Your browser doesn't support javascript.
loading
Mycobacterium tuberculosis Methyltransferase Rv1515c Can Suppress Host Defense Mechanisms by Modulating Immune Functions Utilizing a Multipronged Mechanism.
Rani, Anshu; Alam, Anwar; Ahmad, Faraz; P, Manjunath; Saurabh, Abhinav; Zarin, Sheeba; Mitra, Dipendra Kumar; Hasnain, Seyed E; Ehtesham, Nasreen Z.
Afiliação
  • Rani A; Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT-D), New Delhi, India.
  • Alam A; ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.
  • Ahmad F; ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.
  • P M; ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.
  • Saurabh A; ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.
  • Zarin S; Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India.
  • Mitra DK; ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.
  • Hasnain SE; Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India.
  • Ehtesham NZ; Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi (IIT-D), New Delhi, India.
Front Mol Biosci ; 9: 906387, 2022.
Article em En | MEDLINE | ID: mdl-35813825
ABSTRACT
Mycobacterium tuberculosis (M. tb) gene Rv1515c encodes a conserved hypothetical protein exclusively present within organisms of MTB complex and absent in non-pathogenic mycobacteria. In silico analysis revealed that Rv1515c contain S-adenosylmethionine binding site and methyltransferase domain. The DNA binding and DNA methyltransferase activity of Rv1515c was confirmed in vitro. Knock-in of Rv1515c in a model mycobacteria M. smegmatis (M. s_Rv1515c) resulted in remarkable physiological and morphological changes and conferred the recombinant strain with an ability to adapt to various stress conditions, including resistance to TB drugs. M. s_Rv1515c was phagocytosed at a greater rate and displayed extended intra-macrophage survival in vitro. Recombinant M. s_Rv1515c contributed to enhanced virulence by suppressing the host defense mechanisms including RNS and ROS production, and apoptotic clearance. M. s_Rv1515c, while suppressing the phagolysosomal maturation, modulated pro-inflammatory cytokine production and also inhibited antigen presentation by downregulating the expression of MHC-I/MHC-II and co-stimulatory signals CD80 and CD86. Mice infected with M. s_Rv1515c produced more Treg cells than vector control (M. s_Vc) and exhibited reduced effector T cell responses, along-with reduced expression of macrophage activation markers in the chronic phase of infection. M. s_Rv1515c was able to survive in the major organs of mice up to 7 weeks post-infection. These results indicate a crucial role of Rv1515c in M. tb pathogenesis.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article