Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Neurosci Res ; 65(4): 298-307, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11494365

ABSTRACT

Bone marrow transplantation (BMT) has therapeutic value for twitcher (globoid cell leukodystrophy) mice, which suffer from a genetic deficiency of the lysosomal enzyme galactosylceramidase that leads to progressive demyelination and early death. Preliminary investigations indicated that a semiallogeneic BMT resulted in graft vs. host disease (GVHD) in twitcher mice but not normal mice. Increased production of the cytokine IL-6 has been demonstrated in twitcher mice, and it has been linked with induction of GVHD. We investigated the effects of BMT in twitcher/IL-6 deficient mice and compared these findings with those from transplanted twitcher and control mice. After a semiallogeneic BMT, 11.4% of controls died within few weeks while the rest survived >100 days without GVHD. In contrast, 85% of the transplanted twitcher mice died by 70 days and 65% developed clinical signs of GVHD, e.g., alopecia and weight loss. In transplanted twitcher/IL-6 deficient mice, only 21% died by Day 70, none had alopecia, and 23% had weight loss. There was no difference in the onset day and severity of twitching between twitcher and twitcher/IL-6 deficient mice after BMT. In transplanted twitcher/IL-6 deficient mice, there was improvement of BBB integrity and a decrease in globoid cell number compared with nontransplanted twitcher/IL-6 deficient mice. In summary, these results demonstrate that an underlying pathology like globoid cell leukodystrophy leads to activation of GVHD responses in a donor-host combination that would not normally induce GVHD. Furthermore, IL-6 seems to play a key role because a deficiency of IL-6 results in a better prognosis.


Subject(s)
Bone Marrow Transplantation/immunology , Interleukin-6/genetics , Leukodystrophy, Globoid Cell/immunology , Leukodystrophy, Globoid Cell/therapy , Minor Histocompatibility Antigens/immunology , Animals , Astrocytes/pathology , Blood-Brain Barrier , Body Weight , Brain/blood supply , Brain/immunology , Brain/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Female , Gliosis/pathology , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Immunohistochemistry , Interleukin-6/immunology , Lectins , Leukodystrophy, Globoid Cell/mortality , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Myelin Basic Protein/analysis , Serum Albumin/analysis , Serum Albumin/metabolism , Survival Rate , beta-Galactosidase/genetics
2.
Virology ; 282(1): 123-40, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11259196

ABSTRACT

Previous studies have shown that passage of nonpathogenic SHIV-4 through a series of macaques results in the selection of variants of the virus that are capable of causing rapid subtotal loss of CD4(+) T cells and AIDS within 6-8 months following inoculation into pig-tailed macaques. Using a pathogenic variant of SHIV-4 known as SHIV(KU-1bMC33), we reported that a mutant of this virus with the majority of the vpu deleted was still capable of causing profound CD4(+) T cell loss and neuroAIDS in pig-tailed macaques (McCormick-Davis et al., 2000, Virology 272, 112-116). In this study, we have analyzed the tissue-specific changes in the env and nef in one macaque that developed neuroAIDS (macaque 50 O) and in three macaques that developed only a moderate or no significant loss of CD4(+) T cells and no neurological disease (macaques 50 Y, 20220, 20228) following inoculation with DeltavpuSHIV(KU-1bMC33). Sequence analysis of the gp120 region of env isolated from lymphoid tissues (lymph node and spleen) of macaques 50 Y, 20220, and 20228 revealed no consensus amino acid substitutions. In contrast, analysis of the gp120 sequences isolated from lymphoid and CNS tissues (parietal cortex, basal ganglia, and pons) of macaque 50 O revealed numerous amino acid substitutions. The significance of the amino acid substitutions in gp120 was supported by neutralization assays which showed that the virus isolated from the lymph node of macaque 50 O was neutralization resistant compared to the parental SHIV(KU-1bMC33). Analysis of changes in the nef gene from macaque 50 O revealed in-frame deletions in Nef that ranged from 4 to 13 amino acids in length, whereas the nef genes isolated from the other three macaques revealed no deletions or consensus amino acid substitutions. Inoculation of the virus isolated from the lymph node of the macaque which developed neuroAIDS, SHIV(50OLNV), into four pig-tailed macaques resulted in a severe loss of the circulating CD4(+) T cells within 2 weeks postinoculation, which was maintained for up to 20 weeks postinoculation, confirming that this virus had indeed become more pathogenic in pig-tailed macaques. Taken together, these observations suggest that DeltavpuSHIV(KU-1bMC33) has a low pathogenic phenotype in macaques but that individual pig-tailed macaques can select for additional mutations within the Env and Nef which can compensate for the lack of an intact Vpu and ultimately increase its pathogenicity.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Gene Products, env/genetics , Reassortant Viruses/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/genetics , Acquired Immunodeficiency Syndrome/immunology , Amino Acid Sequence , Animals , Basal Ganglia/virology , Brain/virology , CD4 Lymphocyte Count , Central Nervous System Infections/virology , Consensus Sequence , Disease Models, Animal , Gene Products, nef/genetics , HIV Envelope Protein gp120/genetics , HIV-1/pathogenicity , Human Immunodeficiency Virus Proteins , Leukocytes, Mononuclear/virology , Lymph Nodes/virology , Macaca nemestrina , Molecular Sequence Data , Simian Immunodeficiency Virus/genetics , Spleen/virology , Viral Regulatory and Accessory Proteins/isolation & purification , nef Gene Products, Human Immunodeficiency Virus
3.
Virology ; 272(1): 112-26, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10873754

ABSTRACT

We report on the role of vpu in the pathogenesis of a molecularly cloned simian-human immunodeficiency virus (SHIV(KU-1bMC33)), in which the tat, rev, vpu, env, and nef genes derived from the uncloned SHIV(KU-1b) virus were inserted into the genetic background of parental nonpathogenic SHIV-4. A mutant was constructed (DeltavpuSHIV(KU-1bMC33)) in which 42 of 82 amino acids of Vpu were deleted. Phase partitioning studies revealed that the truncated Vpu was not an integral membrane protein, and pulse-chase culture studies revealed that cells inoculated with DeltavpuSHIV(KU-1bMC33) released viral p27 into the culture medium with slightly reduced kinetics compared with cultures inoculated with SHIV(KU-1bMC33). Inoculation of DeltavpuSHIV(KU-1bMC33) into two pig-tailed macaques resulted in a severe decline of CD4(+) T cells and neurological disease in one macaque and a more moderate decline of CD4(+) T cells in the other macaque. These results indicate that a membrane-bound Vpu is not required for the CD4(+) T cell loss and neurological disease in SHIV-inoculated pig-tailed macaques. Furthermore, because the amino acid substitutions in the Tat and Rev were identical to those previously reported for the nonpathogenic SHIV(PPc), our results indicate that amino acid substitutions in the Env and/or Nef were responsible for the observed CD4(+) T cell loss and neurological disease after inoculation with this molecular clone.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , CD4-Positive T-Lymphocytes/immunology , Capsid Proteins , Central Nervous System Viral Diseases/virology , HIV-1/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/metabolism , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/pathology , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/cytology , Capsid/metabolism , Cell Line , Cell Membrane/metabolism , Central Nervous System/pathology , Central Nervous System/virology , Central Nervous System Viral Diseases/immunology , Cloning, Molecular , DNA, Viral/analysis , DNA, Viral/genetics , Gene Products, gag/metabolism , HIV Envelope Protein gp120/metabolism , HIV-1/genetics , HIV-1/physiology , Human Immunodeficiency Virus Proteins , Humans , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macaca nemestrina , Molecular Sequence Data , Sequence Deletion/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Viral Load , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics , gag Gene Products, Human Immunodeficiency Virus
4.
J Neurosci Res ; 60(2): 231-6, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10740228

ABSTRACT

Globoid cell leukodystrophy (Krabbe's disease) is an autosomal recessive disease that affects the lysosomal enzyme galactosylceramidase. Galactosylceramidase removes galactose from galactosylceramide and psychosine, which are derived from sphingosine. In the present study, L-cycloserine (an inhibitor of 3-ketodyhydrosphingosine synthase) was administered to the twitcher mouse, an authentic model of globoid cell leukodystrophy. Twitcher mice treated with L-cycloserine had a significantly longer life span and a delayed onset of weight loss than vehicle-injected twitcher mice. Pathological features such as macrophage infiltration and astrocyte gliosis also were less in treated twitcher mice. These results indicate that substrate reduction therapy may have therapeutic value for individuals with residual enzymatic activity, e.g., individuals with late onset disease or individuals with partial enzyme replacement via bone marrow transplantation. In these cases, a reduction in galactosylceramide and psychosine synthesis would enable residual enzymatic activity to keep up with the accumulation of these substrates that would otherwise lead to pathology.


Subject(s)
Cycloserine/therapeutic use , Leukodystrophy, Globoid Cell/drug therapy , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Leukodystrophy, Globoid Cell/pathology , Leukodystrophy, Globoid Cell/physiopathology , Mice , Mice, Mutant Strains
5.
Neuropathol Appl Neurobiol ; 25(4): 285-94, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10476045

ABSTRACT

The pattern of neurological disease caused by human immunodeficiency virus (HIV) infection of the central nervous system (CNS) was investigated using a macaque model of acquired immune defiency syndrome (AIDS). Seven of nine macaques inoculated with neurovirulent simian imunodeficiency virus (SIVmac ) developed AIDS within 3 months. Four of these had clinically obvious neurological disease and extensive conduction defects in the form of latency increases in evoked potential (EP) responses. Neuropathologically, all four animals had disseminated white matter disease in the form of multifocal, perivascular and nodular parenchymal mononuclear cell infiltrates, along with extensive involvement of the cortical grey matter, leptomeninges and intracranial portions of cranial nerves. A brisk multinucleated giant cell (MGC) response was a frequent accompaniment in the affected areas. Three of the animals in this group also showed spongiform vacuolation in the occipital grey matter, a lesion described only rarely in HIV encephalitis. In the remaining three animals, there was only minimal evidence of overt neurological impairment or conduction defects. These animals had only mild to moderate neuropathological changes and lesions were virtually confined to the white matter regions of the brain. MGC responses were rare or absent in the CNS of these animals. Neuropathological findings in this SIVmac model have therefore shown good correlation with the severity of clinical and neurophysiological changes, and are reminiscent of HIV-1 encephalitis. More importantly, white matter involvement was a consistent finding in the affected macaques, regardless of the duration and severity of disease, or type of virus inoculated, suggesting an unusual susceptibility for lentiviral infection in these regions of the macaque CNS.


Subject(s)
Nervous System/pathology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/pathogenicity , Animals , Brain/pathology , Chick Embryo , Evoked Potentials/physiology , Female , Humans , Immunohistochemistry , In Situ Hybridization , Infant, Newborn , Macaca mulatta , Macaca nemestrina , Nervous System/physiopathology , Pregnancy , Simian Acquired Immunodeficiency Syndrome/physiopathology , Spinal Cord/pathology
6.
J Med Primatol ; 27(2-3): 59-64, 1998.
Article in English | MEDLINE | ID: mdl-9747944

ABSTRACT

By animal to animal passage in rhesus and pig-tailed macaques, we developed a rhesus model of HIV-1 disease in humans. Rhesus macaques infected with a cell-free stock of SHIVKU-2 developed CD4+ T cell loss, primary lentiviral encephalitis and pneumonia, and AIDS. Six of nine rhesus macaques died within eight months post-inoculation, while the remaining three are at five, five, and eight months post-inoculation, respectively. Animals infected by either mucosal or parenteral routes of infection had a similar course of infection.


Subject(s)
Acquired Immunodeficiency Syndrome/physiopathology , CD4-Positive T-Lymphocytes/immunology , HIV-1/genetics , Reassortant Viruses/genetics , Simian Immunodeficiency Virus/genetics , Acquired Immunodeficiency Syndrome/immunology , Animals , CD4 Lymphocyte Count , Cell Line , Flow Cytometry , Genes, env , Genes, rev , Genes, tat , Genes, vpu , HIV-1/immunology , HIV-1/pathogenicity , Humans , Immune Tolerance , Macaca mulatta , Reassortant Viruses/immunology , Reassortant Viruses/pathogenicity , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , T-Lymphocytes/virology , Virulence
8.
Brain Pathol ; 7(3): 851-61, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9217970

ABSTRACT

We recently reported that a chimeric simian/human immunodeficiency virus (SHIVKU-1) developed in our laboratory caused progressive depletion of CD4+ T lymphocytes and AIDS within 6 months of inoculation into pig-tailed macaques (M. nemestrina). None of the pig-tailed macaques showed productive SHIV infection in the central nervous system (CNS). In this report, we show that by further passage of the pathogenic virus in rhesus macaques [M. mulatta], we have derived a new strain of SHIV (SHIVKU-2) that has caused AIDS and productive CNS infection in 3 of 5 rhesus macaques infected with the virus. Productive replication of SHIV in the CNS was clearly shown by high infectivity titers and p27 protein levels in brain homogenates, and in 2 of the 3 rhesus macaques this was associated with disseminated, nodular, demyelinating lesions, including focal multinucleated giant cell reaction, largely confined to the white matter. These findings were reminiscent of HIV-1 associated neurological disease, and our immunohistochemical and in situ hybridization data indicated that the neuropathological lesions were associated with the presence of SHIV-specific viral antigens and nucleic acid respectively. However, the concomitant reactivation of opportunistic infections in these macaques suggested that such pathogens may have influenced the replication of SHIV in the CNS, or modified the neuropathological sequelae of SHIV infection in the rhesus species, but not in pig-tailed macaques. Our findings in the two species of macaques highlight the complexities of lentiviral neuropathogenesis, the precise mechanisms of which are still elusive.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Central Nervous System Diseases/virology , Chimera/genetics , HIV-1/pathogenicity , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Acquired Immunodeficiency Syndrome/genetics , Animals , Base Sequence , Disease Models, Animal , Female , HIV-1/genetics , Macaca mulatta , Macaca nemestrina , Male , Molecular Sequence Data , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , Virus Replication
9.
AIDS Res Hum Retroviruses ; 13(8): 635-45, 1997 May 20.
Article in English | MEDLINE | ID: mdl-9168232

ABSTRACT

By animal-to-animal passage in macaques we derived a pathogenic chimeric simian-human immunodeficiency virus (SHIV) that caused CD4+ T cell loss and AIDS in pigtail macaques and used it to inoculate 20 rhesus and pigtail macaques by the intravaginal and intravenous routes. On the basis of the outcome of infection and patterns of CD4+ T cell loss and viral load, disease was classified into four patterns: acute, subacute, chronic, and nonprogressive infection. During the study period, 15 of the 20 animals developed fatal disease, including AIDS, encephalitis, pneumonia, and severe anemia. Opportunistic pathogens identified in these animals included Pneumocystis, cytomegalovirus, Cryptosporidium, Toxoplasma, and Candida. No single parameter by itself predicted outcome, although a combination of low CD4+ T cell counts in blood, high plasma virus levels, and presence of autoantibodies to red blood cells reliably predicted a fatal outcome. Five animals (25%) died within 3 months of inoculation and constituted the group with acute disease, whereas the nine animals (45%) with subacute disease died between 3 and 8 months postinoculation. This 70% mortality within 8 months is significantly shorter than in HIV-1-infected human beings, of whom 70% develop fatal disease a decade after infection. SHIV infection in macaques provides a useful model with which to evaluate antiviral strategies, combining all the advantages of the SIVmac system, yet using a virus bearing the envelope gene of HIV-1.


Subject(s)
Acquired Immunodeficiency Syndrome/etiology , Disease Models, Animal , HIV-1/genetics , Macaca mulatta/virology , Macaca nemestrina/virology , Reassortant Viruses/genetics , Simian Immunodeficiency Virus/genetics , AIDS-Related Opportunistic Infections/immunology , AIDS-Related Opportunistic Infections/pathology , Anemia, Hemolytic, Autoimmune/blood , Anemia, Hemolytic, Autoimmune/immunology , Anemia, Hemolytic, Autoimmune/metabolism , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes , Coombs Test , HIV Antibodies/blood , HIV-1/immunology , Humans , Lymphocyte Count , Macaca mulatta/genetics , Macaca mulatta/immunology , Macaca nemestrina/genetics , Macaca nemestrina/immunology , Reassortant Viruses/immunology , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/classification , Simian Acquired Immunodeficiency Syndrome/mortality , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/immunology , Viremia/blood
10.
J Virol ; 71(5): 4016-23, 1997 May.
Article in English | MEDLINE | ID: mdl-9094679

ABSTRACT

Chimeric simian/human immunodeficiency virus (SHIV) consists of the env, vpu, tat, and rev genes of human immunodeficiency virus type 1 (HIV-1) on a background of simian immunodeficiency virus (SIV). We derived a SHIV that caused CD4+ cell loss and AIDS in pig-tailed macaques (S. V. Joag, Z. Li, L. Foresman, E. B. Stephens, L. J. Zhao, I. Adany, D. M. Pinson, H. M. McClure, and O. Narayan, J. Virol. 70:3189-3197, 1996) and used a cell-free stock of this virus (SHIV(KU-1)) to inoculate macaques by the intravaginal route. Macaques developed high virus burdens and severe loss of CD4+ cells within 1 month, even when inoculated with only a single animal infectious dose of the virus by the intravaginal route. The infection was characterized by a burst of virus replication that peaked during the first week following intravenous inoculation and a week later in the intravaginally inoculated animals. Intravaginally inoculated animals died within 6 months, with CD4+ counts of <30/microl in peripheral blood, anemia, weight loss, and opportunistic infections (malaria, toxoplasmosis, cryptosporidiosis, and Pneumocystis carinii pneumonia). To evaluate the kinetics of virus spread, we inoculated macaques intravaginally and euthanized them after 2, 4, 7, and 15 days postinoculation. In situ hybridization and immunocytochemistry revealed cells expressing viral RNA and protein in the vagina, uterus, and pelvic and mesenteric lymph nodes in the macaque euthanized on day 2. By day 4, virus-infected cells had disseminated to the spleen and thymus, and by day 15, global elimination of CD4+ T cells was in full progress. Kinetics of viral replication and CD4+ loss were similar in an animal inoculated with pathogenic SHIV orally. This provides a sexual-transmission model of human AIDS that can be used to study the pathogenesis of mucosal infection and to evaluate the efficacy of vaccines and drugs directed against HIV-1.


Subject(s)
Acquired Immunodeficiency Syndrome/transmission , CD4-Positive T-Lymphocytes/physiology , Mouth Mucosa/virology , Simian Acquired Immunodeficiency Syndrome/transmission , Vagina/virology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , Animals , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Disease Models, Animal , Female , Humans , Lymph Nodes/virology , Macaca mulatta , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology
11.
AIDS Res Hum Retroviruses ; 13(3): 241-6, 1997 Feb 10.
Article in English | MEDLINE | ID: mdl-9115811

ABSTRACT

We evaluated the effects of a reverse transcriptase inhibitor, 9-(2-phosphonylmethoxyethyl)adenine (PMEA), on simian immunodeficiency virus (SIV) infection in rhesus macaques (Macaca mulatta). Four macaques were given PMEA (20 mg/kg) subcutaneously on days 1 and 2 and inoculated with virus on day 2. Drug treatment was continued for 30 consecutive days, after which the virus burdens and course of infection were monitored for a further 6 months. Four control animals that did not receive PMEA all developed high virus burdens and two of the four developed clinical disease. In contrast, virus burdens remained low in three of the four macaques treated with PMEA and all four remained healthy. Our results show that suppression of virus replication early in infection can result in reduced virus burdens for a much longer period.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/pharmacology , Organophosphonates , Simian Acquired Immunodeficiency Syndrome/virology , Viral Load , Adenine/pharmacology , Adenine/therapeutic use , Animals , Antiviral Agents/therapeutic use , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Virus Replication/drug effects
12.
J Virol ; 70(5): 3189-97, 1996 May.
Article in English | MEDLINE | ID: mdl-8627799

ABSTRACT

By animal-to-animal passage of simian/human immunodeficiency virus (SHIV) in pig-tailed macaques, we have developed a macaque model of human immunodeficiency virus type 1 (HIV-1) disease in humans. Passaging was begun with a chimeric virus containing the env gene of HIV-1 HXBc2 and the gag and pol genes of simian immunodeficiency virus SIVmac239. SHIV was passaged serially in cohorts of two macaques each, using bone marrow-to-bone marrow transfers at 5, 5, and 16 weeks for passages 2, 3, and 4, respectively. The fifth passage was done by using cell-free virus isolated from cerebrospinal fluid of a passage 4 macaque. The virus became more virulent with each passage. Virus replication was restricted in all three animals in passages 1 and 2 but not in five of the six animals in passages 3, 4, and 5. In these animals, intense virus replication in the lymphoid tissues resulted in almost total elimination of CD4+ T cells within weeks of inoculation, and three of these animals developed AIDS in less than 1 year. The more uniform virus-host interaction initiated by the cell-free virus in the passage 5 animals contrasted with a more variable pattern of disease initiated by infectious bone marrow cells during earlier passages. The virulent cell-free SHIV can now be used to screen the efficacy of vaccines directed against the envelope of HIV-1.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , CD4-Positive T-Lymphocytes/immunology , Chimera , HIV-1/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Acquired Immunodeficiency Syndrome/pathology , Acquired Immunodeficiency Syndrome/transmission , Animals , Base Sequence , Bone Marrow/virology , CD8-Positive T-Lymphocytes/immunology , DNA Primers , Genes, env , Genes, gag , Genes, pol , HIV-1/genetics , HIV-1/isolation & purification , Humans , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Macaca mulatta , Macaca nemestrina , Molecular Sequence Data , Polymerase Chain Reaction , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Spleen/immunology , Spleen/pathology , Spleen/virology , Thymus Gland/immunology , Thymus Gland/pathology , Thymus Gland/virology , Time Factors
13.
Cell Prolif ; 28(10): 525-31, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7488672

ABSTRACT

This study was designed to evaluate the effects of hypergastrinaemia induced via suppression of gastric acid by omeprazole on carcinogen-induced colon cancer in rats. The carcinogen methylazoxymethanol (MAM), 30 mg/kg, was administered intraperitoneally at 6-weekly intervals to Sprague-Dawley rats. Four weeks after the last MAM injection, the first daily dose of omeprazole, 40 mg/kg, was given by gastric gavage to one group of rats, and the rest were given buffered methylcellulose vehicle. After 10 weeks of daily omeprazole or vehicle, the rats were anaesthetized with ether, blood samples obtained, and animals sacrificed. Gastrin levels in serum from omeprazole-treated rats were elevated nearly six-fold. DNA and RNA levels in gastric mucosa were unchanged by omeprazole, but protein content was somewhat reduced. No biochemical or histological changes related to omeprazole treatment were observed in normal colon. The number of tumours, tumour volumes, and total tumour burden were not significantly different in colons of vehicle- or omeprazole-treated rats. Analysis by flow cytometry revealed that the S phase fraction was lower in tumour cells from omeprazole-treated animals; and that the frequency of DNA aneuploidy was also reduced. The results indicate that while omeprazole-induced suppression of stomach acid in rats elevates levels of gastrin in serum, it does not substantially alter the biochemical or cellular characteristics of carcinogen-induced colon tumours.


Subject(s)
Anti-Ulcer Agents/pharmacology , Carcinogens/toxicity , Cell Cycle/drug effects , Colonic Neoplasms/pathology , Gastric Mucosa/pathology , Methylazoxymethanol Acetate/analogs & derivatives , Omeprazole/pharmacology , Animals , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , DNA Replication/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastrins/analysis , Male , Methylazoxymethanol Acetate/toxicity , Rats , Rats, Sprague-Dawley
14.
Toxicol Pathol ; 23(4): 513-7, 1995.
Article in English | MEDLINE | ID: mdl-7501963

ABSTRACT

Drugs and other chemicals that have the potential to induce or exacerbate systemic autoimmune diseases in humans are of great concern. The aim of this study was to examine the immune-disregulating potential of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) by using the popliteal lymph node (PLN) assay. Chlorpromazine (CPZ) was used as a reference compound for 2 reasons: (a) CPZ is known to elicit a positive response in this assay, and (b) CPZ is a structural analogue of TCDD. Male Sprague-Dawley rats were injected subcutaneously with either TCDD or CPZ into the right hind footpad, whereas vehicle alone was injected into the contralateral footpad. Control rats were injected with vehicle in both hind footpads. Animals were sacrificed on day 7, and their PLNs were removed, weighed, and immersed in 10% formalin. The PLN weight index (the weight ratio of right PLN over left PLN) was significantly higher in both CPZ- and TCDD-treated rats than in controls. Histological examinations of PLNs in the CPZ- and TCDD-treated rats revealed similar morphological changes in both groups (e.g., mild follicular hyperplasia with no evidence of an acute inflammatory response). These results indicate that TCDD has the potential to induce or exacerbate autoimmune-like reactions. Results also suggest that drugs may be useful surrogates to study the mechanism of toxicity of environmental chemicals that cannot be administered to humans.


Subject(s)
Adjuvants, Immunologic/toxicity , Environmental Pollutants/toxicity , Lymph Nodes/drug effects , Polychlorinated Dibenzodioxins/toxicity , Adjuvants, Immunologic/administration & dosage , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Chlorpromazine/administration & dosage , Chlorpromazine/toxicity , Environmental Pollutants/administration & dosage , Environmental Pollutants/immunology , Hindlimb , Injections, Subcutaneous , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Organ Size/drug effects , Polychlorinated Dibenzodioxins/administration & dosage , Polychlorinated Dibenzodioxins/immunology , Rats , Rats, Sprague-Dawley
15.
Gastroenterology ; 108(4): 1068-74, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7698573

ABSTRACT

BACKGROUND/AIMS: Published studies suggest that hypergastrinemia stimulates growth of normal or malignant colon tissue. Other studies dispute these findings. This study was designed to test the hypothesis that hypergastrinemia enhances progression or invasiveness of colon cancer. METHODS: Colonic carcinomas were induced in male Sprague-Dawley rats by six weekly intraperitoneal injections of methylazoxymethanol. Four weeks after the last injection of carcinogen, the animals were randomized into four treatment groups, including vehicle control, low- and high-dose omeprazole, and ranitidine. After 10 weeks of treatment, the animals were bled, stomach weights were recorded, and colon tumors were mapped, enumerated, measured, and scored histopathologically by Dukes' classification. Crypt and mucosal heights were determined in colonic mucosa unaffected by tumor. RESULTS: Drug administration induced a sustained hypergastrinemia that did not enhance tumor burden or invasiveness or crypt height/mucosal height ratios. Ranitidine-treated rats consumed less food, weighed less, and developed fewer tumors. This group also had lower crypt and mucosal heights than rats in the vehicle- or omeprazole-treated rats. CONCLUSIONS: The results suggest that endogenous hypergastrinemia induced by these acid-suppressing drugs has no stimulatory effect on colon mucosal growth or progression or biological behavior of experimental rat colon cancer.


Subject(s)
Colonic Neoplasms/pathology , Gastrins/blood , Omeprazole/adverse effects , Ranitidine/adverse effects , Analysis of Variance , Animals , Colonic Neoplasms/blood , Colonic Neoplasms/chemically induced , Intestinal Mucosa/pathology , Male , Methylazoxymethanol Acetate/analogs & derivatives , Neoplasm Invasiveness , Random Allocation , Rats , Rats, Sprague-Dawley
16.
J Neurovirol ; 1(1): 78-91, 1995 Mar.
Article in English | MEDLINE | ID: mdl-9222344

ABSTRACT

SIVmac239 replicates productivity in activated CD4+ T lymphocytes, but inefficiently in macrophages from rhesus macrophages. Inoculation of the virus into animals results in an acute, highly productive burst of virus replication in activated T lymphocytes in lymphoid tissues and infected cells invade the central nervous system (CNS). This phase lasts a few weeks and is eventually followed by development of immunosuppression of different degrees of severity, opportunistic infections, and tumors related to the loss of T lymphocytes. On rare occasions, infected immunosuppressed animals develop encephalitis and/or interstitial pneumonia, syndromes that are associated with selection of mutant viruses that replicate efficiently in macrophages of these tissues. Usually, however, brains of animals dying with AIDS caused by SIVmac239 appear histologically normal. Is the brain infected with virus? We report here on a macaque dying with AIDS, a neuroinvasive tumor and interstitial pneumonia associated with macrophage-tropic virus. Except for focal infiltration of tumor cells, the brain was normal histologically. We examined the virus and viral DNA from different tissues and found that lymphocytes but not macrophages from lymph nodes and spleen yielded virus, whereas macrophages but not lymphocytes from the lung produced virus. No virus was recovered from the brain but small amounts of viral p27 were present in the brain homogenate. Viral sequences were present in the brain as determined by PCR from tissue DNA. Comparison showed that the viral sequences in the brain closely resembled those from the spleen. Presumably, the virus caused a minimally productive infection detectable by production of small amounts of p27, but was not accompanied by any histopathological changes. It is unclear why the macrophage-tropic virus in the lung failed to 'take-off' in the brain of this animal. To determine whether this virus had encephalitic potential, we inoculated the lung homogenate containing cell-free, macrophage tropic virus into a young pigtail macaque, a species known to be sensitive to primate lentiviral infections. This animal developed severe encephalitis 10 weeks later. Virus from the brain was very similar to the inoculum virus, proving its encephalitic potential. Possible reasons for the differences in neurovirulence of this virus between the two animals remain speculative.


Subject(s)
B-Lymphocytes/virology , Encephalitis, Viral/virology , Macrophages/virology , Simian Immunodeficiency Virus/isolation & purification , T-Lymphocytes/virology , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , Brain/cytology , Brain/immunology , Brain/virology , Carrier State/virology , Cloning, Molecular , Encephalitis, Viral/immunology , Genes, Viral/genetics , Humans , Hybrid Cells/cytology , Hybrid Cells/immunology , Hybrid Cells/virology , Macaca mulatta , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/cytology , Viral Load , Viral Proteins/genetics
17.
J Virol ; 69(2): 1367-9, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7815523

ABSTRACT

We inoculated four rhesus macaques with molecularly cloned simian immunodeficiency virus SIVmac239/17E env, a chimeric virus whose env gene was derived from the brain of an SIV-encephalitic macaque. Blood and lymphoid tissues had high frequencies of infected cells. The virus was neuroinvasive, but productive virus replication did not occur in the brain, and animals did not develop encephalitis.


Subject(s)
Brain/virology , Encephalitis/virology , Genes, env , Macrophages/virology , Simian Immunodeficiency Virus/pathogenicity , Animals , CD4 Lymphocyte Count , Chimera , Gene Products, gag/analysis , Macaca mulatta , Simian Immunodeficiency Virus/genetics , Virulence
18.
J Leukoc Biol ; 56(3): 353-7, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8083607

ABSTRACT

Lymphocyte-tropic (L-tropic) SIVmac predictably causes immunosuppression and AIDS in rhesus macaques. SIV encephalitis, on the other hand, is caused mainly by macrophage-tropic (M-tropic) SIVmac. We have previously described the derivation of M-tropic, neuroadapted SIVmac from molecularly cloned, L-tropic SIVmac239. In this report we show that inoculation of four macaques with neuroadapted virus resulted in L-tropic SIVmac-related diseases in all four but neurological disease in only two of the four animals. Because cocultivation of infected macrophages with CD4+ lymphocytes results in production of tumor necrosis factor alpha and interleukin-6, we asked whether infiltration of supernatant fluids containing these cytokines into the brains of macaques infected with neuroadapted virus would enhance the development of neurological disease. These procedures failed to promote productive virus replication in the brain. Thus, although different degrees of immunosuppression and AIDS could be induced predictably with L-tropic virus, induction of neurological disease was not predictable even when animals were inoculated with neuroadapted M-tropic virus and inflammatory cytokines were infiltrated into the brains of these animals.


Subject(s)
Brain/microbiology , Interleukin-6/pharmacology , Macrophages/microbiology , Monkey Diseases/microbiology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Immunodeficiency Virus/isolation & purification , Tumor Necrosis Factor-alpha/pharmacology , Animals , Biopsy , Brain/pathology , Cells, Cultured , Macaca , Macrophages/pathology , Macrophages/physiology , Monkey Diseases/pathology , Monkey Diseases/physiopathology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/physiology , Virus Replication/drug effects
19.
J Med Primatol ; 23(2-3): 108-16, 1994.
Article in English | MEDLINE | ID: mdl-7966224

ABSTRACT

We studied 15 macaques inoculated with SIV and identified three phases of infection. Phase 1 was characterized by activated lymphocytes in blood and infected cells in the CSF. In phase 2, activated cells were not detected but virus was recovered from mitogen-stimulated PBMC, while in phase 3, virus was recovered from mitogen-stimulated PBMC only after depletion of CD8+ lymphocytes, indicating effective control of the virus in peripheral blood. Early development of phase 3 status correlated with a longer period of clinical normalcy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Animals , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Lymphocyte Count , Lymphocyte Depletion , Macaca mulatta , Predictive Value of Tests , Prognosis , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Immunodeficiency Virus/isolation & purification , Simian Immunodeficiency Virus/pathogenicity , Time Factors , Virus Replication
20.
J Am Vet Med Assoc ; 201(10): 1499; author reply 1501-2, 1992 Nov 15.
Article in English | MEDLINE | ID: mdl-1289324
SELECTION OF CITATIONS
SEARCH DETAIL
...