Russel J. Reiter’s research while affiliated with The University of Texas at San Antonio and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (472)


This figure illustrates the two main apoptotic pathways: the intrinsic (mitochondrial) pathway and the extrinsic (death receptor) pathway. The intrinsic pathway is triggered by cellular stress, such as reactive oxygen species (ROS) and DNA damage, which activate p53, JNK, and pro-apoptotic proteins (BAX, BAD) while inhibiting Bcl-2, leading to mitochondrial membrane damage and cytochrome c (Cyt C) release. Cyt C binds Apaf-1, forming the apoptosome, which activates caspase-9 and executioner caspases (caspase-3, -6, -7), inducing apoptosis. The extrinsic pathway is initiated by the binding of Fas ligand (FasL) to Fas/TNFR, recruiting FADD and activating caspase-8, which directly triggers executioner caspases, leading to apoptos
is. Both pathways ultimately result in controlled cell death through caspase activation
This figure illustrates the molecular mechanisms linking oxidative stress and inflammation to atherosclerosis, as well as the protective effects of melatonin. Reactive oxygen species (ROS) generated by NADPH oxidase (NOX), mitochondria, and uncoupled endothelial nitric oxide synthase (eNOS) contribute to oxidative stress, leading to the activation of inflammatory pathways. Oxidized low-density lipoprotein (ox-LDL) and damage-associated molecular patterns (DAMPs) activate Toll-like receptor 4 (TLR4), which triggers nuclear factor kappa B (NF-κB) signaling. This results in the transcription of pro-inflammatory cytokines, promoting vascular inflammation and endothelial dysfunction. In parallel, oxidative stress and inflammatory stimuli activate the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, which in turn activates caspase-1, leading to the cleavage and release of pro-inflammatory cytokines interleukin-1 beta (IL-1β) and interleukin-18 (IL-18). Additionally, caspase-1 promotes the expression of inducible nitric oxide synthase (iNOS) and adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), and selectins, facilitating immune cell recruitment and plaque progression. Melatonin, a potent antioxidant and anti-inflammatory molecule, inhibits NF-κB activation and NLRP3 inflammasome assembly, thereby reducing cytokine production and endothelial dysfunction. This highlights its potential therapeutic role in mitigating oxidative stress and inflammation in atherosclerosis
Role of melatonin on programmed cell death, inflammation and oxidative stress
Melatonin as a treatment for atherosclerosis: focus on programmed cell death, inflammation and oxidative stress
  • Literature Review
  • Full-text available

April 2025

·

12 Reads

Journal of Cardiothoracic Surgery

Reza Asemi

·

Elham Omidi Najafabadi

·

Zahra Mahmoudian

·

[...]

·

Delaying the development of atherosclerosis (AS) and decreasing cardiac ischemia-reperfusion damage remain serious challenges for the medical community. Chronic arterial disease, i.e., AS, is frequently linked to oxidative stress and inflammation as significant contributing causes. AS risk factors, such as hyperlipidemia, high blood pressure, age, hyperglycemia, smoking, high cholesterol, and irregular sleep patterns, can exacerbate AS in the carotid artery and further shrink its lumen. Finding new approaches that support plaque inhibition or stability is an ongoing problem. The last ten years have shown us that melatonin (MLT) affects the cardiovascular system, although its exact mechanisms of action are yet unknown. MLT’s direct free radical scavenger activity, its indirect antioxidant qualities, and its anti-inflammatory capabilities all contribute to its atheroprotective effects on several pathogenic signaling pathways. Herein, we examine the evidence showing that MLT treatment has significant protective effects against AS and AS-related cardiovascular diseases. The numerous pieces of the puzzle that have been as for epigenetic and biogenetic targets for prevention and therapy against the atherosclerotic pathogenic processes are identified.

Download

A schematic illustration of the autophagy process under stressful settings. Stress signals stimulate autophagy’s early stage via regulating vital molecules like as ULK1, FIP200, ATG13, and ATG101, which are regulated by upstream signaling pathways like AMPK and mTOR. The mechanism starts with the creation of an isolating membrane, also known as a phagophore, which comes from the ER, specifically from specific areas termed omegasomes. The infiltration of central autophagy machinery elements like Beclin1, VPS34, ATG16L, and GABARAP-II allows the phagophore to elongate and mature into an autophagosome. The autophagosome subsequently combines with lysosomes via Rab7 and SNARE proteins to produce an autolysosome. Lysosomal hydrolases breakdown the enclosed cargo within the autolysosome, converting it into simple compounds that the cell may recycle. This strictly controlled mechanism is required to preserve cell equilibrium during stress by eliminating damaged organelles and misfolded proteins
General mechanisms for lncRNA classification. LncRNAs are classified into five major categories depending on their location and transcriptional connection to neighboring mRNAs. (1) Sense lncRNAs are produced from the identical DNA strand as an overlapped protein-coding gene. They may control the expression of the associated mRNA using cis-acting processes like transcriptional interference or chromatin changes; (2) Antisense lncRNAs are produced from the opposite DNA strand of an overlapped mRNA. Antisense transcripts may control the associated mRNA via processes such as RNA-RNA interactions, RNA masking, and transcriptional regulator engagement; (3) Bidirectional lncRNAs are produced from the same promoter area as a protein-coding gene but in the reverse direction. They may affect the transcription of the neighboring genes via promoter competition, transcriptional interference, or enhancer-like actions; (4) Intronic lncRNAs are found fully within the introns of protein-coding genes and can be transcribed either separately or concurrently with the target gene. They may modify host gene activity post-transcriptionally or take part in alternative splicing processes; (5) Intergenic (lincRNAs) lncRNAs are found between protein-coding genes and have no significant overlap. They frequently operate as structural scaffolds, decoys, or enhancers, influencing gene activity via connections to chromosomal modifiers or transcription factors
Several molecular mechanisms of action of LncRNAs. on gene activity and cellular mechanisms. (1) LncRNAs direct chromatin-modifying enzymes to specific genomic loci, affecting chromatin shape and gene expression; (2) LncRNAs act as scaffolds, providing a framework for the construction of ribonucleoprotein complexes that regulate many cellular processes; (3) LncRNAs work as decoys by attaching to and sequestering proteins like transcription factors and other regulatory components, inhibiting their connecting to the desired target DNA or RNA; (4) LncRNAs promote chromatin looping, which brings distant chromosomal areas closer together to control the transcriptional process, which often involves RNA polymerase II; (5) Some lncRNAs operate as progenitors for miRNAs, which are processed to produce functional miRNAs that affect post-transcriptional gene expression; (6) LncRNAs can compete with endogenous RNAs (ceRNAs) by sponging miRNAs, blocking miRNA-mediated repression of targeted mRNAs
The Function of several LncRNAs in various phases of the autophagy formation. LncRNAs like H19, NBR2, and PTENP1 control autophagy initiation via altering autophagy-related proteins, particularly those involved in the AMPK/mTOR signaling cascade. LncRNAs such as AC023115.3 and 10 C146880 affect the activity of the PI3 K complex, which is required for phagophore nucleation. LncRNAs such as TGF2-OT1 and GAS5 are involved in the elongation and closure of autophagosomes. The LncRNA CHAST controls the last stage by modulating ATG5 and PLEKHM1 expression, both of which are required for autophagosome-lysosome fusion
Examples of tumor suppressors and oncogenic lncRNAs that function in leukemia development through autophagy mechanism. Tumor Suppressor LncRNAs (Left Panel): LncRNAs like LINC00265 promote autophagy, which can help to avoid apoptosis and boost cell survival in certain situations. SHLINC00470 can inhibit autophagy to increase tumor growth by activating AKT and, inversely, inhibit autophagy to restore PTEN expression. SHLINC00470 can also destabilize METTL3, which reduces PTEN expression and promotes cancer via m6 A methylation. Oncogenic LncRNAs (right panel): DANCR, UCA1, OIP5-AS1, SNHG5, HOTAIRM1, and LAMPS-AS1 have been found to play oncogenic functions. DANCR interacts with miR- 874 - 3p and ATG16L1 to promote chemoresistance, induce autophagy, and increase chemotherapy effectiveness. UCA1 regulates autophagy through the ATG7 pathway, which contributes cell proliferation and drug resistance. OIP5-AS1 modulates autophagy, increases drug resistance, and overcomes TKT resistance through interactions with miR- 30e- 5p and ATG12. SNHG5 enhances chemoresistance by interacting with miR- 32/DNAJB9. HOTAIRM1 stimulates proliferation, suppresses apoptosis, promotes differentiation, and controls autophagy. LAMPS-AS1 supports survival and self-renewal while inducing autophagy-mediated cell death. PMLRARA is also known to influence autophagy
Interplay between LncRNAs and autophagy-related pathways in leukemia: mechanisms and clinical implications

Medical Oncology

Autophagy is a conserved catabolic process that removes protein clumps and defective organelles, thereby promoting cell equilibrium. Growing data suggest that dysregulation of the autophagic pathway is linked to several cancer hallmarks. Long non-coding RNAs (lncRNAs), which are key parts of gene transcription, are increasingly recognized for their significant roles in various biological processes. Recent studies have uncovered a strong connection between the mutational landscape and altered expression of lncRNAs in the tumor formation and development, including leukemia. Research over the past few years has emphasized the role of lncRNAs as important regulators of autophagy-related gene expression. These RNAs can influence key leukemia characteristics, such as apoptosis, proliferation, epithelial-mesenchymal transition (EMT), migration, and angiogenesis, by modulating autophagy-associated signaling pathways. With altered lncRNA expression observed in leukemia cells and tissues, they hold promise as diagnostic biomarkers and therapeutic targets. The current review focuses on the regulatory function of lncRNAs in autophagy and their involvement in leukemia, potentially uncovering valuable therapeutic targets for leukemia treatment.


Melatonin and its metabolite-induced drop in melanin content in melanotic MNT-1 melanoma cells. After 72 h incubation with melatonin and its metabolites (10–3 M), transmission electron microscopy (TEM) images were obtained as described in Materials and Methods, where the effects of G-coupled membrane receptors (10 μM luzindole or 0.1 μM 4-P-PDOT) were assessed, and their presence did not affect the collapse of melanogenesis. Bars: 1 μm.
Correction: Möller et al. Melanogenesis Is Directly Affected by Metabolites of Melatonin in Human Melanoma Cells. Int. J. Mol. Sci. 2023, 24, 14947

March 2025

·

25 Reads

In the original publication [...]


Melatonin regulation of phase separation in Neuro-PASC: out-maneuvering Janus-faced amyloids

March 2025

·

112 Reads

·

1 Citation

The SAR-CoV-2 virus has evolved to co-exist with human hosts, albeit at a substantial energetic cost resulting in post-infection neurological manifestations [Neuro-post-acute sequelae of SARS-CoV-2 infection (PASC)] that significantly impact public health and economic productivity on a global scale. One of the main molecular mechanisms responsible for the development of Neuro-PASC, in individuals of all ages, is the formation and inadequate proteolysis/clearance of phase-separated amyloid crystalline aggregates—a hallmark feature of aging-related neurodegenerative disorders. Amyloidogenesis during viral infection and persistence is a natural, inevitable, protective defense response that is exacerbated by SARS-CoV-2. Acting as chemical catalyst, SARS-CoV-2 accelerates hydrophobic collapse and the heterogeneous nucleation of amorphous amyloids into stable β-sheet aggregates. The clearance of amyloid aggregates is most effective during slow wave sleep, when high levels of adenosine triphosphate (ATP)—a biphasic modulator of biomolecular condensates—and melatonin are available to solubilize amyloid aggregates for removal. The dysregulation of mitochondrial dynamics by SARS-CoV-2, in particular fusion and fission homeostasis, impairs the proper formation of distinct mitochondrial subpopulations that can remedy challenges created by the diversion of substrates away from oxidative phosphorylation towards glycolysis to support viral replication and maintenance. The subsequent reduction of ATP and inhibition of melatonin synthesis during slow wave sleep results in incomplete brain clearance of amyloid aggregates, leading to the development of neurological manifestations commonly associated with age-related neurodegenerative disorders. Exogenous melatonin not only prevents mitochondrial dysfunction but also elevates ATP production, effectively augmenting the solubilizing effect of the adenosine moiety to ensure the timely, optimal disaggregation and clearance of pathogenic amyloid aggregates in the prevention and attenuation of Neuro-PASC.



Extracellular Signaling Molecules from Adipose-Derived Stem Cells and Ovarian Cancer Cells Induce a Hybrid Epithelial-Mesenchymal Phenotype in a Bidirectional Interaction

March 2025

·

37 Reads

Ovarian cancer (OC) is characterized by high mortality rates due to late diagnosis, recurrence, and metastasis. Here, we show that extracellular signaling molecules secreted by adipose-derived mesenchymal stem cells (ASCs) and OC cells—either in the conditioned medium (CM) or within small extracellular vesicles (sEVs)—modulate cellular responses and drive OC progression. ASC-derived sEVs and CM secretome promoted OC cell colony formation, invasion, and migration while upregulating tumor-associated signaling pathways, including TGFβ/Smad, p38MAPK/ERK1/2, Wnt/β-catenin, and MMP-9. Additionally, OC-derived sEVs and CM induced a pro-tumorigenic phenotype in ASCs, enhancing their invasiveness and expression of tumor-associated factors. Notably, both ASCs and OC cells exhibited increased expression of E-cadherin and Snail/Slug proteins, key markers of epithelial/mesenchymal hybrid phenotype, enhancing cellular plasticity and metastatic potential. We also demonstrated that these cellular features are, at least in part, due to the presence of tumor-supportive molecules such as TNF-α, Tenascin-C, MMP-2, and SDF-1α in the CM secretome of ASCs and OC cells. In silico analyses linked these molecular changes to poor prognostic outcomes in OC patients. These findings highlight the critical role of sEVs and tumor/stem cell-derived secretome in OC progression through bidirectional interactions that impact cellular behavior and phenotypic transitions. We suggest that targeting EV-mediated communication could improve therapeutic strategies and patient outcomes.





Metallothionein rescues doxorubicin cardiomyopathy via mitigation of cuproptosis

February 2025

·

8 Reads

Life Sciences

Doxorubicin (DOX), a chemotherapeutic agent utilized in the management of cancer, provokes cardiotoxicity although effective remedy is lacking. Given that DOX provokes oxidative stress and cell death in cardiomyocytes, this study evaluated the possible involvement of cuproptosis, a newly identified form of cell death, in DOX-instigated cardiac remodeling and contractile dysfunction, alongside the impact of the heavy metal scavenger metallothionein (MT) on DOX cardiomyopathy. Cardiac-specific MT transgenic and wild-type (WT) mice were treated with DOX (5 mg/kg/wk., i.p., for 4 wks) prior to assessment of cardiac morphology and function. DOX evoked cardiac remodeling (interstitial fibrosis), compromised echocardiographic (thinner septum, elevated LVESD, reduced ejection fraction and fractional shortening), cardiomyocyte mechanical and intracellular Ca2+ properties alongside mitochondrial injury (ultrastructure, respiration and mitochondrial membrane potential), oxidative stress (higher O2− and ROS, lower GSH level), apoptosis and cuproptosis (upregulated CTR1 and DLAT, downregulated copper exporter ATP7B, FDX1, the Fesingle bondS cluster proteins ACO2 and NDUFS8). Bioinformatics analysis confirmed enrichment of copper-related genes among DEGs in DOX-challenged hearts. DOX-induced anomalies (except the copper importer CTR1) were reversed by MT overexpression. Co-immunoprecipitation and interaction interface revealed a prominent interaction between MT and ATP7B. Further study indicated that copper chelator tetrathiomolybdate mitigated DOX-induced cardiac remodeling and cardiomyocyte abnormalities whereas copper ionophore elesclomol nullified MT-mediated benefit against DOX. These findings favor a vital role for cuproptosis in DOX cardiomyopathy while the antioxidant MT rescues DOX cardiomyopathy possibly through alleviating cuproptosis.


Citations (69)


... Reiter has long emphasized melatonin's ability to modulate the immune system and its antioxidant capacity, which may help mitigate the cytokine storm-a hyperinflammatory response often seen in severe COVID-19 cases. Given that COVID-19 is associated with increased oxidative stress and inflammation, melatonin's capacity to scavenge free radicals and reduce inflammation could be of significant benefit [16]. Additionally, Reiter's research on melatonin's neuroprotective effects may offer insights into its potential role in mitigating long-term neurological complications in COVID-19 patients that include fatigue, concentration, and memory disturbances. ...

Reference:

Celebrating the legacy of Russel J. Reiter: a pioneer in melatonin research
Melatonin regulation of phase separation in Neuro-PASC: out-maneuvering Janus-faced amyloids

... Notably, at doses of 0.75 mg or higher, the ORR was 41%, PSA50 response reached 59%, and PSA90 response was 45.1%. Adverse events of grade 3 or higher were reported in 66% of patients, with 9% experiencing grade 4 events and 16% discontinuing treatment due to adverse events [70,71]. Amgen is currently advancing Xaluritamig to a phase 3 trial in the second-line mCRPC setting, where the management of CRS and musculoskeletal side effects remains a key consideration [71]. ...

Bispecific antibodies as powerful immunotherapeutic agents for urological cancers: Recent innovations based on preclinical and clinical evidence

International Journal of Biological Sciences

... This accumulation of DNA damage contributes to the initiation and progression of oral squamous cell carcinoma (OSCC). 2,3 In addition to its genotoxic effects, low doses of arecoline promote OSCC cell proliferation by accelerating cell cycle progression and upregulating oncogenes such as c-Myc. 4 This increase in cell proliferation contributes to the rapid expansion of malignant cells in the oral cavity, promoting tumor growth. ...

Correction: Melatonin as an inducer of arecoline and their coordinated roles in anti-oxidative activity and immune responses

Food & Function

... Several studies support the idea that natural MLT secretion through healthy sleep patterns may reduce the risk of AS [34]. A meta-analysis of 71 studies (with 3.8 million participants) found that sleep duration 7.5 h 20:194 per night) was associated with a reduced risk of cardiocerebrovascular disease [35]. ...

Melatonin stabilizes atherosclerotic plaques: an association that should be clinically exploited

... In the former study, the AANAT (arylalkylamine N-acetyl transferase) and ASMT (acetyl serotonin methyltransferase) proteins were identified in mitochondria isolated from mouse brain cells and, moreover, incubating the mitochondria with deuterated serotonin led to the production of deuterated melatonin. AANAT and ASMT are the two enzymes required for the conversion serotonin to melatonin [39,40]. As shown by Venegas and co-workers [15], Suofu and colleagues [38] also found that the brain mitochondrial enzymes do not exhibit a circadian rhythm when collected from mice during exposure to a light:dark cycle as they did in the pineal gland (Fig. 2). ...

Molecular pathways and biological roles of melatonin and vitamin D; effects on immune system and oxidative stress
  • Citing Article
  • November 2024

International Immunopharmacology

... For instance, UCF101, a selective protease inhibitor, has been shown to ameliorate structural and functional cardiac alterations associated with diabetes by regulating AMPK-mediated mitochondrial autophagy. 50 Additionally, studies have demonstrated that inhibiting glycosphingolipids in myocardial cells enhances Sirtuin3 protein expression, scavenges intracellular ROS, and restores mitochondrial autophagy homeostasis, thereby mitigating myocardial hypertrophy. 51 Despite these advances, most research remains foundational, underscoring the need for further clinical investigations. ...

UCF101 Rescues against Diabetes-Evoked Cardiac Remodeling and Contractile Anomalies through AMPK-Mediated Induction of Mitophagy
  • Citing Article
  • October 2024

Pharmacology

... Numerous studies have highlighted the protective effects of Mel in models of oxidative stress, attributed to both its direct scavenging of free radicals and its indirect antioxidant properties (Monteiro et al. 2024). Additionally, Mel has been found to exert anti-tumor effects in several cancers, including breast, prostate, and lung cancers (Reiter et al. 2024). Recent research suggests that Mel enhances the anti-tumor efficacy of Gem in PC by modulating apoptotic pathways (Leja-Szpak et al. 2018) and inhibiting nuclear factor-κB (NF-κB) activation (Ju et al. 2016). ...

Melatonin and vitamin D as potential synergistic adjuvants for cancer therapy

International Journal of Oncology

... In a sepsis mouse model, exposure to LPS triggered cardiomyocyte contractile dysfunction, oxygen molecules accumulation, apoptosis, necroptosis, and ferroptosis. These pathological responses were mitigated by the administration of oleanolic acid, a mitophagy inducer, as well as through the inhibition of ACSL4 and ferroptosis (Li et al., 2024). These findings suggest that cochlear damage induced by the Frontiers in Neuroscience 08 frontiersin.org ...

Ablation of Mitophagy Receptor FUNDC1 Accentuates Septic Cardiomyopathy through ACSL4-Dependent Regulation of Ferroptosis and Mitochondrial Integrity
  • Citing Article
  • September 2024

Free Radical Biology and Medicine

... Due to these reasons, this hormone arises as a multifaceted molecule with significant therapeutic effects to combat cancer. Its property of modulating immune responses and improving cellular resilience reaches the symptoms and pathophysiological pathways associated with cancer [153][154][155][156]. ...

Programmed cell death and melatonin: A comprehensive review

Functional & Integrative Genomics

... Previous studies have demonstrated that impairments in mitophagy, such as too little ROS, result in an accumulation of defective mitochondria in the cell, leading to various diseases [41]. However, conflicting studies have shown that increased ROS production results in deficient mitophagy pathways, also resulting in pathologies such as DPN [42]. Clearly, there is a fine line between too much and not enough ROS within a cell. ...

Investigating the Interplay between Mitophagy and Diabetic Neuropathy: Uncovering the hidden secrets of the disease pathology
  • Citing Article
  • September 2024

Pharmacological Research