Page 75 - Read Online
P. 75

Jyonouchi. J Transl Genet Genom 2023;7:274-90  https://dx.doi.org/10.20517/jtgg.2023.32   Page 282

               JAK inhibitors have also become increasingly popular for treating various autoimmune conditions,
               including rheumatoid arthritis (RA), inflammatory bowel diseases (IBD), atopic dermatitis, etc. The actions
               of a new generation of JAK inhibitors are more selective for specific JAKs, as opposed to the 1st generation
               JAK inhibitors. In ASD subjects in whom type 1 IFN-induced neuroinflammation plays a role in their
               behavioral symptoms or cognitive dysfunctions, orally available JAK inhibitors may provide an additional
               treatment option. Interestingly, multiple open-label studies have reported the beneficial effects of baricitinib
               for severe COVID-19 . With the increase in reports of the favorable effects of baricitinib, the FDA issued
                                 [79]
               an EUA (emergency use authorization) for its use as an adjunct treatment, with remdesivir, for hospitalized
               COVID-19 patients older than 2 years of age. In ASD patients who have a risk of more significant
               neuroinflammation following COVID-19, baricitinib may also offer another treatment option for
               controlling post-COVID inflammation.


               Immunodeficiency affecting PI3K/Akt/mTOR pathways
               PI3k/Akt/mTOR pathways are crucial for immune functions, especially differentiation of Th17 vs. Treg cell
               pathways, but these pathways are also crucial for neuronal cell activation and neuronal plasticity, as shown
                                                          [80]
               in patients diagnosed with tuberous sclerosis (TS) . Dysregulation of these signaling pathways caused by
               multiple pathogenic gene variants including TS complex 1/2 are now categorized as activated PI3Kinase
                                                            [81]
               delta syndrome with highly variable clinical features . As expected, apart from the immune system, this
               condition affects the nervous system, frequently exhibiting neurodevelopmental delay and learning
               disabilities along with autoimmune and autoinflammatory complications [81-83] .


               Class IA PI3kinase (PI3K) molecules consist of a p110 catalytic subunit and a regulatory subunit, and
               catalyze phosphatidylinositol 4.5 bisphosphate to the phosphatidylinositol 2,3,4 trisphosphate (PIP3), which
                                                                                         [84]
               serves as a secondary messenger for cell proliferation, activation, and differentiation . Its catalytic unit
               (p110) is coded by genes  PIK3CA, PIK3CB, and PIK3CD. PIK3CD is predominantly expressed in
                                                                                   [84]
               leukocytes . Its regulatory subunits are coded by PIK3R1, PIK3R2, and PIK3R3 . AD, GOF mutations of
                        [84]
               PIK3CD (Activated PI3Kinase delta syndrome (APDS) 1) or AD, LOF mutations of PIK3R1 (APDS2) can
               cause overactivation of PI3K and downstream Akt/mTOR pathways in immune cells and other lineage cells.
               This results in complex clinical features of immunodeficiency and autoimmune/autoinflammatory
               symptoms, illustrating the importance of balanced activation in this signaling pathway in immune cells [81,85] .
               These patients present with clinical features overlapping with both type 1 interferonopathies and
               CAPS [81,85,86] . Regulatory proteins for PI3K activation also affect this signaling pathway and pathogenic
               variants coding for such regulatory molecules cause clinical features resembling APDS: such conditions are
               often referred to as APDS-like syndrome. LOF PTEN pathogenic variants are best known for causing
               APDS-like syndrome .
                                 [85]
               (1) APDS1 and APDS2: As described above, the clinical features of APDS1/APDS2 are highly variable.
               However, in general, these patients present with combined immunodeficiency with evidence of impaired
               humoral and cellular immunity. Impaired humoral immunity is typically suspected when they suffer from
               recurrent sinopulmonary infection caused by community-acquired organisms covered by infant vaccines
                                                                                               [83]
               (Prevnar and Hib). Ocular infection is reported to be frequently seen in APDS patients . Impaired
               immunity exerted by T cells and innate immune cells is also impaired, as evidenced by an increase in the
               frequency of viral and opportunistic infection: asymptomatic viremia of CMV and EBV is common , as is
                                                                                                   [83]
               persistent or recurrent herpes infection . Chronic viral infection may lead to benign lymphadenopathy and
                                                [87]
               hepatosplenomegaly [83,86] . In addition to symptoms associated with immunodeficiency, APDS patients also
               manifest autoimmune conditions typically after the first decade of life: cytopenia and glomerulonephritis
               are common, along with an increase in the risk of malignant complications . Since both APDS1/APDS2
                                                                                [88]
   70   71   72   73   74   75   76   77   78   79   80