomatic, suggesting that co-morbid factors contribute to thrombo-embolic events. The potential roles of additional genetic or acquired prothrombotic risk factors are poorly understood because detailed family studies are lacking. Herein, we describe a family whose propositus was a young Caucasian man with recurrent venous thrombo-emboli and dysfibrinogenemia due to heterozygosity for an Arg-->Cys substitution at residue 275 in the gamma-chain. The only additional thrombophilic abnormality found in the proband was heterozygosity for a G/A transition at position -455 in the fibrinogen beta-chain promoter; a genotype associated with high acute phase levels of fibrinogen. The proband's father, who died of a cerebral artery thrombosis, carried the gammaR275C substitution but not the beta-promoter -455 variant. Among 14 living relatives, eight were heterozygous for one or the other mutation and only one, a 21-year-old niece, was dually affected. None had suffered bleeding or thrombosis. In vitro studies of the proband's purified fibrinogen revealed markedly abnormal thrombin-catalyzed polymerization and delayed fibrin clot lysis by tPA-activated plasmin. We hypothesize that the gammaR275C substitution predisposes to thrombosis by generating clots that are relatively resistant to fibrinolysis. The clinical risk is low, however, in the absence of an additional thrombophilic mutation. The beta-promoter variant could, theoretically, contribute to this risk by augmenting expression of the dysfibrinogen under conditions of stress. Like the common hereditary thrombophilias, heterozygous familial dysfibrinogenemia induces thrombosis in the setting of multiple prothrombotic influences.


Subject(s)
Fibrinogen/genetics , Point Mutation , Thrombosis/genetics , Adult , Aged , Alleles , Amino Acid Substitution , Female , Humans , Male , Middle Aged , Pedigree , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Thrombosis/etiology
5.
Vet Immunol Immunopathol ; 70(3-4): 223-43, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10507363

ABSTRACT

Tumorigenesis occurs through a multistep process initiated by genetic lesions and facilitated by endogenous and external growth/survival signals. In many malignancies, specific oncogenic mutations correlate with phenotypic characteristics, inferring lineage-specific pathogenic mechanisms. To characterize these relationships in a unique feline tumor, we studied primary cells and two-cell lines independently-derived from a thymic lymphoma that contained and actively expressed a novel feline leukemia virus (FeLV) recombinant with transduced host Notch2 sequences. All three tumor cell populations contained similar FeLV/Notch2 proviral variants and phenotypically resembled mature thymocytes. Multiple Notch2 transcripts were expressed in the cell lines, including species that correspond to viral genomes and spliced subgenomic viral mRNA. Tumor cell line FeLV/Notch2 virus was packaged into virions; however, the variant was not efficiently transmitted to feline cells in vitro. Primary tumor cells constitutively expressed mRNA for interleukin-4 (IL-4), IL-6 and the p40 subunit of IL-12. Lymphokine mRNA was not detected in established tumor cell lines nor was T-cell growth-promoting activity found in culture supernatants. Exogenous IL-4 enhanced primary tumor cell survival, but inhibited proliferation of the cell lines. Interleukin-4 abrogated hydrocortisone-induced apoptosis in all three populations and had divergent effects on cell line clonogenic colony formation. Exogenous IL-7 and, to a lesser degree, IL-6 also had variable positive effects on the growth and viability of the tumor cell populations. Collectively, these data suggest that thymocytes are susceptible to the transforming potential of dysregulated Notch2 and that thymopoietic factors could, through overlapping and distinct mechanisms, promote the survival and outgrowth of FeLV/Notch2-containing neoplastic cells.


Subject(s)
Cat Diseases/virology , Leukemia Virus, Feline , Lymphokines/pharmacology , Receptors, Cell Surface/analysis , Retroviridae Infections/veterinary , Thymus Neoplasms/veterinary , Tumor Virus Infections/veterinary , Animals , Apoptosis/drug effects , Cat Diseases/pathology , Cats , Cell Survival/drug effects , Cell Transformation, Neoplastic , Hydrocortisone/pharmacology , Interleukin-4/pharmacology , Interleukin-6/pharmacology , Interleukin-7/pharmacology , Phenotype , Receptor, Notch2 , Retroviridae Infections/pathology , Retroviridae Infections/virology , Thymus Neoplasms/pathology , Thymus Neoplasms/virology , Tumor Cells, Cultured , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
6.
Vet Immunol Immunopathol ; 72(3-4): 343-68, 1999 Dec 30.
Article in English | MEDLINE | ID: mdl-10628677

ABSTRACT

To identify potential roles of cytokines in retroviral pathogenesis, we used reverse transcription-quantitative competitive polymerase chain reaction (RT-qcPCR) assays to characterize mRNA levels of 19 different lymphokines, chemokines, monokines and hematopoietic growth factors in three feline cell lines productively infected with subgroup A feline leukemia virus (FeLV-A) or various feline immunodeficiency virus (FIV) strains. Infection of a CD8+, CD5- large granular lymphocyte (LGL) cell line with FeLV-A activated expression of interleukin-7 (IL-7), induced modest (4-fold) increases in granulocyte/macrophage colony-stimulating factor (GM-CSF) and leukemia inhibitory factor (LIF) transcripts, and decreased transforming growth factor-beta (TGF-beta) mRNA (4-fold). The LGL cells were not susceptible to infection by FIV. Infection of MYA-1 cells, a CD4+ T-lymphoblastoid cell line, with FeLV-A activated expression of macrophage inflammatory protein-1alpha (MIP-1alpha), increased transcript levels of GM-CSF (8-fold), macrophage CSF (M-CSF) (16-fold) and stem cell factor (SCF) (250-fold), and decreased (4-fold) expression of IL-10 and tumor necrosis factor-alpha (TNF-alpha). Productive infection with four different FIV molecular clones caused progressive MYA-1 cell death; however, the mRNA expression profiles were unchanged except for 2- to 4-fold increases in M-CSF and 16- to 500-fold increases in SCF. Thus, FIV-induced MYA-1 cytopathicity was not associated with dysregulation of pro-apoptotic or survival factor transcript levels. Lastly, productive infection of PNI cells, a marrow-derived fibroendothelial cell line, with FeLV-A or any of three FIV strains induced 4-fold higher levels of IL-12p40 transcripts and variably higher levels (4- to 64-fold) of GM-CSF. Two viral strains, the FIV-14 molecular clone and the clinical isolate FIV-5122, caused syncytia formation and unique activation of IL-1beta and stromal cell-derived factor-1 (SDF-1) expression, suggesting a potential role for those factors in viral spread and/or cytopathicity. In addition, infection with FIV-5122, but not the other FIV strains or FeLV-A, induced significant increases in mRNA levels of the hematopoietic inhibitors TNF-alpha and MIP-1alpha, along with increased concentrations of soluble proteins in culture supernatants. Consistent with this, supernatant from FIV-5122 infected PNI cells suppressed hematopoietic progenitor growth in colony assays, compared to supportive activities in supernatants from other infected or uninfected PNI cell cultures. Together, these data demonstrate that feline retroviruses alter cytokine mRNA levels in general and strain-specific patterns. These changes may result in specific alterations in cell function and contribute to retroviral pathogenesis. Our observations provide a basis for directed studies of candidate factors within the hematopoietic, thymic and lymphoid microenvironments.


Subject(s)
Cytokines/biosynthesis , Leukemia Virus, Feline/pathogenicity , Leukemia, Feline/metabolism , Animals , Cats , Cell Line , Cloning, Molecular , Cytokines/genetics , Cytopathogenic Effect, Viral , Leukemia Virus, Feline/metabolism , Polymerase Chain Reaction/veterinary
7.
Leukemia ; 10(12): 1867-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8946923

ABSTRACT

Feline leukemia virus (FeLV) infection in cats serves as a valuable animal model system for understanding the mechanisms of human diseases such as cancer and immunodeficiency. We have used experimental infection with molecularly cloned viruses to isolate and characterize novel FeLV variants that evolved in vivo and that were associated with the development of thymic lymphoma. One variant, FeLV-81T, contained a mutated envelope gene that conferred cytopathicity, enhanced replication rate, and syncytium induction in feline T cells, and is reminiscent of immunodeficiency-inducing strains of FeLV. Another variant transduced a portion of the feline Notch2 gene, which was expressed as a novel truncated protein in the cell nucleus and which we believe functioned as an oncogene in the development of T cell malignancy. Understanding how FeLV variants that either stimulate or destroy lymphocytes evolve and interrelate during disease progression will help elucidate the mechanisms of retroviral pathogenicity.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Leukemia Virus, Feline/genetics , Lymphoma, T-Cell/virology , Retroviridae Infections/genetics , Retroviridae Infections/virology , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , Animals , Cats , Disease Models, Animal , Feline Acquired Immunodeficiency Syndrome/virology , Humans
8.
J Virol ; 70(11): 8071-80, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8892932

ABSTRACT

Feline leukemia virus (FeLV) is thought to induce neoplastic diseases in infected cats by a variety of mechanisms, including the transduction of host proto-oncogenes. While FeLV recombinants that encode cellular sequences have been isolated from tumors of naturally infected animals, the acquisition of an unrelated host gene has never been documented in an experimental FeLV infection. We isolated recombinant FeLV proviruses encoding feline Notch2 sequences from thymic lymphoma DNA of two cats inoculated with the molecularly cloned virus FeLV-61E. Four recombinant genomes were identified, three in one cat and one in the other. Each had similar but distinct transduction junctions, and in all cases, the insertions replaced most of the envelope gene with a region of Notch2 that included the intracellular ankyrin repeat functional domain. The product of the FeLV/Notch2 recombinant provirus was a novel, truncated 65- to 70-kD Notch2 protein that was targeted to the cell nucleus. This virally encoded Notch2 protein, which resembles previously constructed, constitutively activated forms of Notch, was apparently expressed from a subgenomic transcript spliced at the normal envelope donor and acceptor sequences. The data reported here implicate a nuclear, activated Notch2 protein in FeLV-induced leukemogenesis.


Subject(s)
Cat Diseases/virology , Leukemia Virus, Feline/genetics , Lymphoma/veterinary , Receptors, Cell Surface/genetics , Retroviridae Infections/veterinary , Thymus Neoplasms/veterinary , Amino Acid Sequence , Animals , Base Sequence , Cats , Cell Line , Cloning, Molecular , DNA, Viral , Gene Rearrangement , Genome, Viral , Lymphoma/virology , Molecular Sequence Data , Proviruses , RNA, Viral/analysis , Rats , Receptor, Notch2 , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombination, Genetic , Retroviridae Infections/virology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Thymus Neoplasms/virology , Transfection , Tumor Virus Infections/veterinary , Tumor Virus Infections/virology
10.
Semin Vet Med Surg Small Anim ; 10(4): 220-33, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8820596

ABSTRACT

Feline patients with unexplained peripheral blood cytopenias, circulating immature or neoplastic cells, dysplastic or dysmorphic bone marrow abnormalities, and/or lymphoid tumors are likely suffering from an underlying retroviral infection with feline leukemia virus (FeLV) and/or feline immunodeficiency virus (FIV). Cytopenic hematologic disorders are often caused by the direct or indirect hematosuppressive effects of these retroviruses. Alternatively, secondary infections, nutritional deficiencies, and/or hematopoietic neoplasms may be important cofactors in the development of blood and bone marrow abnormalities in retrovirus-positive patients. Mild to moderate nonregenerative anemia, with or without concurrent granulocytopenia and/or thrombocytopenia, is one of the most frequent hematologic disorders encountered with either infectious agent. Severe, isolated anemia with absent reticulocytes (pure red blood cell aplasia) specifically suggests infection with FeLV subgroup C. Hemolytic (regenerative) anemia, more commonly associated with FeLV infection, may be caused by an autoimmune process and/or coinfection with Haemobartonella felis. Lymphopenia is a hallmark of chronic, symptomatic FIV infection. Neutropenia may accompany a panleukopenia-like syndrome in FeLV-positive cats or it may be associated with acute primary infection or an adverse drug effect in the FIV-infected patient. FeLV and, to a lesser extent, FIV are both causally related to lymphoid neoplasms in domestic cats, but with dissimilar epidemiologic, clinical, and host cell phenotypic features. Clinicians must be cognizant of the wide spectrum of hematologic manifestations of FeLV and FIV infections to recognize and appropriately manage these complications in their feline patients.


Subject(s)
Cat Diseases/etiology , Feline Acquired Immunodeficiency Syndrome/complications , Hematologic Diseases/veterinary , Leukemia, Feline/complications , Animals , Cats , Hematologic Diseases/etiology
11.
Exp Hematol ; 23(10): 1104-14, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7544739

ABSTRACT

To characterize the production of stem cell factor (SCF, the ligand for the c-kit receptor protein) and its regulation by inflammatory cytokines and glucocorticoids, primary marrow stromal fibroblasts were isolated from normal individuals and two patients with Diamond-Blackfan anemia. Unstimulated normal marrow stromal fibroblasts constitutively expressed a low level of SCF mRNA (9 +/- 2 copies/cell [mean +/- SEM]), continually secreted soluble SCF into the supernatant of 1- to 5-day-old cultures (0.16 +/- 0.02 to 0.73 +/- 0.04 ng/mL per 10(6) cells, respectively), and expressed membrane-bound SCF. Stimulation with interleukin-1 beta (IL-1 beta) only modestly increased SCF mRNA levels, soluble SCF production at 24 hours, and membrane-bound SCF. In comparison, hydrocortisone or tumor necrosis factor alpha (TNF-alpha) exposure increased SCF mRNA levels 3.5- to four-fold above controls, but with different kinetics. The peak TNF-alpha effect was at 6 hours, with return to near control levels at 24 hours, whereas hydrocortisone induced maximal mRNA increases at 12 to 18 hours, and the levels remained high at 24 hours. Similarly, a sustained increase in soluble SCF production was detected during 1 to 5 days of hydrocortisone exposure (0.27 +/- 0.03 to 1.10 +/- 0.08 ng/mL per 10(6) cells), while TNF-alpha stimulation modestly increased the production of soluble SCF in 24-hour cultures only. Unstimulated normal marrow fibroblasts expressed predominantly the long species of alternatively spliced SCF mRNA, and the relative amounts of long and short mRNAs did not change after stimulation with IL-1 beta, hydrocortisone, or TNF-alpha. SCF production by marrow stromal fibroblasts from a symptomatic patient with Diamond-Blackfan anemia was equivalent to simultaneously studied normal marrow fibroblasts. In contrast, marrow fibroblasts from a Diamond-Blackfan anemia patient in untreated hematologic remission constitutively expressed high levels of SCF mRNA (21 +/- 4 copies/cell) and soluble protein (0.40 ng/mL per 10(6) cells at 24 hours). Together, these observations suggest that SCF is constitutively produced by fibroblasts in the human marrow microenvironment and that hydrocortisone induces a modest but sustained increase in SCF gene expression and protein production, compared to only a transient increase induced by TNF-alpha. In addition, these findings support the hypothesis that endogenous or corticosteroid-induced increases in the production of SCF could play a physiologic role in the clinical improvement of congenital anemia.


Subject(s)
Bone Marrow/metabolism , Fanconi Anemia/metabolism , Hematopoietic Cell Growth Factors/biosynthesis , Adult , Bone Marrow Cells , Fibroblasts/metabolism , Gene Expression/drug effects , Hematopoiesis , Hematopoietic Cell Growth Factors/genetics , Humans , Hydrocortisone/pharmacology , Interleukin-1/pharmacology , Interleukin-6/genetics , RNA, Messenger/genetics , Stem Cell Factor , Tumor Necrosis Factor-alpha/pharmacology
12.
Exp Hematol ; 23(10): 1069-79, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7656928

ABSTRACT

To determine the role of infected marrow accessory cells in the pathogenesis of viral-associated hematologic disorders, we evaluated whether feline leukemia virus (FeLV) infection alters the cytoadhesive properties of long-term marrow culture (LTMC) stromal cells, the support of stromal-associated progenitors in LTMCs, and the production of progenitor growth-promoting and -inhibiting activities by marrow stromal cells. Our previous studies demonstrated that LTMCs containing FeLV-infected stromal cells generated two- to three-fold higher numbers of total nonadherent cells and nonadherent granulocyte-macrophage progenitors (CFU-GM) compared with uninfected LTMCs. In the present studies, CFU-GM and primitive erythroid progenitors (BFU-E) bound equivalently to FeLV-infected or uninfected LTMC stromal cells in a 2-hour adherence assay. In recharge LTMC studies, the numbers of adherent CFU-GM maintained in cultures containing stromal cells infected with FeLV-A/61E were not significantly different from controls (range 84-191% of uninfected control cultures, p > 0.1); however, the percentages of adherent CFU-GM in S phase of the cell cycle were consistently increased (range 42-62% compared with controls, range 5-23%). FeLV infection had no significant effect on the cell-cycle status of the nonadherent CFU-GM in LTMCs. Agar co-culture assays revealed that multilineage colony-stimulating activity was constitutively and equivalently produced by feeder cell layers consisting of either uninfected or FeLV-infected irradiated heterogeneous LTMC stromal cells, homogeneous marrow stromal fibroblasts, or a fibroendothelial marrow stromal cell line. However, FeLV infection significantly attenuated the soluble progenitor growth-inhibitory activity associated with higher densities of these stromal cells. Assays of conditioned medium from cultures of irradiated stromal cells demonstrated that FeLV infection or hydrocortisone exposure decreased the utilization of glucose, the production of acidic metabolic products, and the constitutive production of active and latent transforming growth factor beta (TGF-beta) bioactivity and TGF-beta 2 immunoreactivity. Levels of macrophage inflammatory protein 1 alpha (MIP-1 alpha) and tumor necrosis factor alpha (TNF-alpha) were undetectable and unchanged in CM samples. Together, these observations suggest that downmodulation of TGF-alpha and/or the basal metabolic status of stromal cells may be responsible for the high basal proliferative activity of adherent CFU-GM in FeLV-infected LTMCs, and by extension, that retroviral infection in vivo could alter hematopoiesis by perturbing the progenitor growth-regulatory and -supportive function of marrow stromal cells.


Subject(s)
Bone Marrow/pathology , Growth Inhibitors/metabolism , Leukemia Virus, Feline/pathogenicity , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Animals , Cats , Cell Cycle , Cytokines/metabolism , Down-Regulation , Hematopoiesis , Hydrocortisone/pharmacology , Transforming Growth Factor beta/metabolism
13.
J Vet Intern Med ; 9(3): 133-40, 1995.
Article in English | MEDLINE | ID: mdl-7674214

ABSTRACT

Prospective studies were performed over a 28- to 77-month period (median, 66 months) on 5 cats with naturally acquired feline immunodeficiency virus (FIV) infection in an attempt to correlate hematologic and clinicopathologic changes with the emergence of clinical disease. On presentation, all cats were asymptomatic; free of opportunistic infections; and had normal complete blood counts, bone marrow morphologies, marrow progenitor frequencies, and progenitor in vitro growth characteristics. During study, 2 cats remained healthy, 2 cats showed mild clinical signs, and 1 cat developed a malignant neoplasm (ie, bronchiolar-alveolar adenocarcinoma). Although persistent hematologic abnormalities were not observed, intermittent peripheral leukopenias were common. In 3 of 5 FIV-seropositive cats, lymphopenia (< 1,500 lymphs/microL; normal reference range, 1,500 to 7,000 lymphs/microL) was a frequent finding and the absolute lymphocyte counts had a tendency to progressively decline. One of the other 2 cats had consistently low to low-normal absolute neutrophil counts (1,300 to 4,800 segs/microL; mean, 2,730 segs/microL; normal reference range, 2,500 to 12,500 segs/microL), and the remaining cat had consistently normal leukograms, except for a transient period (ie, 11 months) of benign lymphocytosis (7,200 to 13,430 lymphs/microL) early in the study. Periodic examinations of bone marrow aspirates revealed normal to slightly depressed myeloid-to-erythroid ratios with normal cellular morphology and maturation. Bone marrow abnormalities observed late in the study included mild dysmorphic changes (ie, megaloblastic features) in 2 cats, and a significant decrease (60% of controls, P < .001) in the frequencies of burst-forming units erythroid (BFU-E) in marrow cultures of FIV-seropositive cats compared with uninfected control cats.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Feline Acquired Immunodeficiency Syndrome/blood , Feline Acquired Immunodeficiency Syndrome/physiopathology , Animals , Bone Marrow/pathology , Cats , Culture Techniques , Feline Acquired Immunodeficiency Syndrome/pathology , Female , Immunodeficiency Virus, Feline/physiology , Male , Prospective Studies
14.
Baillieres Clin Haematol ; 8(1): 73-112, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7663052

ABSTRACT

Feline oncornavirus and lentivirus infections have provided useful models to characterize the virus and host cell factors involved in a variety of marrow suppressive disorders and haematological malignancies. Exciting recent progress has been made in the characterization of the viral genotypic features involved in FeLV-associated diseases. Molecular studies have clearly defined the causal role of variant FeLV env gene determinants in two disorders: the T-lymphocyte cytopathicity and the clinical acute immunosuppression induced by the FeLV-FAIDS variant and the pure red cell aplasia induced by FeLV-C/Sarma. Variant or enFeLV env sequences also appear to play a role in FeLV-associated lymphomas. Additional studies are required to determine the host cell processes that are perturbed by these variant env gene products. In the case of the FeLV-FAIDS variant, the aberrant env gene products appear to impair superinfection interference, resulting in accumulation of unintegrated viral DNA and cell death. In other cases it is likely that the viral env proteins interact with host products that are important in cell viability and/or proliferation. Understanding of these mechanisms will therefore provide insights to factors involved in normal lymphohaematopoiesis. Similarly, studies of FeLV-induced haematological neoplasms should reveal recombination or rearrangement events involving as yet unidentified host gene sequences that encode products involved in normal cell growth regulation. These sequences may include novel protoncogenes or sequences homologous to genes implicated in human haematological malignancies. The haematological consequences of FIV are quite similar to those associated with HIV. As with HIV, FIV does not appear to directly infect myeloid or erythroid precursors, and the mechanisms of marrow suppression likely involve virus, viral antigen, and/or infected accessory cells in the marrow microenvironment. Studies using in vitro experimental models are required to define the effects of each of these microenvironmental elements on haematopoietic progenitors. As little is known about the molecular mechanisms of FIV pathogenesis, additional studies of disease-inducing FIV strains are needed to identify the genotypic features that correlate with virulent phenotypic features. Finally, experimental FIV infection in cats provides the opportunity to correlate in vivo virological and haematological changes with in vitro observations in a large animal model that closely mimics HIV infection in man.


Subject(s)
Feline Acquired Immunodeficiency Syndrome , Immunodeficiency Virus, Feline/physiology , Leukemia Virus, Feline/physiology , Leukemia, Feline , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Bone Marrow/pathology , Bone Marrow/virology , Cats/virology , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/transmission , Genes, Viral , Immunity, Cellular , Immunodeficiency Virus, Feline/genetics , Immunodeficiency Virus, Feline/immunology , Leukemia Virus, Feline/classification , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/immunology , Leukemia, Feline/immunology , Leukemia, Feline/transmission , Lymphoma/epidemiology , Lymphoma/veterinary , Lymphoma/virology , Myelodysplastic Syndromes/veterinary , Myelodysplastic Syndromes/virology , Red-Cell Aplasia, Pure/veterinary , Red-Cell Aplasia, Pure/virology , Retroviridae/classification , Retroviridae Proteins/genetics , Retroviridae Proteins/physiology , Spumavirus/pathogenicity
15.
Blood ; 85(4): 941-51, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7849316

ABSTRACT

Severe neutropenia and bone marrow (BM) morphologic abnormalities occur during experimentally induced primary infection with feline immunodeficiency virus (FIV), a lentivirus biologically similar to human immunodeficiency virus (HIV). To further characterize the mechanisms involved in this acute infection model of lentivirus-induced BM suppression, peripheral blood counts, histologic BM studies, and BM culture assays were performed on 12 cats that underwent necropsy at regular intervals postinoculation (PI) with FIV Petaluma. Plasma viremia developed at week 3 PI and neutropenia was initially detected at week 6 PI. Low neutrophil counts, but normal hematocrits and platelet counts, persisted through week 12 PI. Infected BM mononuclear cells and megakaryocytes were identified by in situ hybridization assays for FIV nucleic acids in BM sections of cats that underwent necropsy at weeks 4 to 12 PI, correlating with detection of soluble FIV p24 antigen and identification of infected mononuclear and macrophage cells in BM buffy-coat cell cultures from these cats. At weeks 1.5 to 4 PI, the mean frequencies (number per 10(5) BM mononuclear cells) of erythroid progenitors (erythroid colony-forming units [CFU-E] and erythroid burst-forming units [BFU-E] and granulocyte/macrophage progenitors (CFU-granulocyte/macrophage [CFU-GM]) were increased to 508 +/- 74, 143 +/- 24, and 110 +/- 17, respectively (n = 5 cats) as compared with controls (172 +/- 24, 86 +/- 26, and 44 +/- 10; n = 3 cats; P < .02), and the percentages of progenitors in the DNA-synthetic phase of the cell cycle were equivalent to controls. In contrast, the progenitor frequencies at weeks 6 to 12 PI were significantly decreased (72 +/- 16, 43 +/- 6, and 19 +/- 4, respectively; n = 7 cats; P < .01), and these progenitors were more frequently in S-phase. Autologous serum significantly inhibited (P < .05) the growth of CFU-GM in 6 of 9 cats and failed to support the maximal growth of BFU-E in 4 of 9 cats studied at weeks 4 to 12 PI, whereas no such abnormalities were observed in colony assays containing autologous sera from control cats (n = 3) or cats studied at weeks 1.5 or 3 PI (n = 3). In comparison, sera from FIV-infected cats did not inhibit the growth of normal, allogeneic progenitors. However, FIV serum frequently failed to support maximal in vitro growth of normal CFU-GM as compared with uninfected allogeneic sera, further suggesting a lack of progenitor growth-promoting substances in infected cat sera.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Bone Marrow/virology , Feline Acquired Immunodeficiency Syndrome/pathology , Feline Acquired Immunodeficiency Syndrome/physiopathology , Hematopoiesis , Hematopoietic Stem Cells/pathology , Immunodeficiency Virus, Feline/physiology , Neutropenia/etiology , Animals , Bone Marrow/pathology , Bone Marrow/physiopathology , Cats , Cell Cycle , Cells, Cultured , DNA, Viral/analysis , Feline Acquired Immunodeficiency Syndrome/blood , Immunodeficiency Virus, Feline/isolation & purification , In Situ Hybridization , Kinetics , RNA, Viral/analysis , Time Factors , Viremia/blood , Viremia/pathology , Viremia/physiopathology
16.
J Virol ; 68(4): 2458-67, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8139030

ABSTRACT

In order to study retroviral variation, selection, and viral correlates of in vivo pathogenicity, we documented the evolution of feline leukemia virus (FeLV) variants in cats that died with thymic lymphoma after infection with molecularly cloned subgroup A FeLV. Using genomic DNA from cat necropsy samples, we employed PCR to amplify and clone the envelope gene, which is a major determinant of the specific pathogenicity of different FeLV variants. In the envelope gene, mutations encoded scattered amino acid changes that did not cluster into clearly definable variable regions; however, characterization of these terminal variant sequences revealed a predominance of G-to-A and A-to-G nucleotide substitutions. Additionally, some cats harbored variants with recombinant subgroup B-like envelope genes, while the major variant from one cat had a 12-bp insertion in a region previously characterized as an immunodeficiency-inducing determinant. Finally, proviruses from tumor DNA frequently possessed envelope genes predicted to encode a protein truncated in the N-terminal half because of either premature termination codons or deletions ranging from 29 to 1,666 bp. In contrast, all envelope genes cloned from the bone marrow of one cat were predicted to encode full-length envelope product, and only a minority of proviral clones from a cat that did not develop a tumor had defective envelope genes. Thus, in the cat, viruses evolved from subgroup A FeLV that had point mutations, insertions, deletions, or recombinant envelope genes. Furthermore, defective variants were particularly prominent in T-cell tumors.


Subject(s)
Genes, env/genetics , Leukemia Virus, Feline/genetics , Lymphoma/microbiology , Retroviridae Infections/microbiology , Thymus Neoplasms/microbiology , Tumor Virus Infections/microbiology , Amino Acid Sequence , Animals , Base Sequence , Cats , Cloning, Molecular , Genetic Variation , Genome, Viral , Molecular Sequence Data , Mutagenesis , Proviruses/genetics , Sequence Analysis, DNA , Sequence Deletion , Sequence Homology, Amino Acid
17.
Blood ; 82(7): 2096-103, 1993 Oct 01.
Article in English | MEDLINE | ID: mdl-8400259

ABSTRACT

The kinetics of hematopoietic stem cells were investigated in glucose-6-phosphate dehydrogenase (G-6-PD) heterozygous cats treated with dimethylbusulfan. Because of X-chromosome inactivation during embryogenesis, each somatic cell from these animals contains either maternal- or paternal-type G-6-PD. Therefore, all hematopoietic progenitor cells carry the G-6-PD phenotype of the most primitive cell (stem cell) from which they originate. For up to 6.5 years after dimethylbusulfan therapy, we determined the percentages of erythroid and granulocyte/macrophage progenitor cells with each G-6-PD phenotype. Significant variations were seen in studies from five of six cats, showing that the population of stem cells contributing to hematopoiesis was neither large nor constant. With mathematical analyses, we estimated that the proliferative potential of residual stem cells was much less than that of normal stem cells reduced in number by autologous transplantation (Abkowitz et al, Proc Natl Acad Sci USA 87:9062, 1990). There was no evidence for the regeneration of a normal stem cell reserve over time; rather, damage was most pronounced years after dimethylbusulfan exposure. These data may help explain the high clinical incidence of aplastic anemia and myelodysplasia after alkylating agent therapies.


Subject(s)
Busulfan/analogs & derivatives , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Cells , Busulfan/pharmacology , Cats , Cells, Cultured , Clone Cells , Female , Glucosephosphate Dehydrogenase/analysis , Glucosephosphate Dehydrogenase/genetics , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Phenotype , Probability , Stochastic Processes , Time Factors
18.
Exp Hematol ; 20(8): 1022-7, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1324184

ABSTRACT

Marrow stromal fibroblasts (FBs) likely play an important role in the regulation of hematopoiesis within the marrow microenvironment. Infection of these cells by feline leukemia virus (FeLV) might not only contribute to the pathogenesis of FeLV-induced hematologic diseases, but could provide a reservoir for virus in the infected cat. To determine the frequency of FeLV infection among marrow FB precursor cells (fibroblast colony-forming units, CFU-F) of cats viremic with FeLV-C/Sarma and FeLV-A/61E, marrow FBs and FB cell clones were isolated and assayed for expression of FeLV gag protein. From 30% to 86% and 64% to 88% of marrow FB precursors were infected with FeLV-C/Sarma and FeLV-A/61E, respectively. CFU-F from a cat viremic with FeLV-A/61E were not affected by exposure to antibody against FeLV envelope glycoprotein gp70 and heterologous complement, whereas similarly treated hematopoietic progenitors (erythroid colony-forming units, CFU-E; erythroid burst-forming units, BFU-E; and granulocyte-macrophage colony-forming units, CFU-GM) and culture-propagated, FeLV-infected marrow FBs were effectively lysed, suggesting that infected CFU-F within the marrow microenvironment do not express a significant amount of gp70 on their cell membranes. Thus, marrow FB precursor cells appear to be a major target for FeLV in vivo. Furthermore, the low level of gp70 antigen expression on the surface of these cells in vivo may allow them to escape immune surveillance and provide a reservoir of virus during active or latent infection.


Subject(s)
Bone Marrow/microbiology , Bone Marrow/pathology , Fibroblasts/microbiology , Leukemia Virus, Feline/isolation & purification , Leukemia, Experimental/microbiology , Animals , Bone Marrow/metabolism , Cats , Cell Death/physiology , Complement System Proteins/physiology , Female , Fibroblasts/metabolism , Gene Expression Regulation, Leukemic/genetics , Gene Products, gag/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Hematopoietic Stem Cells/pathology , Leukemia, Experimental/genetics , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/metabolism , Tumor Cells, Cultured/microbiology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
19.
Blood ; 80(3): 651-62, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1322201

ABSTRACT

To study the effects of feline leukemia virus (FeLV) on the hematopoietic microenvironment, a two-step feline long-term marrow culture (LTMC) system was developed and characterized. The adherent, stromal layer of these cultures is composed of fibroblastoid cells (50% to 80%), macrophages (10% to 30%), fat cells (10% to 20%), and large, polygonal cells that express muscle actin (1% to 2%). When fresh, enriched marrow mononuclear cells (MMNC) were added to 3-week-old irradiated stromal cultures, nonadherent erythroid progenitors (BFU-E) and granulocyte/macrophage progenitors (CFU-GM) could be detected for up to 5 and 12 weeks, respectively. LTMC stromal layers established from marrow cells from cats viremic with either a nonpathogenic strain of FeLV (FeLV-A/61E) or the anemogenic strain FeLV-C/Sarma were morphologically equivalent to uninfected LTMC stromal layers, although more than 80% of the stromal cells expressed FeLV gag protein. When FeLV-infected stromal cultures were recharged with uninfected MMNC, altered patterns of hematopoiesis were observed, compared with recharged, uninfected stromal cultures. In cultures with infected stroma, fewer nonadherent cells (NAC), nonadherent BFU-E, and nonadherent CFU-GM were detected during the first 4 to 5 weeks after recharge. In contrast, greater numbers of NAC and nonadherent CFU-GM were found from weeks 5 to 12 after recharge. When FeLV-infected stromal cultures were recharged with MMNC from a cat heterozygous for the X-chromosome-linked enzyme glucose-6-phosphate dehydrogenase (G-6-PD), the percentage of nonadherent CFU-GM expressing the domestic type G-6-PD isoenzyme remained stable over time (mean % domestic [%d], 53% +/- 3%), and was equivalent to that of nonadherent CFU-GM maintained in uninfected cultures (mean %d, 56% +/- 3%), indicating that clonal drift or clonal selection was not responsible for the enhanced maintenance of CFU-GM. Furthermore, as only 10% to 20% of recharged hematopoietic cells became infected with FeLV in vitro, it is unlikely that the altered pattern was due to progenitor infection. We hypothesize that the increase in NAC and nonadherent CFU-GM in FeLV-infected cultures resulted from enhanced growth factor production by stromal cells. The two-step LTMC system may facilitate the characterization of stromal-derived factors that affect progenitor cell engraftment and proliferation.


Subject(s)
Bone Marrow/physiology , Hematopoiesis , Hematopoietic Stem Cells/physiology , Leukemia Virus, Feline , Leukemia, Experimental/physiopathology , Acid Phosphatase/analysis , Alkaline Phosphatase/analysis , Animals , Biomarkers , Bone Marrow/physiopathology , Bone Marrow Cells , Carboxylesterase , Carboxylic Ester Hydrolases/analysis , Cats , Cell Adhesion , Cells, Cultured , Collagen/analysis , Culture Techniques/methods , Female , Fibronectins/analysis , Fluorescent Antibody Technique , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Laminin/analysis , Leukemia, Experimental/pathology , Male , Peroxidase/analysis , Reference Values , Time Factors , von Willebrand Factor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...