Studies using the SCID-hu mouse showed similar abnormalities [19]

Studies using the SCID-hu mouse showed similar abnormalities [19]. Damage to the thymic epithelium may alter the thymic microenvironment and contribute to the immune suppression observed in acquired immune deficiency syndrome (AIDS) patients and models. Importantly, it has been observed that thymic epithelial fragments from AIDS children arrest T cell differentiation of normal bone marrow-derived CD34+ stem cells in vitro[25]. Similarly, HIV-1 infection has been shown to interrupt thymopoiesis in vivo in the SCID-hu mouse model [26]. The thymus releases mature lymphocytes into the periphery of the immune system. This

function can Erlotinib datasheet be evaluated through analysis of recent thymic emigrants (RTEs) [27], that themselves can be estimated by the presence of T cell receptor excision circles (Trecs), circular DNA fragments derived from the rearrangement of TCR genes, that remain within RTEs

[28]. Trec analysis in HIV and simian immunodefiency virus (SIV) infections revealed decreased numbers of Trec+ T lymphocytes in the peripheral blood compared with uninfected individuals [29,30]. Interestingly, specific highly active anti-retroviral therapy seems to correct this defect in AIDS patients [31]. Another important feature is that the thymic secretory function is also affected in HIV-infected individuals, as the blood levels INCB018424 clinical trial of thymic peptides are abnormal [23]. For example, thymosin α1 levels are elevated in many patients with AIDS, especially in the early stages PD-1 antibody inhibitor [23,32]. In contrast, a consistent and long-term diminution of thymulin secretion has been documented in AIDS patients, in terms of both serum levels and intrathymic contents of the hormone [24,33,34]. It is known that mouse hepatitis viruses (MHV), which are members of the Coronaviridae family, show a tropism to thymic stromal cells [35] and T lymphocytes [36]. Otherwise, thymus involution was described in MHV-A59-infected BALB/c mice

[37]. That involution was characterized by a severe transient atrophy resulting from apoptosis of immature CD4+CD8+ T cells that might be caused by infection of a small proportion of TEC. Marked thymic involution characterized by striking diminution of thymus weight and cellularity was also observed in CBA mice infected intraperitoneally with MHV-3, together with a significant decrease in thymocyte subpopulations and significant numbers of apoptotic cells [38]. In humans, Trec quantification revealed an impairment of RTEs, reflecting a thymic dysfunction in hepatitis C virus (HCV)-infected patients [39]. Measles, a member of the Paramyxoviridae family, is generally followed by immune suppression with transient lymphopenia and impaired cell-mediated immunity [40,41]. Impaired thymic function seems to contribute to measles virus-induced immune suppression. Indeed, measles virus infects TEC and monocytes in the thymus of humans and monkeys [42,43], leading to a decrease in the size of the thymic cortex [44,45].

Comments are closed.