Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Indian Med Assoc ; 2000 Feb; 98(2): 56-9
Article in English | IMSEAR | ID: sea-103836

ABSTRACT

It is not clear how much genetic factor is responsible for causing cervical cancer, but certainly there has some mutation abnormality in the suppressive p53 gene which is to be proved beyond doubt. In addition to this, there are some environmental factors which make insults on cervical epithelium. Immunology of cancer cervix is well represented by (1) immunology of cancer, (2) immunology of virally controlled antigen and (3) cancer cervix antigen markers. The cancer is an example of transplantation immunology changes in the surface of tumour cells during replication. Human papillomavirus (HPV) DNA has been shown to be associated with high grade cervical intra-epithelial neoplasia (CIN). Squamous cell carcinoma antigen has been investigated for predictive value in cancer cervix. Colony stimulating factor-1 causes monocytic cells aggregation in neoplastic area and plays as initiators of the sequence of inflammation up to neoplasia.


Subject(s)
Antigens, Viral/immunology , Cell Transformation, Neoplastic/immunology , Uterine Cervical Dysplasia/immunology , Female , Humans , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Risk Factors , Tumor Virus Infections/immunology , Uterine Cervical Neoplasms/immunology
2.
Arch. med. res ; 30(4): 269-74, jul.-ago. 1999. ilus, tab
Article in English | LILACS | ID: lil-266529

ABSTRACT

Infection by certain human papillomaviruses (HPV), most notably HPV types 16 and 18, is the mejor risk factor cervical cancer. Worldwide, this disease represents the second most frequent malignant tumor in women; thus, there is urgent need for efficient therapy and prevention. The natural history of cervical cancer and its precursors (cervical intraepithelial neoplasias), as well as animal experiments, strogly suggest that immune system controls both the primary infection (by neutralizing antibodies directed against the major structural protein L1) and the progression o the disease (via cytotoxic T cell specific for the viral oncoproteins expressed in transformed cells, e.g., E7). By the expression of an HPV 16 L1E7 fusion proteins, we have generated chimeric virus-like particles (CVLP). Immunization of mice with CVLPs induces neutralizing antibodies directed against L1 virus-like particles (devoid of the E7 portion) and E7-specific t cells as measured in vitro. Vaccinated animals are protected against tumor growth followings inoculation of syngeneic HPV 16-transformed cells. In addition, we observed a therapeutic effect of vaccination on pre-existing tumors. This data allowed us to concelude that CVLPs are suitable for prevention and therapy of HPV infection. A vaccine based on HPV 16 L1E7 CVLPs is currently under development


Subject(s)
Humans , Animals , Female , Mice , Papillomaviridae/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/prevention & control , Tumor Virus Infections/virology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Virion
3.
Medicina (B.Aires) ; 57(Supl.2): 34-42, Aug. 1997.
Article in English | LILACS | ID: lil-320010

ABSTRACT

All animals, including humans, show differential susceptibility to infection with viruses. Study of the genetics of susceptibility or resistance to specific pathogens is most easily studied in inbred mice. We have been using mouse mammary tumor virus (MMTV), a retrovirus that causes mammary tumors in mice, to study virus/host interactions. These studies have focused on understanding the mechanisms that determine genetic susceptibility to MMTV-induced mammary tumors, the regulation of virus gene expression in vivo and how the virus is transmitted between different cell types. We have found that some endogenous MMTVs are only expressed in lymphoid tissue and that a single base pair change in the long terminal repeat of MMTV determines whether the virus is expressed in mammary gland. This expression in lymphoid cells is necessary for the infectious cycle of MMTV, and both T and B cells express and shed MMTV. Infected lymphocytes are required not only for the initial introduction of MMTV to the mammary gland, but also for virus spread at later times. Without this virus spread, mammary tumorigenesis is dramatically reduced. Mammary tumor incidence is also affected by the genetic background of the mouse and at least one gene that affects infection of both lymphocytes and mammary cells has not yet been identified. The results obtained from these studies will greatly increase our understanding of the genetic mechanisms that viruses use to infect their hosts and how genetic resistance to such viruses in the hosts occurs.


Subject(s)
Animals , Mice , Genetic Predisposition to Disease , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Nucleotides/genetics , Mammary Tumor Virus, Mouse/genetics , Gammaretrovirus/genetics , B-Lymphocytes , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Virus Integration/genetics , Virus Integration/immunology , Carbohydrate Sequence/genetics , T-Lymphocytes , Mammary Tumor Virus, Mouse/immunology , Gammaretrovirus/immunology
4.
Medicina (B.Aires) ; 57(Supl.2): 21-33, Aug. 1997.
Article in Spanish | LILACS | ID: lil-320011

ABSTRACT

Hosts and their pathogens have co-evolved for millions of years, developing multiple and intimate interactions. Vertebrates have evolved a very complex immune system which pathogens have often been able to circumvent, in some cases even managing to appropriate some of its components for their own purpose. Among the pathogens which do use components of the immune system to survive and propagate, those coding for the expression of superantigens (SAgs) are now under intense scrutiny. Investigations concerning one of these pathogens, the mouse mammary tumor virus (MMTV), led to the understanding of how the expression of such components is a critical step in their life cycle. A number of milk-borne exogenous MMTV infect mice shortly after birth and, when expressed, produce superantigens. Herein, we describe the biological effects of new variants of MMTV. Two of these, BALB14 and BALB2 encoding SAgs with V beta 14+ and V beta 2+ specificities, respectively, were present in BALB/c mice of our colony (BALB/cT); a third variant, termed MMTV LA, originated in (BALB/cTxAKR)F1 mice from recombination between BALB 14 and Mtv-7 endogenous provirus. The recombinant LA virus induces the deletion of V beta 6+ and V beta 8.1+ T cells as a consequence of the acquisition of SAg hypervariable coding region of Mtv-7. The SAg encoded by MMTV LA strongly stimulates cognate T cells in vivo leading to a very effective amplification of lymphoid cells in BALB/c mice, correlating with a high incidence of mammary tumors. These results suggest that the presence of non-productive endogenous proviruses--generally considered to confer a selective advantage to the host by protecting it from infection with exogenous MMTVs encoding cross-reactive SAgs--could also be advantageous for the pathogen by increasing its variability, thus broadening the host range and allowing the expansion of highly tumorigenic variants.


Subject(s)
Animals , Female , Mice , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Superantigens/immunology , Gammaretrovirus/immunology , Disease Susceptibility , Genetic Predisposition to Disease , Genome, Viral , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Virus Integration/genetics , Virus Integration/immunology , Mice, Inbred BALB C , RNA-Directed DNA Polymerase , Gammaretrovirus/genetics
5.
Medicina (B.Aires) ; 57(Suppl.2): 34-42, Aug. 1997.
Article in Spanish | LILACS, BINACIS | ID: biblio-1165035

ABSTRACT

All animals, including humans, show differential susceptibility to infection with viruses. Study of the genetics of susceptibility or resistance to specific pathogens is most easily studied in inbred mice. We have been using mouse mammary tumor virus (MMTV), a retrovirus that causes mammary tumors in mice, to study virus/host interactions. These studies have focused on understanding the mechanisms that determine genetic susceptibility to MMTV-induced mammary tumors, the regulation of virus gene expression in vivo and how the virus is transmitted between different cell types. We have found that some endogenous MMTVs are only expressed in lymphoid tissue and that a single base pair change in the long terminal repeat of MMTV determines whether the virus is expressed in mammary gland. This expression in lymphoid cells is necessary for the infectious cycle of MMTV, and both T and B cells express and shed MMTV. Infected lymphocytes are required not only for the initial introduction of MMTV to the mammary gland, but also for virus spread at later times. Without this virus spread, mammary tumorigenesis is dramatically reduced. Mammary tumor incidence is also affected by the genetic background of the mouse and at least one gene that affects infection of both lymphocytes and mammary cells has not yet been identified. The results obtained from these studies will greatly increase our understanding of the genetic mechanisms that viruses use to infect their hosts and how genetic resistance to such viruses in the hosts occurs.


Subject(s)
Animals , Gammaretrovirus/genetics , Tumor Virus Infections/genetics , Retroviridae Infections/genetics , Nucleotides/genetics , Genetic Predisposition to Disease , Mammary Tumor Virus, Mouse/genetics , Gammaretrovirus/immunology , Tumor Virus Infections/immunology , Retroviridae Infections/immunology , Virus Integration/genetics , Virus Integration/immunology , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Carbohydrate Sequence/genetics , Mammary Tumor Virus, Mouse/immunology
6.
Anon.
Medicina (B.Aires) ; 57(supl.2): 43-52, 1997.
Article in English | LILACS | ID: lil-321743

ABSTRACT

Mouse mammary tumor virus (MMTV) is a retrovirus which can induce mammary carcinomas in mice late in life by activation of proto-oncogenes after integration in their vicinity. Surprisingly, it requires a functional immune system to achieve efficient infection of the mammary gland. This requirement became clear when it was discovered that it has developed strategies to exploit the immune response. Instead of escaping immune detection, it induces a vigorous polyclonal T-B interaction which is required to induce a chronic infection. This is achieved by activating and then infecting antigen presenting cells (B cells), expressing a superantigen on their cell surface and triggering unlimited help by the large number of superantigen-specific T cells. The end result of this strong T-B interaction is the proliferation and differentiation of the infected B cells leading to their long term survival.


Subject(s)
Animals , Female , Mice , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Virus Integration/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse/immunology , B-Lymphocytes , Proto-Oncogenes , Superantigens/immunology , T-Lymphocytes
7.
Medicina (B.Aires) ; 57(3): 327-31, 1997. tab, graf
Article in Spanish | LILACS | ID: lil-209650

ABSTRACT

En este trabajo se describen los efectos biológicos de nuevas variantes virales de MMTV exógenos. Una de ellas denominada BALB14 está presente en la cepa BALB/c e induce un bajo porcentaje de adecarcinomas mamarios. Otra de ellas, (MMTV-7) se originó por recombinación entre BALB14 y transcriptos derivados del provirus endógeno Mtv-7. El desarrollo de una línea de ratones BALB/c infectada con ambas variantes virales a través del amamantamiento con una nodriza F1 (BABL/cxAKR) en la que surge el MMTV-7, permitió demonstrar que el virus recombinante - que expressa el SAg del provirus endógeno - es amplificado en los huéspedes BALB/c y resulta en ellos altamente tumorigénico. Se discute el rol de la adquisición de superantígenos estimulatorios en la amplificación del virus recombinante. Los resultados obtenidos permiten hipotetizar que la presencia en el genoma del ratón de provirus endógenos no productivos - considerada hasta ahora como protectora frente a la infección con virus MMTV exógenos - oferecía además ventajas selectivas para el virus al aumentar su variabilidad poblacional, permitiendo así la ampliación del rango de huéspedes susceptibles y la expanción de variantes con alta patogenicidad.


Subject(s)
Mice , Animals , Carcinogenicity Tests , In Vitro Techniques , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse/immunology , Retroviridae Infections/immunology , Superantigens , Tumor Virus Infections/immunology , Mice, Inbred BALB C , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL