3.1. NAD+ Metabolism in Infectious Diseases
The PARP family in humans comprises 17 members, with PARP1 being the most prevalent and extensively researched. PARP1 has crucial functions in programmed cell death, DNA repair, replication, and transcription. DNA strand breaks lead to elevated PARP1 activity, which results in NAD+ depletion and initiates an inflammatory response [
50,
51,
52]. In hepatitis B virus-infected cells, heightened oxidative stress induced double-strand DNA (dsDNA) breaks, subsequently activating PARP1. Upon activation, PARP1 recognized the damaged dsDNA and promoted repair through the non-homologous end joining (NHEJ) pathway [
53]. This process increased NAD+ consumption, suggesting a progressive NAD+ depletion. DNA ligation errors during repair occasionally caused virus-host DNA fusion events, potentially contributing to hepatocellular carcinoma development. In HIV-infected cells stimulated by TNFα, PARP1 overexpression led to a swift reduction in intracellular NAD+ levels. Notably, suppression of PARP1 expression was shown to inhibit viral replication, although the precise mechanisms remained uncharacterized [
54]. Another study demonstrated that PARP1 inactivation in human monocyte-derived macrophages (MDM) also suppressed HIV replication. This inhibition occurred via disruption of long terminal repeat sequences (LTRs) and GTPase activity, which consequently reduced actin cytoskeletal rearrangements [
55]. These findings underscore the multifaceted role of PARP1 in viral pathogenesis and its potential as a therapeutic target. Beyond PARP1, other PARP family members contribute to DNA repair processes during infections. For instance, disturbances in NAD+/NADH redox homeostasis in Mycobacterium tuberculosis (Mtb)-infected cells resulted in bacterial growth arrest and hindered mitochondrial aerobic respiration [
56]. PARP9 expression was significantly upregulated in both Mtb-infected human and mouse cells. PARP9 modulated Mtb susceptibility by downregulating cGAS and mitochondrial oxidative stress-induced type I interferon (IFN) production, thereby reducing bacterial proliferation. Additionally, PARP9 interacted with DTX3L, a protein known to protect cells from DNA damage [
57].
Similarly, the SIRT family comprises seven members, with SIRT1 emerging as a focal point in antiviral research. SIRT1 serves as both a metabolic sensor and a regulator of transcription factors, modulating pathways involved in glycolipid metabolism, DNA replication, and inflammatory responses [
58]. In hepatitis C virus (HCV)-infected human hepatocellular carcinoma cells (HepG2), overexpression of the HCV core protein induced oxidative stress, leading to a reduction in the NAD+/NADH ratio and downregulation of the SIRT1-AMPK pathway. This downregulation decreased the expression of genes associated with glucose and lipid metabolism, contributing to metabolic disturbances in hepatocytes [
59]. In HBV infection, SIRT1 upregulated viral replication by modulating the transcription factor AP-1. Inhibition of SIRT1 with sirtinol significantly suppressed HBV DNA replication, indicating that SIRT1 inhibitors hold promise as a potential treatment for HBV [
60]. Conversely, in Kaposi’s sarcoma-associated herpesvirus (KSHV) infection, a reduction in NAD+ levels lowered SIRT1 activity, which subsequently induced the expression of the transcriptional activator RTA, facilitating viral lysis and replication [
61]. In septic mouse models, a marked decrease in NAD+ levels inhibited the NAD+/SIRT1 pathway in the hippocampus, while inflammatory pathways such as NF-κB and P38-MAPK were upregulated. Supplementation with NMN, a precursor of NAD+, restored NAD+/SIRT1 pathway activity, thereby attenuating the inflammatory response [
62]. In addition to SIRT1, SIRT3 activity also relies heavily on NAD+ levels. [
63]. In macrophages infected with Mycobacterium tuberculosis (M.tb), the downregulation of SIRT3 leads to reduced expression of enzymes involved in central metabolism and components of the electron transport chain. This reduction results in increased mitochondrial ROS production and subsequent cell necrosis. Studies using Sirt3-deficient (Sirt3 -/-) mice infected with M.tb further demonstrate that SIRT3 plays a protective role in defending the host against M.tb infection [
64].
Finally, CD38, which is expressed in a variety of immune cells, is strongly induced to regulate NAD+ levels during infection and inflammation [
65]. In HBV-specific CD8+ T cells, CD38 overexpression led to NAD+ depletion and the dysregulation of DNA repair mechanisms. As a result, the enzymatic activity of SIRTs was reduced, leading to mitochondrial dysfunction, increased ROS production, and DNA damage [
66]. In HIV patients, increased CD38 activity in CD4+ T cells depleted NAD+, decreased SIRT activity, and impaired oxidative phosphorylation, ultimately causing mitochondrial dysfunction and T cell depletion. CD38 catalytic products increased intracellular Ca
2+ as well, exacerbating mitochondrial oxidative stress and contributing to T cell depletion [
67]. In RSV-infected mononuclear-derived dendritic cells (MDDCs), activation of the CD38-cADPR axis led to increased production of type I interferons, initiating an antiviral immune response. However, the precise role of the inflammatory response induced by Ca
2+ channels activated by CD38 requires further investigation [
41]. The changes of NAD+ levels in different infectious diseases are given in
Table 1.
3.2. NAD+ Metabolism in COVID-19
NAD+ exhibits significant metabolic imbalances at various stages of COVID-19. During the acute phase, viral replication and cellular stress disrupt cellular energy metabolism, leading to an inflammatory response and elevated NAD+ consumption [
80]. This reduction in NAD+ is identified as a risk factor for severe COVID-19 [
81]. Besides, the intense inflammatory response and oxidative stress during this phase exacerbate the depletion of NAD+, which in turn causes cellular metabolic disorders, impaired organ function, and respiratory failure, along with other complications [
82]. Interestingly, chronic fatigue in long COVID patients is linked to persistently low levels of NAD+ [
83]. This deficiency affects cellular repair mechanisms, leading to chronic inflammation, mitochondrial dysfunction, and metabolic abnormalities [
84]. The levels of NAD+ metabolites in the blood of COVID-19 patients decreased as the disease worsened. Concurrently, cytokines such as IL-6, IL-10, IL-8, M-CSF, and IL-1α showed elevated levels, suggesting that disturbances in NAD+ metabolism were closely associated with the host immune response [
85]. On top of that, SARS-CoV-2 infection interfered with NAD+ metabolism in a mouse model, evidenced by increased expression of NAD+-depleting enzymes such as RARPs (RARP9, RARP10, RARP14) and the CD38 gene, along with downregulation of SIRT1 expression [
86]. This suggested that, following SARS-CoV-2 infection, host-pathogen interactions may disrupt NAD+ metabolism by modulating the activities of these enzymes, leading to massive NAD+ depletion.
Once SARS-CoV-2 binds to the ACE2 receptor, it enters the cell and releases its RNA genome [
87]. Host cells recognize viral RNA through pattern recognition receptors, such as TLR, RIG-I, and MDA5, which activate the production of IFN, especially IFN-α and IFN-β [
88] (
Figure 2). This interferon signaling then triggers the transcription and activation of PARP1, which plays a role in DNA repair and inhibition of viral replication. However, overactivation of PARP1 reduces NAD+ and NMN levels, causing cellular metabolic dysfunction, cell death, and even tissue damage [
80]. In addition, the excessive activation of PARP1 indirectly reduces SIRT1 activity [
89]. This inhibition of SIRT1 disrupts energy metabolism homeostasis and activates NF-κB, which further induces inflammatory responses, contributing to the production of chemokine storms. Inhibition of SIRT1 also leads to NLRP3 overactivation, triggering a cytokine storm [
58]. Inflammatory pathways regulated by SIRT1, such as NRF2/HMOX1, are also suppressed, which diminishes antioxidant defense functions and reduces the ability to inhibit viral replication [
90]. Interestingly, SIRT5 was implicated in SARS-CoV-2 infection, as it interacted with NSP14, a viral protein of SARS-CoV-2, to promote viral replication [
91]. In contrast, another study found that NSP14 interacted with SIRT5 to regulate host protein succinylation after SARS-CoV-2 infection, inhibiting viral replication [
92]. Although these two studies present contradictory findings, they collectively suggest that SIRT5 regulates innate immunity during SARS-CoV-2 infection. The above highlights the complex relationship between NAD+ depletion, PARP activation, and the role of SIRTs in innate immunity during viral infections.
Along with the aforementioned immune modulators, CD38 may play a crucial role in innate and adaptive immunity during COVID-19. Studies have shown that CD38 expression on immune cells, such as T cells and monocytes, is elevated in COVID-19 patients, particularly in acute and critically ill individuals. This upregulation of CD38 expression may be associated with the severity of the disease [
93,
94]. Upon SARS-CoV-2 entries into host cells via the ACE2 receptor, angiotensin II (Ang II) binds to its receptor, activating CD38. This activation stimulates the release of Ca
2+ from calcium channel proteins, leading to increased cytoplasmic Ca
2+ concentrations, elevated ROS, and activation of type I IFN and IFN-stimulated gene (ISG) pathways. Subsequently, the NF-κB signaling pathway is activated, triggering NLRP3 activation and the release of large quantities of cytokines and chemokines, thereby inducing a cytokine storm [
82]. CD38 is implicated in the innate immune response to SARS-CoV-2 infection by regulating Ca
2+ homeostasis. Moreover, HLA-DR+CD38hiCD8+ T cells accumulated in severe cases of COVID-19 patients, exhibiting high levels of co-stimulatory and co-inhibitory molecules. This suggested that these cells were in a state of simultaneous hyperactivation and depletion. As a marker of this population, CD38 may be a key regulator of T-cell depletion in COVID-19 patients. Therefore, CD38 is also critical in modulating adaptive immunity during SARS-CoV-2 infection [
95].
CD38 also regulates extracellular adenosine levels. It catalyzes the hydrolysis of NAD+ to cADPR, which acts as a second messenger to regulate Ca
2+ homeostasis, indirectly influencing adenosine metabolism [
96]. In addition, CD38 hydrolyzes extracellular NAD+ to ADPR, which generates AMP and adenosine [
97]. Although the role of adenosine in regulating adaptive immunity in COVID-19 patients is not yet clear, its immunosuppressive effects have been demonstrated in conditions like sepsis. For example, adenosine inhibited macrophage-mediated bacterial killing by hydrolyzing ATP to adenosine [
98]. Furthermore, in Sézary syndrome, a rare cutaneous T-cell lymphoma, T-cell activation induced adenosine production, which regulated T-cell immunosuppression and prevented excessive T-cell responses [
99]. In diseases characterized by inflammation, such as COVID-19, extracellular adenosine may function as a negative immune checkpoint molecule [
49,
100]. Consequently, the elevated levels of CD38 in SARS-CoV-2 infection likely lead to increased adenosine levels, suppressing immune function. Modulating this pathway represents a potential therapeutic target for COVID-19.
3.3. NAD+ Metabolism in Non-Infectious Diseases
PARPs also play a critical role in NAD+-dependent DNA damage repair in non-infectious diseases. In a Drosophila model of Alzheimer's disease (AD), reduced NAD+ levels were observed; however, introducing a RARP mutation increased NAD+ levels and improved mitochondrial function [
68]. Likewise, another study demonstrated that treatment with NR, a NAD+ precursor, increased NAD+ levels in the brain of a mouse model of Alzheimer’s disease. This intervention led to a reduction in DNA release into the cytoplasm, which in turn reduced the aberrant activation of the DNA-sensing pathway. As a result, neuroinflammation levels decreased significantly, suggesting that PARPs could be targeted to enhance NAD+ levels as a potential therapeutic strategy for AD [
69]. Moreover, in the context of BRCA-mutated ovarian cancer, a study found that the combination of DNA damage checkpoint kinase 1 (CHK1) inhibitors and poly (ADP-ribose) glycohydrolase (PARG) inhibitors acted synergistically to increase DNA damage. This, in turn, activated PARP1/2 to reduce NAD+ levels, causing metabolic stress and a decrease in tumor stemness [
70]. The FDA-approved PARP inhibitors, such as olaparib, rucaparib, and niraparib, are applied to treat tumors with BRCA1/2-associated mutations, including ovarian and breast cancers with impaired homologous recombination (HR) repair. These inhibitors regulate DNA repair by recruiting MRE11 and NBS1, enzymes that play a crucial role in homologous recombination [
71,
72].
Apart from PARPs, SIRTs have been implicated in maintaining mitochondrial function and cellular energy balance, especially in non-infectious diseases. For example, in inflammatory bowel disease (IBD), depletion of NAD+ decreased SIRT1 activity and led to increased acetylation of PGC1α, contributing to mitochondrial dysfunction. However, treatment with NR restored the SIRT1-PGC1α axis, leading to improved mitochondrial function in mice with infectious colitis [
73]. Similarly, in age-induced type 2 diabetes (T2D), mice exhibited significantly decreased NAD+ levels across several organs. Treatment with NMN normalized oxidative stress and inflammatory response pathways. Remarkably, SIRT1 deacetylated NF-κB, modulating hepatic insulin sensitivity and improving glucose intolerance and hyperlipidemia [
16]. Furthermore, supplementation with NR increased NAD+ levels in mammalian cells and mouse tissues, thereby improving oxidative metabolism by activating SIRT1 and SIRT3. This helped prevent obesity induced by a high-fat diet [
74]. In a mouse model of nonalcoholic fatty liver disease (NAFLD), SIRT2 regulated deacetylation and deubiquitylation of the fibronectin type III structural domain Fndc5, a process dependent on NAD+. This regulation reversed pathological processes such as steatosis, insulin resistance, mitochondrial dysfunction, and liver fibrosis in NAFLD [
75].
In addition to its role in metabolic disorders, CD38 is also implicated in autoimmune diseases and tumors, primarily through its regulation of signaling pathways and mitochondrial oxidative stress. For instance, in Systemic Lupus Erythematosus (SLE) patients, CD8+ CD38hi T-cell subsets were increased, and NAD+ levels were depleted. CD38 activation led to the acetylation of EZH2, which inhibited SIRT1 activity, ultimately reducing the cytotoxic response [
76]. Similarly, in chronic lymphocytic leukemia (CLL), CD38, a marker of poor prognosis, increased intracellular Ca
2+ concentrations by converting NAD+ to ADPR and cADPR. This activation of chemokine receptors and integrins promoted proliferation and increased the invasiveness of CLL cells. Inhibition of CD38 activity blocked the homing of CLL cells from the bloodstream to lymphoid organs, suggesting that targeting CD38 could be a potential therapeutic strategy to inhibit CLL proliferation [
77]. CD38 also played a crucial role in promoting oxidative stress in multiple myeloma (MM). Overexpression of CD38 in MM led to a generalized depletion of NAD+, triggering mitochondrial metabolic reprogramming and an increase in superoxide anion production, which contributed to increased oxidative stress. Interestingly, CD38 upregulation also enhanced the efficacy of NAD+-depleting agents in treating MM, though the exact mechanism of CD38’s involvement in MM remains unclear [
78]. Additionally, in human non-small cell lung cancer (NSCLC) cell line A549 and HepG2, CD38 overexpression led to significant NAD+ depletion. This depletion activated multiple signaling pathways, including integrin, PI3K/AKT, and ERK/MAPK, promoting epithelial-mesenchymal transition (EMT). Supplementation with NAD+ precursors, however, inhibited STAT3 activity, reversed EMT, and ultimately inhibited tumor cell metastasis [
79]. The alterations of NAD+ metabolism in different non-infectious diseases are detailed in
Table 2.