Next Article in Journal
Drugs of the Kallikrein–Kinin System: An Overview
Previous Article in Journal
Antimicrobial Nanoformulations Based on Schinus areira Essential Oil
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Black Cumin Seed (Nigella sativa) in Inflammatory Disorders: Therapeutic Potential and Promising Molecular Mechanisms

1
Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza 12585, Egypt
2
Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
*
Author to whom correspondence should be addressed.
Drugs Drug Candidates 2023, 2(2), 516-537; https://doi.org/10.3390/ddc2020027
Submission received: 8 April 2023 / Revised: 25 May 2023 / Accepted: 12 June 2023 / Published: 17 June 2023
(This article belongs to the Section Drug Candidates from Natural Sources)

Abstract

:
Inflammation is an essential defense mechanism against harmful stimuli. However, uncontrolled inflammatory mechanisms culminate in disturbed responses that contribute to multiple serious diseases. Besides common synthetic drugs, there is a growing interest in optimizing the use of natural products as therapeutic or protective supplements against inflammatory disorders. Black cumin seed (BCS), or Nigella sativa (Family Ranunculaceae), is widely used as a health-supportive herb in the Middle East, Far East and West Asia. BCS is a rich source of phytochemicals, and studies have reported its promising effects against a variety of metabolic, proliferative, respiratory, and neurological disorders associated with disrupted inflammatory pathways. This review presents an updated comprehensive assessment of BCS’s effects against various inflammatory disorders and highlights the role of BCS’s bioactive constituents in inflammation and oxidative stress pathways. Moreover, it outlines the future possibilities for enhancing therapeutic activity through efficient pharmaceutical formulations. Thorough analysis of international research studies published between the years 1998 and 2023 reveals the promising anti-inflammatory potential of BCS’s bioactive constituents through modulating inflammation and crucial oxidative stress players in inflammatory disorders. Thus, the bioactive constituents of BCS can be further boosted by updated technologies such as nano-incorporation for the improved management of inflammatory diseases.

1. Introduction

Inflammation is a vital and complicated process in the body used to defend against numerous chemical, biological, and physical stimuli [1]. The concept of inflammation has developed from being described basically as a burning sensation to an intense understanding of its pathology and contributing molecules. Inflammation varies in accordance with different stimuli and their consequent inflammatory responses [2]. Prolonged exposure to inflammation creates a state of chronic inflammation which involves complicated processes of macrophage and cytokine release and the emergence of a high-oxidative-stress status, which in turn can cause irreversible cellular, genetic, and epigenetic disruptions. Such alterations are now established as being highly involved in the pathogenesis of different diseases. Consequently, trying to modify inflammatory status and the implicated inflammatory molecules through anti-inflammatory agents is a therapeutic target that can improve the management of inflammation-related pathologies. This review discusses the inflammation process and the most prominent molecules involved in inflammation. Moreover, it highlights the promising potential of black cumin seed constituents as powerful inflammatory modulators in various disorders. Finally, supporting research for using nano-formulations of black cumin seed as a future approach for optimized therapeutic applications is also included.

2. Methods

The studies included in the current article were obtained from several databases and websites, such as Elsevier, Springer, Wiley online library, Google Scholar, PubMed, MDPI, Science Direct, ResearchGate and Hindawi. All possible data were collected up until 2023. Examples of inflammatory disorders were classified and different synonyms of black cumin seed such as ‘Nigella sativa’, ‘black seed’, ‘black cumin’, ‘thymoquinone’ and ‘nigella oil’ were used. Both in vivo and in vitro studies were included. All studies were peer reviewed and published in the English language or were translated into English.

3. Inflammation: Different Types and the Involved Key Players

3.1. Types of Inflammation

3.1.1. Acute Inflammation

Acute inflammation is a self-limited process that lasts for a few hours to a few days. It is associated with plasma proteins and fluid perfusion (edema) and leukocyte arrival (mainly neutrophils and later macrophages) at the inflammation site. The inflammation process starts with vascular changes, followed by cellular and molecular recruitment [3]. The immune cells control inflammation by producing anti-inflammatory mediators. The recruitment of monocytes, macrophages and dendritic cells critically clears remnant cell debris and antigens [4]. When the immune system eliminates the harmful stimuli, the inflammation ends. However, if these stimuli are not removed successfully, the inflammation turns into a chronic phase [5].

3.1.2. Chronic Inflammation

Uncontrolled inflammation due to extended exposure to stimuli or an improper inflammatory response for a long time, results in chronic inflammation. Inflammation is characterized by the infiltration of lymphocytes and macrophages mononuclear cells, vascular proliferation, fibrosis, and the progression of tissue damage [6]. Persistent inflammation causes extreme pro-inflammatory mediators and cytokine production that can contribute to systemic inflammatory response syndrome (SIRS), sepsis and multiple organ failure. Several studies linked chronic inflammation to several diseases such as type 2 diabetes mellitus [7], arthritis and joint diseases [8], chronic obstructive pulmonary disease (COPD) [9], neurodegenerative diseases [10], multiple sclerosis [11], inflammatory bowel disease [12] and more (Figure 1).
Inflammatory reactions frequently begin with the detection of foreign stimuli by pattern recognition receptors (PRRs) on the cell surface, which then activate several inflammatory pathways. As a result, inflammatory mediators are produced, and inflammatory cells are recruited. The PRRs can identify different molecules such asPAMPs (pathogen-associated molecular patterns) andDAMPs (danger associated molecular patterns). These receptors are expressed on immune cells and other non-immune cells. They contain many classes, such as receptors for advanced glycation end products (RAGE), C-type lectin receptors (CLRs), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) and Toll-like receptors (TLRs). Crucial intracellular signaling pathways are activated through these receptors, followed by inflammatory cytokine and chemokine release [13].

3.2. Signaling Pathways Involved in the Inflammation Process

3.2.1. Mitogen-Activated Protein Kinase (MAPK) Pathway

Many extracellular stimuli can cause the MAPK pathway to be activated, such as growth factors, pathogens, toxins cytokines, and stress signals. The extracellular-signal-regulated kinases ERK1/2 and ERK5, p38 MAP kinases, and c-Jun N-terminal kinases (JNKs) comprise the signaling families of mammalian MAPKs. The MAP kinase pathways have been implicated in a wide range of pathological illnesses, including cancer and other diseases [14,15].

3.2.2. Nuclear Factor Kappa B (NF-κB) Pathway

The NF-κB pathway plays a crucial role in the regulation of immunity, inflammation, and cell survival. Five proteins, namely p105/p50 (NF-B1), p100/52 (NF-B2), p65 (RelA), RelB and c-Rel, compensate the NF-B transcription factors in mammals. NF-κB activation occurs through at least two different pathways; T- and B-cell receptors, pathogen-related molecules, and pro-inflammatory cytokines such as TNF-α and IL-1 stimulate the “canonical” pathway, while the tumor necrosis factor (TNF)-family cytokines (LTβ, CD40L, BAFF, and RANKL except for TNF-α) activate the “alternative” NF-κB pathway. The noncanonical NF-B regulates the adaptive immune system concomitantly with the canonical NF-B pathway, whereas canonical NF-B plays a major role in immune responses. NF-κB directly regulates inflammation by increasing the production of inflammatory cytokines, chemokines, and adhesion molecules [16,17].

3.2.3. The Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) Pathway

The JAK-STAT pathway is another important pathway that regulates many cellular events. Cytokines are a major activator of this pathway. STAT is phosphorylated by JAK and moves to the nucleus to influence specific gene expressions [18]. All STATs except for STAT4 localize to the mitochondrion and enhance oxidative phosphorylation and membrane permeability, while STAT3 can move to the endoplasmic reticulum and resists excessive apoptosis induced by oxidative stress. Dysregulation of JAK-STAT in humans can contribute to diseases such as cancer and autoimmune diseases [19,20].

3.3. Key Inflammatory Molecules

-
Tumor necrosis factor alpha (TNF-α): is a potent pro-inflammatory cytokine that is crucial for the immune system’s function during inflammation, cell proliferation, differentiation, and apoptosis. TNF-alpha exerts its effects through TNF-alpha receptor I, expressed almost in all kinds of cells, while TNF-alpha receptor II is only expressed in immune system cells, fibroblasts and endothelial cells [21].
-
Nitric oxide (NO): is involved in physiological processes such as neurotransmission, vasodilation, platelets aggregation and adhesion, host defense, and immune regulation. In pathological conditions, NO acts as a cytotoxic agent especially in inflammatory diseases. Upon immunological stimulation, NF-κB stimulates inducible nitric oxide synthase (iNOS, also called NOS2), which synthesizes NO. Overproduction of NO causes high oxidative stress and cell death [22].
-
Prostaglandin E2 (PGE2): The cell membrane’s phospholipids and phospholipase A2 (PLA2) release arachidonic acid (AA). AA is the main precursor for prostanoids which are converted into Prostaglandin H2 (PGH2) by cyclooxygenase (COX) and peroxidase. PGH2 is then transformed by Prostaglandin E (PGE) synthase into Prostaglandin E2 (PGE2) [23]. PGE2 acts as a vasodilator to promote the migration of white blood cells to the site of inflammation causing edema. Additionally, it increases the pain response and acts as a fever mediator [24,25]. Therefore, the inhibition of cyclooxygenase enzyme suppresses prostanoid synthesis and decreases vasodilatation, vascular permeability, and the recruitment of immune cells in inflammation.
-
Interleukin-1β (IL-1β), as a member of the Interleukin 1 (IL-1) family, is a pivotal pro-inflammatory mediator. It is usually linked to acute and chronic inflammation, where the levels of NOS2, COX-2, adhesion molecules, IL-6 and TNF-α are elevated with IL-1β overexpression [26].
-
Interleukin-6 (IL-6): is a main pro-inflammatory member of the IL-6 family. It is involved in acute and chronic inflammation. NF-κB and activator protein 1 (AP-1) are important transcription factors for IL-6. IL-6 is capable of inducing differentiation in T-helper cells and B cells. Elevated IL-6 levels were observed in autoimmune diseases, inflammatory diseases and cancer [27].
-
Interleukin-17 (IL-17): is produced by a subset of T-helper cells known as Th17 cells as a primary source as well as other immune cells including natural killer T cells, gamma-delta T cells, microglia, mast cells, neutrophils, and others. IL-17 is involved in host defense mechanisms through releasing antimicrobial peptides (AMPs), chemokines, and proinflammatory cytokines. On the other hand, it may be implicated in the pathogenesis of autoimmune disorders [28].

3.4. Chronic Inflammation and Oxidative Stress: A Vicious Cycle

Chronic inflammation triggers a status of oxidative stress leading to the excessive release of reactive oxygen species (ROS), which opposes the body’s antioxidant defense mechanisms and causes cellular damage. In turn, excessive oxidative stress caused by reactive species triggers an inflammatory response, which produces more free radicals, which can lead to further oxidative stress and chronic inflammation, creating a vicious cycle (Figure 1). Chronic inflammation due to oxidative stress has been shown to be involved in several conditions, including cancer, diabetes, cardiovascular disease, arthritis, and neurodegenerative disorders [29].

4. Natural Herbs as Anti-Inflammatory Agents

Natural herbs have been used for centuries in folk medicine for the treatment of various diseases, including inflammatory disorders. Many of these herbs have been shown to possess anti-inflammatory properties with few side effects compared with synthetic drugs. Scientific research is ongoing to further explore their potential health benefits. Among the promising constituents in natural herbs with anti-inflammatory activity are resveratrol in grapes [30], quercetin in citrus fruits [31], curcumin in turmeric [32], epigallo-catechin-3-gallate in green tea [33], gingerol in ginger [34], allicin in garlic [35], 1,8-cineole in eucalyptus [36] leaves and thymoquinone in black cumin seed [37]. Black cumin seed has been extensively studied for its potential health benefits. In this review, we highlighted the updated studies of black cumin seed in inflammatory disorders and its different pharmacological formulations.

4.1. Black Cumin Seed

Black cumin seed, or Nigella sativa Linneaus (N. sativa), is an annual flowering herb belonging to the Ranunculaceae family. It is widely distributed in Middle Eastern and Southeast Asian countries [38]. Black cumin seed (BCS) and its oil are used in many therapeutic and cosmetic applications. Traditionally, they are used as a spice, appetizer and food additive, a source of energy and an immune stimulant, a carminative and bowel movement regulator, as well as an analgesic and antipyretic supplement. Additionally, they are used in many respiratory conditions such as the common cold, coughing and asthma. BCS is used to treat dyslipidemia, obesity, diabetes and hypertension as well [39,40]. Women use BCS to relieve menstrual discomfort and as a galactagogue herb [40]. They also use it for skin and hair beauty purposes [41]. Moreover, BCS is used to support the general health of the neurons, stomach, intestines, kidneys, and liver. N. sativa has gained attention as a potential therapeutic agent due to its diverse pharmacological properties [42].

4.2. Chemical Constituents

Many compounds have been identified and quantified in black cumin seed by using different techniques such as high-performance liquid chromatography (HPLC) [43], gas chromatography [44] and thin-layer chromatography (TLC) [45]. BCS is a rich source of carbohydrates (24.9%), fats (28.5%), of which the essential (volatile) oils constitute about 0.4–2.5%, while the fixed oil, which is rich in saturated and unsaturated fatty acids, constitutes about 26–34%, proteins (26.7%), fibers (8.4%), ash (4.8%), vitamins (1.8–3.7%), minerals (3.7–7%) and water [38]. Thymoquinone (TQ) is a putative phytochemical compound of the BCS (Figure 2). It constitutes around 30–48% of the volatile oil. Table 1 demonstrates different compounds that have been identified in BCS.

4.3. Safety and Toxicological Profile of Black Cumin Seed

Studies that evaluated the toxicity of BCS typically described it generally as a safe therapeutic plant as it has a broad safe margin [49,50,51]. In animal models, TQ exerted a median lethal dose (LD50) of 2.4 g/kg in acute oral administration in mice, while in sub-chronic administration, a cytoprotective effect was revealed in mice that received 0.01, 0.02, and 0.03% of TQ in their drinking water, with no signs of toxicity or morbidity [52]. Oral administration of BCS oil for 3 weeks at 2 mL/kg showed a hepatic and renal-protective effect in rats [53]. Different forms of BCS showed LD50 values of 3020, 3371, and 1853 mg/kg for aqueous extract, fixed oil and volatile oil, respectively, in mice [54]. BCS extracts in water, methanol, and chloroform at oral doses of 6, 9, 14, and 21 g/kg did not cause substantial morbidity or mice death [55]. BCS powder integrated in rats pellets at doses of 0.01, 0.1, and 1 g/kg administered daily for 28 days did not cause any toxicity or alteration in liver function markers [56]. BCS fixed oil had LD50 values of 28.8 mL/kg orally and 2.06 mL/kg intraperitoneally in mice after being delivered in single doses [51]. Rats tested for the chronic toxicity of BCS fixed oil (2 mL/kg orally for 12 weeks) showed no histological changes in the kidneys, liver, heart or pancreas or any alteration in liver enzymes as alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyl transferase (GGT). Moreover, a significant elevation in hemoglobin and hematocrit values was observed along with a marked decrease in glucose, serum cholesterol, triglyceride, platelet and leukocyte values [51]. However, BCS oil doses should be carefully adjusted, as some histological changes were observed in rats’ kidneys and livers after one month of administering oral BCS oil at doses of 15 and 25 mL/kg [57].
In humans, the clinical utilization of BCS has also provided evidence of its safety. For example, BCS oil administration for 8 weeks reduced blood pressure and showed no adverse effects on healthy candidates [58] or in hypertensive patients [59]. BCS and TQ were both well tolerated and showed no side effects considering the management of dyslipidemia [60]. Despite the lack of information regarding the maximum tolerable dose of TQ, it was reported that adult patients with solid tumors tolerated TQ oral doses of up to 2600 mg/day [61]. BCS oil was well tolerated by both adults and children when given at doses of 40 mg/kg. However, children who received high doses of BCS oil (80 mg/kg) experienced gastrointestinal side effects. Therefore, BCS oil is recommended to be used in children at weight-appropriate doses and given after meals [62].

4.4. Therapeutic Activity of Black Cumin Seed in Inflammatory Disorders

The health benefits of BCS have been largely attributed to its powerful anti-inflammatory-antioxidant properties (Figure 3). A summary of updated studies that reported the efficiency of using BCS constituents in in-vivo and in-vitro models for a variety of inflammation-related disorders is represented in Table 2.

4.4.1. Rheumatic Arthritis (RA)

Rheumatic arthritis is a chronic autoimmune disease that affects the joints and other parts of the body. It causes inflammation and swelling in the joints, leading to pain, stiffness, and difficulty in movement [81]. Over time, RA can cause significant joint damage, leading to disability. In 2019, a study performed by Cinzia Nasuti et al., on the healing impact of BCS oil (at oral doses of 0.91 mL/kg and 1.82 mL/kg) on arthritic rats showed a marked inhibition of the arthritis score, the percentage inhibition of paw edema and disease progression. In addition, inflammatory markers such as C-reactive protein (CRP) and interleukin 6 were significantly lowered [82]. A clinical study that recruited 40 rheumatic arthritic females revealed the significant therapeutic activity of BCS oil (500 mg of BCS oil capsules twice/day for a month after a month of placebo intake), which was reflected by the improved activity score in swollen joints, pain and morning stiffness [83]. Another study for the effect of BCS oil (500 mg capsules twice a day for 8 weeks) on 23 rheumatic arthritic female patients showed that the disease activity score was significantly lower in the BCS oil group, with a remarkable decrease in serum nitric oxide (NO) and malondialdehyde (MDA) relative to the baseline and elevated protective IL-10 levels [84]. Moreover, one gram of BCS oil given orally to 23 rheumatic arthritic females for 8 weeks caused a significant decrease in CRP levels and CD8+, together with the alleviation of disease activity scores of 28 joints compared with baseline and placebo groups [85]. In addition, pre-treatment of rheumatoid arthritis synovial fibroblast cells with TQ inhibited the production of IL-6, IL-8, and the adhesion molecules: Intercellular Adhesion Molecule 1 (ICAM-1), Vascular Cell Adhesion Molecule 1 (VCAM-1) and Human Cadherin-11 (Cad-11). TQ is considered a potential therapy for RA by inhibiting apoptosis-regulated signaling kinase 1 (ASK1) and the consequent activation of p38 and JNK [86]. The histological indices of joint inflammation and paw weight were both significantly lowered by TQ in pristane induced arthritis in rats [87].

4.4.2. Inflammatory Bowel Disease and Ulcerative Colitis

Ulcerative colitis is a chronic idiopathic disease that affects the large intestine and rectum, causing inflammation and ulcers in the inner lining area, leading to inconvenient symptoms such as abdominal pain, diarrhea, and rectal bleeding [88]. Thymoquinone at a rectal dose of 100 mg/kg was found to reduce fecal inflammatory bowel disease (IBD) biomarkers such as calprotectin, hydroxyproline, and lactoferrin, as well as oxidative stress index (OSI) and total oxidant status (TOS) levels in rats treated with dextran sulfate sodium (DSS) [89]. In line with these findings, TQ was found to have powerful anti-inflammatory activity in HT-29 cells and an ulcerative colitis mouse model at doses of 10 and 20 μM and 20 and 40 mg/kg, respectively. It reduced the gene expression and protein levels of IL-1β, IL-6, TNF-α, COX-2, and iNOS and decreased the myeloperoxidase (MPO) activity. Furthermore, it regulated the phosphorylation of NF-κB and MAPK proteins. Likewise, TQ reduced IL-8 and C-X-C Motif Chemokine Ligand 1(CXCL-1) levels and increased the expression and protein levels of peroxisome proliferator- activated receptor gamma (PPAR-γ) modulator, while it also enhanced the disease activity index (DAI) score and altered the histopathological findings caused by DSS [90]. Another study revealed that more favorable clinical illness scores, macroscopic assessment, and weight gain were noticed in rats treated with 2 mL/kg BCS oil, with a lower extent of bowel damage, apoptosis, and histopathologic findings. Additionally, significantly lower MDA and MPO levels with high activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx) were observed [91].

4.4.3. Neurodegenerative Disorders

Alzheimer’s Disease (AD)

Alzheimer’s disease is the most common dementia-causing factor, characterized by the gradual loss of neurons and synapses in the brain [92]. Several factors are thought to contribute to its development, including genetics, lifestyle, and age [93]. Azar Hosseini et al., showed that BCS oil exerted an anti-inflammatory effect in LPS-induced microglial neuroinflammation. BCS oil markedly decreased IL-1β, IL-6, TNF-α, PGE2, and iNOS levels while elevating IL-10 and arginase-1 (Arg1) levels [94]. Moreover, rats treated orally with 3 mL/Kg BCS oil for 6 weeks showed decreased inflammatory and oxidative stress markers by regulating the levels of MDA, caspase 3, TNF-α, acetylcholinesterase (AChE), GPx, catalase (CAT), and SOD, along with improved histopathological changes in the brain such as congestion, hemorrhage, degeneration, and edema in emamectin benzoate-induced neurotoxicity [95]. In the same context, the anti-oxidant activity of the hydro-alcoholic extract of BCS was suggested to improve learning and memory capabilities in neonatal and juvenile rats [96]. BSC oil also improved behavioral test assessments when administered at a dose of 2 mL/kg for 4 weeks in mercuric chloride-induced neurotoxicity when used alone or combined with ginger [97]. Moreover, hydro-alcoholic extract treatment of BCS resulted in better memory performance and decreased levels of AChE activity and MDA in scopolamine-treated rats [98]. In another study, BCS oil used in combination with virgin olive oil and an aqueous extract of roasted date seeds revealed high prophylactic and therapeutic efficiency against CuSO4-induced AD [99]. Meanwhile, a low dose of BCS (500 mg per day for four weeks) was sufficient to reduce anxiety, maintain mood, and positively enhance cognition in healthy participants compared to a placebo [100]. Thymoquinone administration for 2 weeks at different doses was recently found to counteract cognitive impairment and morphological abnormalities in D-Gal/aluminum chloride-induced AD in rats by decreasing the levels of pro-inflammatory cytokines such as TNF-α and IL1-β and the mRNA expression of NF-κB, TLR-2, TLR-4, MyD88, TRIF and IRF-3, while reducing amyloid-β formation and accumulation in rat brain striata [101]. Meanwhile, Abulfadl et al., reported that TQ administration to treat D-gal- and AlCl3-induced neurotoxicity significantly enhanced the levels of SOD, total antioxidant capacity (TAC), brain-derived neurotrophic factor (BDNF), B-cell lymphoma 2 protein (BCL-2) and acetylcholine (Ach) immunoreactivity and lowered the levels of AChE activity, MDA, NO as well as TNF-α immunoreactivity [102]. TQ was also found to inhibit IκB and p65 subunits in an LPS-induced microglia inflammation model, and thus inhibit NF-κB activation. Additionally, its anti-inflammatory activity was confirmed via the activation of Nrf2/ARE signaling pathway [103].

Parkinson’s Disease (PD)

Parkinson’s disease is a progressive neurodegenerative disorder that affects movement, muscle control, and balance [104]. Mice orally treated with 200 mg/kg of hydroalcoholic BCS for 12 days exhibited improved muscle stiffness in a Parkinson’s model [105]. Furthermore, the ethanolic extract of BCS significantly reduced the decline in climbing capacity and lengthened life in a PD genetic model of Drosophila in a dose-dependent manner [106]. Additionally, BCS ethanolic extract was significantly able to increase reduced Glutathione (GSH) and total protein levels and reduce the levels of thiobarbituric acid reactive substances (TBARS) and nitrites in rats treated with chlorpromazine [107]. The oral administration of 0.2 mL of BCS oil for 6 weeks exerted a marked neuroprotective effect in extrapyramidal symptoms (EPS) such as behavior in rats receiving haloperidol, improving the motor functions and enhancing the histopathological findings with the decreased alteration of microglia and astrocyte activities [108].

4.4.4. Respiratory Diseases

Chronic Obstructive Pulmonary Disease (COPD)

Chronic obstructive pulmonary disease is a progressive inflammatory lung disease that causes difficulty in breathing, coughing, and wheezing. A major cause of COPD is the persistent inflammation brought on by long-term exposure to harmful particles or gases such as cigarette smoke [109]. A clinical study compared COPD patients that were divided into a control group, who received medications only, and the experimental group, who received medications and encapsulated BCS oil for three weeks. The BCS oil group showed a significant decrease in TBARS, protein carbonyl (PC), IL-6 and TNF-α, while a significant enhancement was observed in pulmonary function tests, vitamins levels such as vitamin C and E, and antioxidant markers such as GSH, GPx, CAT, SOD [110]. Furthermore, the chemotaxis of neutrophils triggered by N-Formylmethionyl-leucyl-phenylalanine (fMLP) and the migration of random cells was found to be inhibited by an essential oil from BCS in a dose-dependent manner, in addition to its effect on human neutrophil elastase (HNE) secretion [111].

Asthma

Oral administration of BCS oil capsules for four weeks as an adjuvant therapy led to an improvement in the asthma control test (ACT) score and pulmonary function tests along with a marked decrease in blood eosinophil count compared to the placebo group [112]. Moreover, BCS oil administration for eight weeks orally as an adjunctive therapy at a dosage of 15–30 mg/kg/day in asthmatic children revealed an enhancement in IFN-γ/IL-4 levels with no significant differences in the Th1 and Th2 cell number or the Th1/Th2 ratio when compared to control [113]. Meanwhile, Koshak and colleagues showed that BCS suppressed the release of pro-inflammatory mediators (IL-2, IL-6, PGE2) in human T-lymphocytes, monocytes, and A549 human lung epithelial cells [114]. Moreover, BCS ethanolic extract reduced the amounts of histamine in rat peritoneal mast cells and exhibited no toxicity to mast cells [115].

Coronavirus Disease 2019 (COVID-19)

COVID-19 emerged as a pandemic in early 2020 [116]. It was found to cause a massive disturbance in the immune system and correlate with NF-κB, IRF, and MAPK pathway activation, ending with an elevation of different proinflammatory cytokines and chemokines [117]. Black cumin seed was suggested as a supplement or treatment for COVID-19 due to its known immunomodulatory and antiviral properties [118]. BCS was hypothesized to interfere with SARS-CoV-2 receptors [119]. Using the molecular docking technique, BCS’s active constituents, namely β-sitosterol, α-hederin, nigellidine-4-O-sulfite, sterol-3-D-glucoside, nigellidine, and dithymoquinone, were reported to have binding affinity to SARS-CoV-2 Mpro [120]. Clinically, BCS was considered as a promising prophylactic herb against COVID-19 incidence [121]. Additionally, it decreased disease severity significantly when administrated with the standard COVID-19 treatment [122]. BCS oil was also shown to improve the symptoms of COVID-19 patients [123]. BCS combined with honey remedies showed accelerated viral load removal, ameliorated disease symptoms and a lower number of deaths compared to the placebo group [124]. However, there is a lack of studies that measured biochemical markers such as pro-inflammatory mediators in clinical and animal models.

4.4.5. Metabolic Disorders

Metabolic disorders are a group of medical conditions that affect the metabolism of the body, leading to health problems such as obesity, diabetes, and cardiovascular disease. Many studies have shown that chronic inflammation plays a critical role in the development and progression of metabolic disorders, and vice versa [125].

Obesity, Diabetes Mellitus (DM) and Cardiovascular Disease (CVDs)

Essential and fixed oil of BCS was found to minimize free radicals’ release and boost the antioxidant capacity and immunity in streptozotocin-induced DM rats [126]. A clinical study on obese females supplied with BCS for 8 weeks (2 g/day) together with an aerobic training program exhibited a significant decrease in the lipid profile, total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C) and body mass index, with a significant increase in high-density lipoprotein cholesterol (HDL-C) levels and maximal aerobic capacity (max VO2) [127]. Another study on obese females who received 3 g of BCS oil for 8 weeks observed a marked reduction in their weight and waist circumference, in addition to a decline in triglycerides and VLDL compared to the placebo group [128]. Indeed, a pilot study on healthy male candidates who received 1 g/day of BCS powder for 4 weeks showed no significant changes in serum lipids or in glucose regulation, while this treatment also did not change insulin sensitivity. However, a significant correlation between the change in total cholesterol, LDL-C and their baseline levels was observed in the BCS group only [129].

Non-Alcoholic Fatty Liver Disease (NAFLD)

NAFLD is a condition where there is an excessive accumulation of fat in the liver, which is not caused by alcohol consumption. The accumulation of fat in the liver can trigger an inflammatory response, leading to the recruitment of immune cells to the liver, which can cause progressive damage [130]. NAFLD patients who received BCS oil for 3 months showed a significant decrease in liver ALT and AST enzymes as well as LDL-C and triglycerides [131]. Another clinical study used 2 g of BCS for 12 weeks as a supplement in NAFLD patients, showing a marked decline in glucose and insulin levels and a remarkable rise in quantitative insulin sensitivity check index [132]. Meanwhile, using 1 g of BCS capsules twice daily for 12 weeks revealed remarkable reductions in ALT and AST and the body mass index, with 57.14% of patients reporting normal ultrasound grades when compared to the placebo group [133]. Additionally, an NAFLD patient group who ingested 2 g of BCS per day for 12 weeks showed a better reduction in inflammatory markers such as NF-κB, high-sensitivity C-reactive protein (hs-CRP), TNF-α as well as hepatic steatosis percentage compared to the placebo patient group [134]. Another clinical study by Rashidmayvan and colleagues revealed that BCS oil consumption at 1000 mg/day for 8 weeks decreased fasting blood sugar levels, lipid profile (TC, TG, VLDL-C, LDL-C), liver enzymes (AST and ALT), hs-CRP inflammatory markers, TNF-α and IL-6, while it increased HDL-C levels. BCS oil showed no significant effect on insulin levels, blood pressure, and GGT in comparison to the placebo group [135].

4.4.6. Anticancer Activity

Thymoquinone, as a major constituent of BCS, has shown promising antineoplastic activity against a wide variety of cancers [136], where TQ is involved in multiple biological pathways that are critically involved in proliferation, cell migration and cell cycle regulation. Emerging studies are continuously highlighting the distinguished potential of TQ against pancreatic cancer, where it significantly enhances apoptosis and decreases metastasis through modulating the NF-κB, tumor growth factor-B (TGF-β) and MAPK signaling pathways [137]. Moreover, TQ remarkably increases chemosensitivity in pancreatic cancers resistant to therapy by modulating matrix metalloproteinase enzyme (MMP-1) and NF-κB levels [138]. Table 3 presents updated studies that reported the efficiency of using BCS constituents in various types of cancer.

4.5. Updated and Future Approach: Nano-Formulation for Optimized Therapeutic Applications of Black Cumin Seed

BCS oil represents a natural source of rich anti-inflammatory capacity. A big challenge for the wider use of BCS and particularly TQ is the low water solubility, photosensitivity, and poor bioavailability. These drawbacks have been thoroughly tackled by the current and progressive advances of nano-formulations, leading to remarkable and continuous improvements in BCS bioavailability and stability. Table 4 illustrates some of the nano-formulations used for incorporating BCS in a variety of therapeutic applications.

5. Conclusions

Inflammatory disorders such as rheumatoid arthritis, inflammatory bowel disease, ulcerative colitis, asthma, diabetes, and neurodegenerative diseases have received increased interest due to their adverse consequences. In this review, we briefly discussed the possible health benefits of various forms of black cumin seed (BCS) and thymoquinone (TQ) in these conditions. Their potential therapeutic effects were attributed to their anti-inflammatory and antioxidant activities. BCS modulates pro-inflammatory molecules such as IL-1β, IL-2, IL-6, IL-12, IL-8 TNF-α, COX-2, iNOS, PGE2, MDA, and MPO, anti-oxidant enzymes such as SOD, CAT, GSH, GPx, and AChE, as well as adhesive molecules such as ICAM-1, VCAM-1, and Cad-11. BCS regulates the blood pressure, lipid profile and blood glucose levels in a favorable manner. The anticancer activities of TQ were proven in different types of cancers. However, more studies are required to investigate the role of black cumin seed constituents other than TQ. Combinations of BCS with other remedies have exerted more beneficial effects. Finally, recent studies have shown that BCS and TQ nano-formulations are important approaches to enhance the outcomes of their biocompatibility and efficiency.

Author Contributions

H.K.R.; writing—original draft preparation, S.M. and N.Z.A.; writing—review and editing, H.K.A.; supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this review are available in the references cited in the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Medzhitov, R. Origin and Physiological Roles of Inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  2. Abdulkhaleq, L.A.; Assi, M.A.; Abdullah, R.; Zamri-Saad, M.; Taufiq-Yap, Y.H.; Hezmee, M.N.M. The Crucial Roles of Inflammatory Mediators in Inflammation: A Review. Vet. World 2018, 11, 627–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Janeway, C.A.J.; Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Weisheit, C.K.; Engel, D.R.; Kurts, C. Dendritic Cells and Macrophages: Sentinels in the Kidney. Clin. J. Am. Soc. Nephrol. 2015, 10, 1841–1851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Ambriz-Pérez, D.L.; Leyva-López, N.; Gutierrez-Grijalva, E.P.; Heredia, J.B. Phenolic Compounds: Natural Alternative in Inflammation Treatment. A Review. Cogent. Food Agric. 2016, 2, 1131412. [Google Scholar]
  6. Pahwa, R.; Goyal, A.; Jialal, I. Chronic Inflammation; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK493173/ (accessed on 21 March 2023).
  7. Tsalamandris, S.; Antonopoulos, A.S.; Oikonomou, E.; Papamikroulis, G.-A.; Vogiatzi, G.; Papaioannou, S.; Deftereos, S.; Tousoulis, D. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur. Cardiol. Rev. 2019, 14, 50–59. [Google Scholar] [CrossRef] [Green Version]
  8. Mititelu, R.R.; Pădureanu, R.; Băcănoiu, M.; Pădureanu, V.; Docea, A.O.; Calina, D.; Barbulescu, A.L.; Buga, A.M. Inflammatory and Oxidative Stress Markers—Mirror Tools in Rheumatoid Arthritis. Biomedicines 2020, 8, 125. [Google Scholar] [CrossRef]
  9. Butler, A.; Walton, G.M.; Sapey, E. Neutrophilic Inflammation in the Pathogenesis of Chronic Obstructive Pulmonary Disease. COPD J. Chronic Obstr. Pulm. Dis. 2018, 15, 392–404. [Google Scholar] [CrossRef]
  10. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS Neurodegenerative Diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [Green Version]
  11. Faissner, S.; Plemel, J.R.; Gold, R.; Yong, V.W. Progressive Multiple Sclerosis: From Pathophysiology to Therapeutic Strategies. Nat. Rev. Drug Discov. 2019, 18, 905–922. [Google Scholar] [CrossRef]
  12. Desai, D.; Faubion, W.A.; Sandborn, W.J. Review Article: Biological Activity Markers in Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2006, 25, 247–255. [Google Scholar] [CrossRef]
  13. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory Responses and Inflammation-Associated Diseases in Organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef] [Green Version]
  14. Bohush, A.; Niewiadomska, G.; Filipek, A. Role of Mitogen Activated Protein Kinase Signaling in Parkinson’s Disease. Int. J. Mol. Sci. 2018, 19, 2973. [Google Scholar] [CrossRef] [Green Version]
  15. Zhang, W.; Liu, H.T. MAPK Signal Pathways in the Regulation of Cell Proliferation in Mammalian Cells. Cell Res. 2002, 12, 9–18. [Google Scholar] [CrossRef]
  16. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-ΚB Signaling in Inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
  17. Oeckinghaus, A.; Ghosh, S. The NF-ΚB Family of Transcription Factors and Its Regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  18. Dutta, P.; Li, W.X. Role of the JAK-STAT signalling pathway in cancer. In Encyclopedia of Life Sciences; Wiley: Hoboken, NJ, USA, 2013. [Google Scholar]
  19. Seif, F.; Khoshmirsafa, M.; Aazami, H.; Mohsenzadegan, M.; Sedighi, G.; Bahar, M. The Role of JAK-STAT Signaling Pathway and Its Regulators in the Fate of T Helper Cells. Cell Commun. Signal. 2017, 15, 23. [Google Scholar] [CrossRef] [Green Version]
  20. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT Signaling Pathway: From Bench to Clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  21. Sethi, G.; Sung, B.; Aggarwal, B.B. TNF: A Master Switch for Inflammation to Cancer. Front. Biosci. Landmark 2008, 13, 5094–5107. [Google Scholar] [CrossRef] [Green Version]
  22. Aktan, F. INOS-Mediated Nitric Oxide Production and Its Regulation. Life Sci. 2004, 75, 639–653. [Google Scholar] [CrossRef]
  23. Saper, C.B.; Romanovsky, A.A.; Scammell, T.E. Neural Circuitry Engaged by Prostaglandins during the Sickness Syndrome. Nat. Neurosci. 2012, 15, 1088–1095. [Google Scholar] [CrossRef] [PubMed]
  24. Blomqvist, A.; Engblom, D. Neural Mechanisms of Inflammation-Induced Fever. Neuroscientist 2018, 24, 381–399. [Google Scholar] [CrossRef] [PubMed]
  25. Nakanishi, M.; Rosenberg, D.W. Multifaceted Roles of PGE2 in Inflammation and Cancer. Semin. Immunopathol. 2013, 35, 123–137. [Google Scholar] [CrossRef] [PubMed]
  26. Dinarello, C.A. Interleukin-1β. Crit. Care Med. 2005, 33, S460–S462. [Google Scholar] [CrossRef] [PubMed]
  27. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in Inflammation, Immunity and Disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef]
  28. Ge, Y.; Huang, M.; Yao, Y. Biology of Interleukin-17 and Its Pathophysiological Significance in Sepsis. Front. Immunol. 2020, 11, 1558. [Google Scholar] [CrossRef]
  29. Sharifi-Rad, M.; Anil Kumar, N.V.; Zucca, P.; Varoni, E.M.; Dini, L.; Panzarini, E.; Rajkovic, J.; Tsouh Fokou, P.V.; Azzini, E.; Peluso, I.; et al. Lifestyle, Oxidative Stress and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front. Physiol. 2020, 11, 694. [Google Scholar] [CrossRef]
  30. Bahar, B.; Singhrao, S.K. An Evaluation of the Molecular Mode of Action of Trans-Resveratrol in the Porphyromonas gingivalis Lipopolysaccharide Challenged Neuronal Cell Model. Mol. Biol. Rep. 2021, 48, 147–156. [Google Scholar] [CrossRef]
  31. Wei, X.; Meng, X.; Yuan, Y.; Shen, F.; Li, C.; Yang, J. Quercetin Exerts Cardiovascular Protective Effects in LPS-Induced Dysfunction in vivo by Regulating Inflammatory Cytokine Expression, NF-ΚB Phosphorylation and Caspase Activity. Mol. Cell. Biochem. 2018, 446, 43–52. [Google Scholar] [CrossRef]
  32. Yu, Y.; Shen, Q.; Lai, Y.; Park, S.Y.; Ou, X.; Lin, D.; Jin, M.; Zhang, W. Anti-Inflammatory Effects of Curcumin in Microglial Cells. Front. Pharmacol. 2018, 9, 386. [Google Scholar] [CrossRef] [Green Version]
  33. Fu, M.; Fu, S.; Ni, S.; Zou, L.; Liu, Y.; Hong, T. Anti-Inflammatory Effect of Epigallocatechin Gallate in a Mouse Model of Ovalbumin-Induced Allergic Rhinitis. Int. Immunopharmacol. 2017, 49, 102–108. [Google Scholar] [CrossRef]
  34. Zahoor, A.; Yang, C.; Yang, Y.; Guo, Y.; Zhang, T.; Jiang, K.; Guo, S.; Deng, G. 6-Gingerol Exerts Anti-Inflammatory Effects and Protective Properties on LTA-Induced Mastitis. Phytomedicine 2020, 76, 153248. [Google Scholar] [CrossRef]
  35. Mousa, A.M.; Soliman, K.E.A.; Alhumaydhi, F.A.; Almatroudi, A.; Allemailem, K.S.; Alsahli, M.A.; Alrumaihi, F.; Aljasir, M.; Alwashmi, A.S.S.; Ahmed, A.A.; et al. Could Allicin Alleviate Trastuzumab-Induced Cardiotoxicity in a Rat Model through Antioxidant, Anti-Inflammatory and Antihyperlipidemic Properties? Life Sci. 2022, 302, 120656. [Google Scholar] [CrossRef]
  36. Juergens, L.J.; Racké, K.; Tuleta, I.; Stoeber, M.; Juergens, U.R. Anti-Inflammatory Effects of 1,8-Cineole (Eucalyptol) Improve Glucocorticoid Effects In Vitro: A Novel Approach of Steroid-Sparing Add-on Therapy for COPD and Asthma. Synergy 2017, 5, 1–8. [Google Scholar] [CrossRef]
  37. Umar, S.; Zargan, J.; Umar, K.; Ahmad, S.; Katiyar, C.K.; Khan, H.A. Modulation of the Oxidative Stress and Inflammatory Cytokine Response by Thymoquinone in the Collagen Induced Arthritis in Wistar Rats. Chem. Biol. Interact. 2012, 197, 40–46. [Google Scholar] [CrossRef]
  38. Srinivasan, K. Cumin (Cuminum cyminum) and Black Cumin (Nigella sativa) Seeds: Traditional Uses, Chemical Constituents and Nutraceutical Effects. Food Qual. Saf. 2018, 2, 1–16. [Google Scholar] [CrossRef]
  39. Ali, B.H.; Blunden, G. Pharmacological and Toxicological Properties of Nigella sativa. Phytother. Res. 2003, 17, 299–305. [Google Scholar] [CrossRef]
  40. Ahmad, A.; Husain, A.; Mujeeb, M.; Khan, S.A.; Najmi, A.K.; Siddique, N.A.; Damanhouri, Z.A.; Anwar, F. A Review on Therapeutic Potential of Nigella sativa: A Miracle Herb. Asian Pac. J. Trop. Biomed. 2013, 3, 337–352. [Google Scholar] [CrossRef] [Green Version]
  41. Haque, M.; Sapna; Singh, R.; Nadeem, A.; Rasool, S.; Wani, J.A.; Khan, A.; Ashafaq, M.; Makeen, H.A.; Zehra, U. Nigella sativa: A promise for industrial and agricultural economic growth. In Black Seeds (Nigella sativa); Elsevier: Amsterdam, The Netherlands, 2022; pp. 439–460. [Google Scholar]
  42. Kooti, W.; Hasanzadeh-Noohi, Z.; Sharafi-Ahvazi, N.; Asadi-Samani, M.; Ashtary-Larky, D. Phytochemistry, Pharmacology and Therapeutic Uses of Black Seed (Nigella sativa). Chin. J. Nat. Med. 2016, 14, 732–745. [Google Scholar] [CrossRef]
  43. Ghosheh, O.A.; Houdi, A.A.; Crooks, P.A. High Performance Liquid Chromatographic Analysis of the Pharmacologically Active Quinones and Related Compounds in the Oil of the Black Seed (Nigella sativa L.). J. Pharm. Biomed. Anal. 1999, 19, 757–762. [Google Scholar] [CrossRef]
  44. Nivetha, K.; Prasanna, G. GC-MS and FT-IR Analysis of Nigella sativa L. Seeds. Int. J. Adv. Res. Biol. Sci. 2016, 19, 757–762. [Google Scholar]
  45. Tiji, S.; Benayad, O.; Berrabah, M.; El Mounsi, I.; Mimouni, M. Phytochemical Profile and Antioxidant Activity of Nigella sativa L. Growing in Morocco. Sci. World J. 2021, 2021, 6623609. [Google Scholar] [CrossRef] [PubMed]
  46. Yimer, E.M.; Tuem, K.B.; Karim, A.; Ur-Rehman, N.; Anwar, F. Nigella sativa L. (Black Cumin): A Promising Natural Remedy for Wide Range of Illnesses. Evid.-Based Complement. Alternat. Med. 2019, 2019, 1528635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Jayamurali, D.; Manoharan, N.; Govindarajulu, S.N. Phytochemical Composition, Therapeutical and Pharmacological Potential of Nigella sativa: A Review. Tradit. Med. Res. 2021, 6, 32. [Google Scholar]
  48. Hannan, M.A.; Rahman, M.A.; Sohag, A.A.M.; Uddin, M.J.; Dash, R.; Sikder, M.H.; Rahman, M.S.; Timalsina, B.; Munni, Y.A.; Sarker, P.P.; et al. Black Cumin (Nigella sativa L.): A Comprehensive Review on Phytochemistry, Health Benefits, Molecular Pharmacology and Safety. Nutrients 2021, 13, 1784. [Google Scholar] [CrossRef]
  49. Burdock, G.A. Assessment of Black Cumin (Nigella sativa L.) as a Food Ingredient and Putative Therapeutic Agent. Regul. Toxicol. Pharmacol. 2022, 128, 105088. [Google Scholar] [CrossRef]
  50. Mashayekhi-Sardoo, H.; Rezaee, R.; Karimi, G. Nigella sativa (Black Seed) Safety: An Overview. Asian Biomed. 2020, 14, 127–137. [Google Scholar] [CrossRef]
  51. Zaoui, A.; Cherrah, Y.; Mahassini, N.; Alaoui, K.; Amarouch, H.; Hassar, M. Acute and Chronic Toxicity of Nigella sativa Fixed Oil. Phytomedicine 2002, 9, 69–74. [Google Scholar] [CrossRef] [Green Version]
  52. Badary, O.A.; Al-Shabanah, O.A.; Nagi, M.N.; Al-Bekairi, A.M.; Elmazar, M.M.A. Acute and Subchronic Toxicity of Thymoquinone in Mice. Drug Dev. Res. 1998, 44, 56–61. [Google Scholar] [CrossRef]
  53. Hamed, M.A.; El-Rigal, N.S.; Ali, S.A. Effects of Black Seed Oil on Resolution of Hepato-Renal Toxicity Induced Bybromobenzene in Rats. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 569–581. [Google Scholar]
  54. EI-Hadiyah, T.M.; Raza, M.; Mohammed, O.Y.; Abdallah, A.A. Evaluation of Nigella sativa Seed Constituents for Their In Vivo Toxicity in Mice. Nat. Prod. Sci. 2003, 9, 22–27. [Google Scholar]
  55. Vahdati-Mashhadian, N.; Rakhshandeh, H.; Omidi, A. An investigation on LD50 and subacute hepatic toxicity of Nigella sativa seed extracts in mice. Die Pharm. 2005, 60, 544–547. [Google Scholar]
  56. Dollah, M.A.; Parhizkar, S.; Latiff, L.A.; Bin Hassan, M.H. Toxicity Effect of Nigella sativa on the Liver Function of Rats. Adv. Pharm. Bull. 2013, 3, 97–102. [Google Scholar]
  57. Zaghlol, D.A.A.; Kamel, E.S.; Mohammed, D.S.; Abbas, N.H. The Possible Toxic Effect of Different Doses of Nigella sativa Oil on the Histological Structure of the Liver and Renal Cortex of Adult Male Albino Rats. Egypt. J. Histol. 2012, 35, 127. [Google Scholar] [CrossRef]
  58. Fallah Huseini, H.; Amini, M.; Mohtashami, R.; Ghamarchehre, M.E.; Sadeqhi, Z.; Kianbakht, S.; Fallah Huseini, A. Blood Pressure Lowering Effect of Nigella sativa L. Seed Oil in Healthy Volunteers: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Phytother. Res. 2013, 27, 1849–1853. [Google Scholar] [CrossRef]
  59. Shoaei-Hagh, P.; Kamelan Kafi, F.; Najafi, S.; Zamanzadeh, M.; Heidari Bakavoli, A.; Ramezani, J.; Soltanian, S.; Asili, J.; Hosseinzadeh, H.; Eslami, S.; et al. A Randomized, Double-Blind, Placebo-Controlled, Clinical Trial to Evaluate the Benefits of Nigella sativa Seeds Oil in Reducing Cardiovascular Risks in Hypertensive Patients. Phytother. Res. 2021, 35, 4388–4400. [Google Scholar] [CrossRef]
  60. Asgary, S.; Sahebkar, A.; Goli-malekabadi, N. Ameliorative Effects of Nigella sativa on Dyslipidemia. J. Endocrinol. Investig. 2015, 38, 1039–1046. [Google Scholar] [CrossRef]
  61. Al-Amri, A.M.; Bamosa, A.O. Phase I Safety and Clinical Activity Study of Thymoquinone in Patients with Advanced Refractory Malignant Disease. Shiraz E-Med. J. 2009, 10, 107–111. [Google Scholar]
  62. Kalus, U.; Pruss, A.; Bystron, J.; Jurecka, M.; Smekalova, A.; Lichius, J.J.; Kiesewetter, H. Effect of Nigella sativa (Black Seed) on Subjective Feeling in Patients with Allergic Diseases. Phytother. Res. 2003, 17, 1209–1214. [Google Scholar] [CrossRef]
  63. Taka, E.; Mazzio, E.; Goodman, C.; Redmon, N.; Flores-Rozas, H.; Reams, R.; Darling-Reed, S.; Soliman, K. Anti-Inflammatory Effects of Thymoquinone in Activated BV-2 Microglia Cells. J. Neuroimmunol. 2015, 286, 5–12. [Google Scholar] [CrossRef] [Green Version]
  64. Fanoudi, S.; Alavi, M.S.; Hosseini, M.; Sadeghnia, H.R. Nigella sativa and Thymoquinone Attenuate Oxidative Stress and Cognitive Impairment Following Cerebral Hypoperfusion in Rats. Metab. Brain Dis. 2019, 34, 1001–1010. [Google Scholar] [CrossRef] [PubMed]
  65. Sedaghat, R.; Roghani, M.; Khalili, M. Neuroprotective Effect of Thymoquinone, the Nigella sativa Bioactive Compound, in 6-Hydroxydopamine-Induced Hemi-Parkinsonian Rat Model. Iran. J. Pharm. Res. 2014, 13, 227. [Google Scholar] [PubMed]
  66. Ebrahimi, S.S.; Oryan, S.; Izadpanah, E.; Hassanzadeh, K. Thymoquinone Exerts Neuroprotective Effect in Animal Model of Parkinson’s Disease. Toxicol. Lett. 2017, 276, 108–114. [Google Scholar] [CrossRef] [PubMed]
  67. Kooshki, A.; Tofighiyan, T.; Rastgoo, N.; Rakhshani, M.H.; Miri, M. Effect of Nigella sativa Oil Supplement on Risk Factors for Cardiovascular Diseases in Patients with Type 2 Diabetes Mellitus. Phytother. Res. 2020, 34, 2706–2711. [Google Scholar] [CrossRef] [PubMed]
  68. Badr, G.; Mahmoud, M.H.; Farhat, K.; Waly, H.; Al-Abdin, O.Z.; Rabah, D.M. Maternal Supplementation of Diabetic Mice with Thymoquinone Protects Their Offspring from Abnormal Obesity and Diabetes by Modulating Their Lipid Profile and Free Radical Production and Restoring Lymphocyte Proliferation via PI3K/AKT Signaling. Lipids Health Dis. 2013, 12, 37. [Google Scholar] [CrossRef] [Green Version]
  69. Medhet, M.; El-Bakly, W.M.; Badr, A.M.; Awad, A.; El-Demerdash, E. Thymoquinone Attenuates Isoproterenol-Induced Myocardial Infarction by Inhibiting Cytochrome C and Matrix Metalloproteinase-9 Expression. Clin. Exp. Pharmacol. Physiol. 2022, 49, 391–405. [Google Scholar] [CrossRef]
  70. Khalifa, A.A.; Rashad, R.M.; El-Hadidy, W.F. Thymoquinone Protects against Cardiac Mitochondrial DNA Loss, Oxidative Stress, Inflammation and Apoptosis in Isoproterenol-Induced Myocardial Infarction in Rats. Heliyon 2021, 7, e07561. [Google Scholar] [CrossRef]
  71. Simeonova, R. Comparative Study on the Protective Effects of Nigella sativa Oil, Curcumin, and Hydroxytyrosol against Dextran Sulphate Sodium-Induced Colitis in Mice. Farmacia 2022, 70, 447–455. [Google Scholar] [CrossRef]
  72. Arjumand, S.; Shahzad, M.; Shabbir, A.; Yousaf, M.Z. Thymoquinone Attenuates Rheumatoid Arthritis by Downregulating TLR2, TLR4, TNF-α, IL-1 and NFκB Expression Levels. Biomed. Pharmacother. 2019, 111, 958–963. [Google Scholar] [CrossRef]
  73. Yang, Y.; Bai, T.; Yao, Y.-L.; Zhang, D.-Q.; Wu, Y.-L.; Lian, L.-H.; Nan, J.-X. Upregulation of SIRT1-AMPK by Thymoquinone in Hepatic Stellate Cells Ameliorates Liver Injury. Toxicol. Lett. 2016, 262, 80–91. [Google Scholar] [CrossRef]
  74. Kong, L.Y.; Li, G.P.; Yang, P.; Xi, Z. Protective Effect of Thymoquinone on Cholestatic Rats with Liver Injury. Genet. Mol. Res. 2015, 14, 12247–12253. [Google Scholar] [CrossRef]
  75. Dur, A.; Kose, H.; Kocyigit, A.; Kocaman, O.; Ismayilova, M.; Sonmez, F.C. The anti-inflammatory and antioxidant effects of thymoquinone on ceruleine induced acute pancreatitis in rats. Bratisl. Lekárske Listy 2016, 117, 614–618. [Google Scholar] [CrossRef] [Green Version]
  76. Boskabady, M.; Khazdair, M.R.; Bargi, R.; Saadat, S.; Memarzia, A.; Mohammadian Roshan, N.; Hosseini, M.; Askari, V.R.; Boskabady, M.H. Thymoquinone Ameliorates Lung Inflammation and Pathological Changes Observed in Lipopolysaccharide-Induced Lung Injury. Evid.-Based Complement. Alternat. Med. 2021, 2021, 6681729. [Google Scholar] [CrossRef]
  77. Parlar, A.; Arslan, S.O. Thymoquinone Exhibits Anti-Inflammatory, Antioxidant, and Immunomodulatory Effects on Allergic Airway Inflammation. Arch. Clin. Exp. Med. 2019, 4, 60–65. [Google Scholar] [CrossRef] [Green Version]
  78. Rahman, I.; Mohammed, A.; AlShiddi, M.; Algazlan, A.; Alwably, A.; Hebbal, M.; Omar, M.G. The Assessment of Nigella sativa Oil as a Therapeutic Aid for Gingivitis: A Randomized Clinical Trial; Research Square: Durham, NC, USA, 2022. [Google Scholar]
  79. Ahmed, J.H.; Ibraheem, A.Y.; Al-Hamdi, K.I. Evaluation of Efficacy, Safety and Antioxidant Effect of Nigella sativa in Patients with Psoriasis: A Randomized Clinical Trial. J. Clin. Exp. Investig. 2014, 5, 186–193. [Google Scholar] [CrossRef] [Green Version]
  80. Aslam, H.; Shahzad, M.; Shabbir, A.; Irshad, S. Immunomodulatory Effect of Thymoquinone on Atopic Dermatitis. Mol. Immunol. 2018, 101, 276–283. [Google Scholar] [CrossRef]
  81. Guo, Q.; Wang, Y.; Xu, D.; Nossent, J.; Pavlos, N.J.; Xu, J. Rheumatoid Arthritis: Pathological Mechanisms and Modern Pharmacologic Therapies. Bone Res. 2018, 6, 15. [Google Scholar] [CrossRef] [Green Version]
  82. Nasuti, C.; Fedeli, D.; Bordoni, L.; Piangerelli, M.; Servili, M.; Selvaggini, R.; Gabbianelli, R. Anti-Inflammatory, Anti-Arthritic and Anti-Nociceptive Activities of Nigella sativa Oil in a Rat Model of Arthritis. Antioxidants 2019, 8, 342. [Google Scholar] [CrossRef] [Green Version]
  83. Gheita, T.A.; Kenawy, S.A. Effectiveness of Nigella sativa Oil in the Management of Rheumatoid Arthritis Patients: A Placebo Controlled Study. Phytother. Res. 2012, 26, 1246–1248. [Google Scholar] [CrossRef]
  84. Hadi, V.; Kheirouri, S.; Alizadeh, M.; Khabbazi, A.; Hosseini, H. Effects of Nigella sativa Oil Extract on Inflammatory Cytokine Response and Oxidative Stress Status in Patients with Rheumatoid Arthritis: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Avicenna J. Phytomed. 2016, 6, 34. [Google Scholar]
  85. Kheirouri, S.; Hadi, V.; Alizadeh, M. Immunomodulatory Effect of Nigella sativa Oil on T Lymphocytes in Patients with Rheumatoid Arthritis. Immunol. Investig. 2016, 45, 271–283. [Google Scholar] [CrossRef] [PubMed]
  86. Umar, S.; Hedaya, O.; Singh, A.K.; Ahmed, S. Thymoquinone Inhibits TNF-α-Induced Inflammation and Cell Adhesion in Rheumatoid Arthritis Synovial Fibroblasts by ASK1 Regulation. Toxicol. Appl. Pharmacol. 2015, 287, 299–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Faisal, R.; Ahmad, N.; Fahad, Y.S.; Chiragh, S. Anti-Arthritic Effect of Thymoquinone in Comparison with Methotrexate on Pristane Induced Arthritis in Female Sprague Dawley Rats. J. Ayub Med. Coll. Abbottabad 2018, 30, 3–7. [Google Scholar]
  88. Gajendran, M.; Loganathan, P.; Jimenez, G.; Catinella, A.P.; Ng, N.; Umapathy, C.; Ziade, N.; Hashash, J.G. A Comprehensive Review and Update on Ulcerative Colitis. Dis. Mon. 2019, 65, 100851. [Google Scholar] [CrossRef] [PubMed]
  89. Ozden, H.; Karaca, G. The Healing Effects of prp and Thymoquinone on dss Induced Inflammatory Bowel Disease in Rats. Ann. Med. Res. 2021, 28, 1496–1499. [Google Scholar] [CrossRef]
  90. Venkataraman, B.; Almarzooqi, S.; Raj, V.; Alhassani, A.T.; Alhassani, A.S.; Ahmed, K.J.; Subramanian, V.S.; Ojha, S.K.; Attoub, S.; Adrian, T.E.; et al. Thymoquinone, a Dietary Bioactive Compound, Exerts Anti-Inflammatory Effects in Colitis by Stimulating Expression of the Colonic Epithelial PPAR-γ Transcription Factor. Nutrients 2021, 13, 1343. [Google Scholar] [CrossRef]
  91. Tayman, C.; Cekmez, F.; Kafa, I.M.; Canpolat, F.E.; Cetinkaya, M.; Uysal, S.; Tunc, T.; Sarıcı, S.U. Beneficial Effects of Nigella sativa Oil on Intestinal Damage in Necrotizing Enterocolitis. J. Investig. Surg. 2012, 25, 286–294. [Google Scholar] [CrossRef]
  92. Kumar, A.; Sidhu, J.; Goyal, A. Alzheimer Disease; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK499922/ (accessed on 21 March 2023).
  93. Imtiaz, B.; Tolppanen, A.-M.; Kivipelto, M.; Soininen, H. Future Directions in Alzheimer’s Disease from Risk Factors to Prevention. Biochem. Pharmacol. 2014, 88, 661–670. [Google Scholar] [CrossRef]
  94. Hosseini, A.; Baradaran Rahimi, V.; Rakhshandeh, H.; Askari, V.R. Nigella sativa Oil Reduces LPS-Induced Microglial Inflammation: An Evaluation on M1/M2 Balance. Evid.-Based Complement. Alternat. Med. 2022, 2022, 5639226. [Google Scholar] [CrossRef]
  95. Madkour, D.A.; Ahmed, M.M.; Orabi, S.H.; Sayed, S.M.; Korany, R.M.S.; Khalifa, H.K. Nigella sativa Oil Protects against Emamectin Benzoate-Induced Neurotoxicity in Rats. Environ. Toxicol. 2021, 36, 1521–1535. [Google Scholar] [CrossRef]
  96. Beheshti, F.; Hosseini, M.; Vafaee, F.; Shafei, M.N.; Soukhtanloo, M. Feeding of Nigella sativa during Neonatal and Juvenile Growth Improves Learning and Memory of Rats. J. Tradit. Complement. Med. 2016, 6, 146–152. [Google Scholar] [CrossRef] [Green Version]
  97. Benkermiche, S.; Samir, D.; Haloui, M.; Bena-bed, M.; Tahraoui, A. Preventive Effects of Ginger Extract and Nigella sativa Oil on Anxiety and Depression Behavior in Wistar Rats Exposed to Mercuric Chloride. Pharmacogn. Res. 2022, 14, 1–4. [Google Scholar] [CrossRef]
  98. Hosseini, M.; Mohammadpour, T.; Karami, R.; Rajaei, Z.; Reza Sadeghnia, H.; Soukhtanloo, M. Effects of the Hydro-Alcoholic Extract of Nigella sativa on Scopolamine-Induced Spatial Memory Impairment in Rats and Its Possible Mechanism. Chin. J. Integr. Med. 2015, 21, 438–444. [Google Scholar] [CrossRef]
  99. Saleh, S.R.; Abdelhady, S.A.; Khattab, A.R.; El-Hadidy, W.F. Dual Prophylactic/Therapeutic Potential of Date Seed, and Nigella and Olive Oils-Based Nutraceutical Formulation in Rats with Experimentally-Induced Alzheimer’s Disease: A Mechanistic Insight. J. Chem. Neuroanat. 2020, 110, 101878. [Google Scholar] [CrossRef]
  100. Bin Sayeed, M.S.; Shams, T.; Fahim Hossain, S.; Rahman, M.R.; Mostofa, A.; Fahim Kadir, M.; Mahmood, S.; Asaduzzaman, M. Nigella sativa L. Seeds Modulate Mood, Anxiety and Cognition in Healthy Adolescent Males. J. Ethnopharmacol. 2014, 152, 156–162. [Google Scholar] [CrossRef]
  101. Abulfadl, Y.; El-Maraghy, N.; Ahmed, A.E.; Nofal, S.; Abdel-Mottaleb, Y.; Badary, O. Thymoquinone Alleviates the Experimentally Induced Alzheimer’s Disease Inflammation by Modulation of TLRs Signaling. Hum. Exp. Toxicol. 2018, 37, 1092–1104. [Google Scholar] [CrossRef]
  102. Abulfadl, Y.S.; El-Maraghy, N.N.; Ahmed, A.A.E.; Nofal, S.; Badary, O.A. Protective Effects of Thymoquinone on D-Galactose and Aluminum Chloride Induced Neurotoxicity in Rats: Biochemical, Histological and Behavioral Changes. Neurol. Res. 2018, 40, 324–333. [Google Scholar] [CrossRef]
  103. Velagapudi, R.; Kumar, A.; Bhatia, H.S.; El-Bakoush, A.; Lepiarz, I.; Fiebich, B.L.; Olajide, O.A. Inhibition of Neuroinflammation by Thymoquinone Requires Activation of Nrf2/ARE Signalling. Int. Immunopharmacol. 2017, 48, 17–29. [Google Scholar] [CrossRef]
  104. Chu, Y.; Dodiya, H.; Aebischer, P.; Olanow, C.W.; Kordower, J.H. Alterations in Lysosomal and Proteasomal Markers in Parkinson’s Disease: Relationship to Alpha-Synuclein Inclusions. Neurobiol. Dis. 2009, 35, 385–398. [Google Scholar] [CrossRef]
  105. Jahromy, M.H.; Jalili, M.; Mohajer, A.J.; Poor, F.K.; Dara, S.M. Effects of Nigella sativa Seed Extract on Perphenzine-Induced Muscle Rigidity in Male Mice. World J. Neurosci. 2014, 2014, 313–318. [Google Scholar] [CrossRef] [Green Version]
  106. Bawani, S.S.; Anandhi, U. GC-MS Analysis of Nigella sativa Seed Extract and Its Ameliorative Effects on Transgenic Drosophila Model of Parkinson Disease. Int. J. Eng. Technol. 2021, 4, 16–22. [Google Scholar]
  107. Sandhua, K.S.; Ranab, A.C. Evaluation of Anti Parkinson’s Activity of Nigella sativa (Kalonji) Seeds in Chlorpromazine Induced Experimental Animal Model. Int. J. Pharm. Pharm. Sci. 2013, 5, 884–888. [Google Scholar]
  108. Malik, T.; Hasan, S.; Pervez, S.; Fatima, T.; Haleem, D.J. Nigella sativa Oil Reduces Extrapyramidal Symptoms (EPS)-Like Behavior in Haloperidol-Treated Rats. Neurochem. Res. 2016, 41, 3386–3398. [Google Scholar] [CrossRef]
  109. Agarwal, A.K.; Raja, A.; Brown, B.D. Chronic Obstructive Pulmonary Disease; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK559281/ (accessed on 3 April 2023).
  110. Al-Azzawi, M.A.; AboZaid, M.M.N.; Ibrahem, R.A.L.; Sakr, M.A. Therapeutic Effects of Black Seed Oil Supplementation on Chronic Obstructive Pulmonary Disease Patients: A Randomized Controlled Double Blind Clinical Trial. Heliyon 2020, 6, e04711. [Google Scholar] [CrossRef] [PubMed]
  111. Kacem, R. Neutrophils Involvements in COPD: Effects of Essential Oil Extracted from Nigella sativa (L.) Seeds on Human Neutrophil Functions and Elastase Activity. Ph.D. Thesis, Faculté des Sciences de la Nature et de la Vie, Setif, Algeria, 2018. [Google Scholar]
  112. Koshak, A.; Wei, L.; Koshak, E.; Wali, S.; Alamoudi, O.; Demerdash, A.; Qutub, M.; Pushparaj, P.N.; Heinrich, M. Nigella sativa Supplementation Improves Asthma Control and Biomarkers: A Randomized, Double-Blind, Placebo-Controlled Trial. Phytother. Res. 2017, 31, 403–409. [Google Scholar] [CrossRef] [Green Version]
  113. Barlianto, W.; Rachmawati, M.; Irawan, M.; Wulandari, D. Effects of Nigella sativa Oil on Th1/Th2, Cytokine Balance and Improvement of Asthma Control in Children. Paediatr. Indones. 2017, 57, 223. [Google Scholar] [CrossRef]
  114. Koshak, A.E.; Yousif, N.M.; Fiebich, B.L.; Koshak, E.A.; Heinrich, M. Comparative Immunomodulatory Activity of Nigella sativa L. Preparations on Proinflammatory Mediators: A Focus on Asthma. Front. Pharmacol. 2018, 9, 1075. [Google Scholar] [CrossRef] [Green Version]
  115. Ikhsan, M.; Hiedayati, N.; Maeyama, K.; Nurwidya, F. Nigella sativa as an Anti-Inflammatory Agent in Asthma. BMC Res. Notes 2018, 11, 744. [Google Scholar] [CrossRef]
  116. Cucinotta, D.; Vanelli, M. WHO Declares COVID-19 a Pandemic. Acta Bio-Med. Atenei Parm. 2020, 91, 157–160. [Google Scholar]
  117. Upadhyay, J.; Tiwari, N.; Ansari, M.N. Role of Inflammatory Markers in Corona Virus Disease (COVID-19) Patients: A Review. Exp. Biol. Med. 2020, 245, 1368–1375. [Google Scholar] [CrossRef]
  118. Salem, M.L. Immunomodulatory and Therapeutic Properties of the Nigella sativa L. Seed. Int. Immunopharmacol. 2005, 5, 1749–1770. [Google Scholar] [CrossRef]
  119. Rahman, M.T. Potential Benefits of Combination of Nigella sativa and Zn Supplements to Treat COVID-19. J. Herb. Med. 2020, 23, 100382. [Google Scholar] [CrossRef]
  120. Khan, S.L.; Siddiqui, F.A.; Jain, S.P.; Sonwane, G.M. Discovery of Potential Inhibitors of SARS-CoV-2 (COVID-19) Main Protease (Mpro) from Nigella sativa (Black Seed) by Molecular Docking Study. Coronaviruses 2021, 2, 384–402. [Google Scholar] [CrossRef]
  121. Ali, K.; Faiq, T.; Ghareeb, O. Preventive Value of Black Seed in People at Risk of Infection with COVID-19. Pak. J. Med. Health Sci. 2021, 15, 384–387. [Google Scholar]
  122. Al-Haidari, K.A.A.; Faiq, T.; Ghareeb, O. Clinical Trial of Black Seeds against COVID-19 in Kirkuk City/Iraq. Indian J. Forensic Med. Toxicol. 2021, 15, 3393–3399. [Google Scholar]
  123. Koshak, A.E.; Koshak, E.A.; Mobeireek, A.F.; Badawi, M.A.; Wali, S.O.; Malibary, H.M.; Atwah, A.F.; Alhamdan, M.M.; Almalki, R.A.; Madani, T.A. Nigella sativa for the Treatment of COVID-19: An Open-Label Randomized Controlled Clinical Trial. Complement. Ther. Med. 2021, 61, 102769. [Google Scholar] [CrossRef]
  124. Ashraf, S.; Ashraf, S.; Ashraf, M.; Imran, M.A.; Kalsoom, L.; Siddiqui, U.N.; Farooq, I.; Akmal, R.; Akram, M.K.; Ashraf, S.; et al. Honey and Nigella sativa against COVID-19 in Pakistan (HNS-COVID-PK): A Multicenter Placebo-Controlled Randomized Clinical Trial. Phytother. Res. 2023, 37, 627–644. [Google Scholar] [CrossRef]
  125. Rochlani, Y.; Pothineni, N.V.; Kovelamudi, S.; Mehta, J.L. Metabolic Syndrome: Pathophysiology, Management and Modulation by Natural Compounds. Ther. Adv. Cardiovasc. Dis. 2017, 11, 215–225. [Google Scholar] [CrossRef] [Green Version]
  126. Sultan, M.T.; Butt, M.S.; Karim, R.; Iqbal, S.Z.; Ahmad, S.; Zia-Ul-Haq, M.; Aliberti, L.; Ahmad, A.N.; De Feo, V. Effect of Nigella sativa Fixed and Essential Oils on Antioxidant Status, Hepatic Enzymes, and Immunity in Streptozotocin Induced Diabetes Mellitus. BMC Complement. Altern. Med. 2014, 14, 193. [Google Scholar] [CrossRef] [Green Version]
  127. Farzaneh, E.; Nia, F.R.; Mehrtash, M.; Mirmoeini, F.S.; Jalilvand, M. The Effects of 8-Week Nigella sativa Supplementation and Aerobic Training on Lipid Profile and VO2 Max in Sedentary Overweight Females. Int. J. Prev. Med. 2014, 5, 210. [Google Scholar]
  128. Mahdavi, R.; Namazi, N.; Alizadeh, M.; Farajnia, S. Effects of Nigella sativa Oil with a Low-Calorie Diet on Cardiometabolic Risk Factors in Obese Women: A Randomized Controlled Clinical Trial. Food Funct. 2015, 6, 2041–2048. [Google Scholar] [CrossRef] [PubMed]
  129. Pelegrin, S.; Galtier, F.; Chalançon, A.; Gagnol, J.-P.; Barbanel, A.-M.; Pélissier, Y.; Larroque, M.; Lepape, S.; Faucanié, M.; Gabillaud, I.; et al. Effects of Nigella sativa Seeds (Black Cumin) on Insulin Secretion and Lipid Profile: A Pilot Study in Healthy Volunteers. Br. J. Clin. Pharmacol. 2019, 85, 1607. [Google Scholar] [CrossRef] [PubMed]
  130. Kudaravalli, P.; John, S. Nonalcoholic Fatty Liver; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK541033/ (accessed on 11 March 2023).
  131. Khonche, A.; Huseini, H.F.; Gholamian, M.; Mohtashami, R.; Nabati, F.; Kianbakht, S. Standardized Nigella sativa Seed Oil Ameliorates Hepatic Steatosis, Aminotransferase and Lipid Levels in Non-Alcoholic Fatty Liver Disease: A Randomized, Double-Blind and Placebo-Controlled Clinical Trial. J. Ethnopharmacol. 2019, 234, 106–111. [Google Scholar] [CrossRef] [PubMed]
  132. Darand, M.; Darabi, Z.; Yari, Z.; Hedayati, M.; Shahrbaf, M.A.; Khoncheh, A.; Hosseini-Ahangar, B.; Alavian, S.M.; Hekmatdoost, A. The Effects of Black Seed Supplementation on Cardiovascular Risk Factors in Patients with Nonalcoholic Fatty Liver Disease: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Phytother. Res. 2019, 33, 2369–2377. [Google Scholar] [CrossRef]
  133. Hussain, M.; Tunio, A.G.; Akhtar, L.; Shaikh, G.S. Effects of Nigella sativa on Various Parameters in Patients of Non-Alcoholic Fatty Liver Disease. J. Ayub Med. Coll. Abbottabad 2017, 29, 403–407. [Google Scholar]
  134. Darand, M.; Darabi, Z.; Yari, Z.; Saadati, S.; Hedayati, M.; Khoncheh, A.; Hosseini-Ahangar, B.; Alavian, S.M.; Hekmatdoost, A. Nigella sativa and Inflammatory Biomarkers in Patients with Non-Alcoholic Fatty Liver Disease: Results from a Randomized, Double-Blind, Placebo-Controlled, Clinical Trial. Complement. Ther. Med. 2019, 44, 204–209. [Google Scholar] [CrossRef]
  135. Rashidmayvan, M.; Mohammadshahi, M.; Seyedian, S.S.; Haghighizadeh, M.H. The Effect of Nigella sativa Oil on Serum Levels of Inflammatory Markers, Liver Enzymes, Lipid Profile, Insulin and Fasting Blood Sugar in Patients with Non-Alcoholic Fatty Liver. J. Diabetes Metab. Disord. 2019, 18, 453–459. [Google Scholar] [CrossRef]
  136. Mahmoud, Y.K.; Abdelrazek, H.M.A. Cancer: Thymoquinone Antioxidant/pro-Oxidant Effect as Potential Anticancer Remedy. Biomed. Pharmacother. 2019, 115, 108783. [Google Scholar] [CrossRef]
  137. Ansary, J.; Giampieri, F.; Forbes-Hernandez, T.Y.; Regolo, L.; Quinzi, D.; Gracia Villar, S.; Garcia Villena, E.; Tutusaus Pifarre, K.; Alvarez-Suarez, J.M.; Battino, M.; et al. Nutritional Value and Preventive Role of Nigella sativa L. and Its Main Component Thymoquinone in Cancer: An Evidenced-Based Review of Preclinical and Clinical Studies. Molecules 2021, 26, 2108. [Google Scholar] [CrossRef]
  138. Zhao, Z.; Liu, L.; Li, S.; Hou, X.; Yang, J. Advances in Research on the Relationship between Thymoquinone and Pancreatic Cancer. Front. Oncol. 2023, 12, 1092020. [Google Scholar] [CrossRef]
  139. Khurshid, Y.; Syed, B.; Simjee, S.U.; Beg, O.; Ahmed, A. Antiproliferative and Apoptotic Effects of Proteins from Black Seeds (Nigella sativa) on Human Breast MCF-7 Cancer Cell Line. BMC Complement. Med. Ther. 2020, 20, 5. [Google Scholar] [CrossRef] [Green Version]
  140. Shanmugam, M.K.; Ahn, K.S.; Hsu, A.; Woo, C.C.; Yuan, Y.; Tan, K.H.B.; Chinnathambi, A.; Alahmadi, T.A.; Alharbi, S.A.; Koh, A.P.F.; et al. Thymoquinone Inhibits Bone Metastasis of Breast Cancer Cells Through Abrogation of the CXCR4 Signaling Axis. Front. Pharmacol. 2018, 9, 1294. [Google Scholar] [CrossRef]
  141. Subburayan, K.; Thayyullathil, F.; Pallichankandy, S.; Rahman, A.; Galadari, S. Par-4-Dependent P53 up-Regulation Plays a Critical Role in Thymoquinone-Induced Cellular Senescence in Human Malignant Glioma Cells. Cancer Lett. 2018, 426, 80–97. [Google Scholar] [CrossRef]
  142. Almajali, B.; Nagi Al-Jamal, H.A.; Wan Taib, W.R.; Ismail, I.; Johan, M.F.; Doolaanea, A.A.; Ibrahim, W.N.; Tajudin, S.A. Thymoquinone Suppresses Cell Proliferation and Enhances Apoptosis of HL60 Leukemia Cells through Re-Expression of JAK/STAT Negative Regulators. Asian Pac. J. Cancer Prev. 2021, 22, 879–885. [Google Scholar] [CrossRef]
  143. Samarghandian, S.; Azimi-Nezhad, M.; Farkhondeh, T. Thymoquinone-Induced Antitumor and Apoptosis in Human Lung Adenocarcinoma Cells. J. Cell. Physiol. 2019, 234, 10421–10431. [Google Scholar] [CrossRef]
  144. Alhassani, M.Y.; Zohny, S.F.; Sheikh, R.A.; Hassan, M.A.; Kalantan, A.A.; Hosawi, S.; Alhosin, M. Thymoquinone Exerts Anti-Tumor Activities on Human Hepatocellular Carcinoma Cells: Role of Angiogenesis-Related Genes VCAN, Grb2 and EZH2. Eur. J. Cell Sci. 2019, 10, 10–16. [Google Scholar] [CrossRef]
  145. Dera, A.; Rajagopalan, P. Thymoquinone Attenuates Phosphorylation of AKT to Inhibit Kidney Cancer Cell Proliferation. J. Food Biochem. 2019, 43, e12793. [Google Scholar] [CrossRef]
  146. Zhou, X.; Wang, F.; Wu, H.; Chen, X.; Zhang, Y.; Lin, J.; Cai, Y.; Xiang, J.; He, N.; Hu, Z.; et al. Thymoquinone Suppresses the Proliferation, Migration and Invasiveness through Regulating ROS, Autophagic Flux and MiR-877-5p in Human Bladder Carcinoma Cells. Int. J. Biol. Sci. 2021, 17, 3456–3475. [Google Scholar] [CrossRef]
  147. Narayanan, P.; Farghadani, R.; Nyamathulla, S.; Rajarajeswaran, J.; Thirugnanasampandan, R.; Bhuwaneswari, G. Natural Quinones Induce ROS-Mediated Apoptosis and Inhibit Cell Migration in PANC-1 Human Pancreatic Cancer Cell Line. J. Biochem. Mol. Toxicol. 2022, 36, e23008. [Google Scholar] [CrossRef]
  148. Kale, E.; Kale, A.; Bozali, K.; Gulgec, A.S.; Ozdemir, M.; Yalcin, B.; Guler, E.M. TQ-Ox, a Novel Synthetic Derivative of Thymoquinone on Ovarian Cancer Cells In Vitro. Nat. Prod. Res. 2022, 8, 1–10. [Google Scholar] [CrossRef]
  149. Doolaanea, A.A.; Mansor, N.I.; Mohd Nor, N.H.; Mohamed, F. Co-Encapsulation of Nigella sativa Oil and Plasmid DNA for Enhanced Gene Therapy of Alzheimer’s Disease. J. Microencapsul. 2016, 33, 114–126. [Google Scholar] [CrossRef] [PubMed]
  150. Ramachandran, S.; Thangarajan, S. A Novel Therapeutic Application of Solid Lipid Nanoparticles Encapsulated Thymoquinone (TQ-SLNs) on 3-Nitroproponic Acid Induced Huntington’s Disease-like Symptoms in Wistar Rats. Chem. Biol. Interact. 2016, 256, 25–36. [Google Scholar] [CrossRef] [PubMed]
  151. Alkhalaf, M.I.; Hussein, R.H.; Hamza, A. Green Synthesis of Silver Nanoparticles by Nigella sativa Extract Alleviates Diabetic Neuropathy through Anti-Inflammatory and Antioxidant Effects. Saudi J. Biol. Sci. 2020, 27, 2410–2419. [Google Scholar] [CrossRef] [PubMed]
  152. Jihad, M.A.; Noori, F.T.M.; Jabir, M.S.; Albukhaty, S.; AlMalki, F.A.; Alyamani, A.A. Polyethylene Glycol Functionalized Graphene Oxide Nanoparticles Loaded with Nigella sativa Extract: A Smart Antibacterial Therapeutic Drug Delivery System. Molecules 2021, 26, 3067. [Google Scholar] [CrossRef] [PubMed]
  153. Nauroze, T.; Ali, S.; Kanwal, L.; Ara, C.; Akbar Mughal, T.; Andleeb, S. Ameliorative Effect of Nigella sativa Conjugated Silver Nanoparticles against Chromium-Induced Hepatotoxicity and Renal Toxicity in Mice. Saudi J. Biol. Sci. 2023, 30, 103571. [Google Scholar] [CrossRef]
  154. Yan, W.; Liu, Y.; Mansooridara, S.; Kalantari, A.S.; Sadeghian, N.; Taslimi, P.; Zangeneh, A.; Zangeneh, M.M. Chemical Characterization and Neuroprotective Properties of Copper Nanoparticles Green-Synthesized by Nigella sativa L. Seed Aqueous Extract against Methadone-Induced Cell Death in Adrenal Phaeochromocytoma (PC12) Cell Line. J. Exp. Nanosci. 2020, 15, 280–296. [Google Scholar] [CrossRef]
  155. Saghir, S.A.M.; Al-Gabri, N.A.; Ali, A.A.; Al-Attar, A.-S.R.; Al-Sobarry, M.; Al-Shargi, O.Y.A.; Alotaibi, A.; Al-Zharani, M.; Nasr, F.A.; Al-Balagi, N.; et al. Ameliorative Effect of Thymoquinone-Loaded PLGA Nanoparticles on Chronic Lung Injury Induced by Repetitive Intratracheal Instillation of Lipopolysaccharide in Rats. Oxid. Med. Cell. Longev. 2021, 2021, e5511523. [Google Scholar] [CrossRef]
  156. Saghir, S.A.M.; Al-Gabri, N.A.; Khafaga, A.F.; El-shaer, N.H.; Alhumaidh, K.A.; Elsadek, M.F.; Ahmed, B.M.; Alkhawtani, D.M.; Abd El-Hack, M.E. Thymoquinone-PLGA-PVA Nanoparticles Ameliorate Bleomycin-Induced Pulmonary Fibrosis in Rats via Regulation of Inflammatory Cytokines and INOS Signaling. Animals 2019, 9, 951. [Google Scholar] [CrossRef] [Green Version]
  157. Negi, P.; Sharma, I.; Hemrajani, C.; Rathore, C.; Bisht, A.; Raza, K.; Katare, O.P. Thymoquinone-Loaded Lipid Vesicles: A Promising Nanomedicine for Psoriasis. BMC Complement. Altern. Med. 2019, 19, 334. [Google Scholar] [CrossRef] [Green Version]
  158. Rohini, B.; Akther, T.; Waseem, M.; Khan, J.; Kashif, M.; Hemalatha, S. AgNPs from Nigella sativa Control Breast Cancer: An In Vitro Study. J. Environ. Pathol. Toxicol. Oncol. 2019, 38, 185–194. [Google Scholar] [CrossRef]
Figure 1. Interrelationship between Inflammation and oxidative stress. TNF-α: Tumor necrosis factor alpha; IL-1: Interleukin-1; ROS: Reactive oxygen species; NF-κB: Nuclear factor-kappa B; AP-1: activator protein-1; Nrf2: Nuclear factor erythroid 2-related factor 2; TGF-β: transforming growth factor-beta; IL-10: Interleukin-10.
Figure 1. Interrelationship between Inflammation and oxidative stress. TNF-α: Tumor necrosis factor alpha; IL-1: Interleukin-1; ROS: Reactive oxygen species; NF-κB: Nuclear factor-kappa B; AP-1: activator protein-1; Nrf2: Nuclear factor erythroid 2-related factor 2; TGF-β: transforming growth factor-beta; IL-10: Interleukin-10.
Ddc 02 00027 g001
Figure 2. The molecular structure of thymoquinone.
Figure 2. The molecular structure of thymoquinone.
Ddc 02 00027 g002
Figure 3. Most prominent effects of black cumin seed related to key molecules involved in inflammation and oxidative stress. ↑ increase; ↓ decrease.
Figure 3. Most prominent effects of black cumin seed related to key molecules involved in inflammation and oxidative stress. ↑ increase; ↓ decrease.
Ddc 02 00027 g003
Table 1. The different compounds identified in black cumin seed [40,44,46,47,48].
Table 1. The different compounds identified in black cumin seed [40,44,46,47,48].
GroupCompounds
Proteins
(Amino acids)
Cysteine, methionine, glutamate, aspartate, arginine, alanine, valine, glycine, isoleucine, leucine, tyrosine, lysine, proline, threonine, serine, phenylalanine
CarbohydratesArabinose, glucose, rhamnose, xylose
SterolsCholesterol, campesterol, β-sitosterol, stigmasterol, 5-avenasterol
Terpenes and TerpenoidsThymoquinone, thymohydroquinone, dithymoquinone, thymol, p-cymene (7–15%), longifolene, limonene, longifolene, α-pinene, citronellol, carvone, 4-terpineol (2–7%), carvacrol (6–12%), t-anethole
Fatty acidsStearic acid, palmitic acid, oleic acid (20%), linoleic acid (50–60%), eicosadienoic acid (3%), eicosanoic acid, dihomolinoleic acid, tetradecanoic acid
AlkaloidsNigellidine, nigellicine, nigellicimine, nigellicimine-n-oxide
Tocolsα- tocopherol, γ-tocopherol, β-tocotrienol (Vit. E)
Saponinα-hederin, hederagenin
Cumarins7-oxy-coumarin, 7-hydroxy-coumarin, 6-methoxy-coumarin
PolyphenolsApigenin, caffeic acid, caftaric acid, chlorogenic acid, cichoric acid, gentisic acid, ferulic acid, fisetin, hyperoside, isoquercitrin, kaempferol, luteolin, myricetin, p-coumaric acid, patuletin, quercitrin, quercetin, rutin, sinapic acid
Steroidal
glycosides
Stigma-5,22-dien-3-β-D-glucopyranoside,
3-O-[β-Doxylopyranosyl-(1-2)-α-L-rhamnopyranosyl-(1-2)-β-D-glucopyranosyl]-11-methoxy-16,
23-dihydroxy-28-methylolean-12-enoate,
3-O-[β-D-xylopyranosyl-(1-3)-α-L-rhamnopyranosyl-(1-4)-β-D-glucopy-ranosyl]-11-methoxy-16-hydroxy-17-acetoxy hederagenin
PhospholipidsPhosphatidylinositol, phosphatidylglycerol, phosphatidylcholine
MineralsCalcium, copper, iron, potassium, magnesium, manganese, phosphorus, sodium, selenium, zinc
VitaminsFolic acid, riboflavin, niacin, Vit. A, Vit. C, thiamin, pyridoxine
Table 2. Studies of BCS’s therapeutic effects in inflammation-related disorders.
Table 2. Studies of BCS’s therapeutic effects in inflammation-related disorders.
DisorderTQ/BCS FormulaModelImproved ParametersRef.
Neuro-inflammationTQ (10 μM)BV-2 cells↓ NO2− and iNOS
↓ IL-6, IL-12, p40/70
↓ Granulocyte colony-stimulating factor
↓ CCL12/MCP-5 and CCL2/MCP-1
[63]
Cognitive impairment
(Alzheimer’s disease)
BCS (100, 200, and 400 mg/kg) i.p.
TQ (10, 20, and 40 mg/kg) i.p.
Rats↓ MDA
↑ SOD
↓ AChE
- Ameliorate learning and memory impairments
[64]
Parkinson’s diseaseTQ (5 and/10 mg/Kg) orallyRats↓ MDA, Nitrite levels
↑ SOD
↓ loss of substantia nigra pars compacta neurons
- Improved the rotational
Behavior
[65]
TQ (7.5 and 15 mg/kg)
Orally
Rats↑ Tyrosine hydroxylase, dopamine, and parkin levels
↓ Dynamin-related protein-1
- Enhance the motor functions
[66]
Diabetes mellitusBCS oil (500 mg twice a day for 8 weeks) orallyDiabetic patients↑ HDL-C levels
↓ MDA and hs-CRP, TC, L DL-C
↓ Triglycerides and FBG levels
[67]
TQ (20 mg/kg)Pregnant mice and their offspring↓ Body weight
↓ BGL, ↑ Insulin
↓ LDL-C, TC, MDA
↑ IL-2, IL-4, IL-7,
↓ IL-6, IL-1β and TNF-α
[68]
Myocardial infarctionTQ at (10 and 20 mg/kg) orallyRats↑ GSH and TAC
↓ TNF-α and NF-κB
↓ Metalloproteinase 9
↓ Cytochrome-C and Caspase 3 and 9
[69]
TQ (20 mg/kg) orally↑ GSH, GPx, SOD, CAT, BCL-2
↓ IL-6, IL-1β, and TNF-α MDA
↓ BAX, Caspase-3
↓ Troponin I and LDH
[70]
Ulcerative colitisBCS oil
(2.0 mL/kg)
Mice↑ CAT, SOD
↑ GSH, GPx
↓ IL6, MDA, NO
↓ CRP, MPO
[71]
Rheumatic arthritisTQ at (10 mg/kg)
i.p.
Rats↓ IL-1, TNF-α, NF-κB
↓ TLR4 and TLR2
↓ CRP
[72]
Liver injuryTQ (12.5 μM)(LX-2) hepatic stellate cells↑ LKB1 and AMPK phosphorylation
↓ Collagen-Ι, α-SMA, TIMP-1
↑ MMP-13
↑ (PPAR-γ) expression
[73]
TQ (20 or 40 mg/kg) orallyMice↓ Serum aminotransferase
↓ Hepatic triglyceride
↓ Collagen-Ι, α-SMA
↑ SIRT1
↑ LKB1 and AMPK phosphorylation
TQ (25, 50 mg/kg)
Orally
Rats↓ Hydroxyproline and MDA
↑ SOD and GPx
[74]
PancreatitisTQ (5 mg/kg) i.p.Rats↓ IL-1β
↑ TAC
↓ TOS
- Ameliorate histopathologic findings
[75]
Lung InjuryTQ (5 or 10 mg/kg) i.p.Rats↓ PGE2, TGF-β, IFN-γ
↑ IL-4
↓Epithelial damage emphysema scores of lung tissue
↓ Eosinophil, neutrophil, and
monocyte%
↑ lymphocyte%
[76]
AsthmaTQ (1, 10, and 30 mg/kg)
i.p.
Rats↓ IL-6 and TNF-α in BALF
↓ MDA in lung tissue
↓ Total WBCs in BALF
↓ Lymphocyte Eosinophil, Monocyte, Neutrophil
↓ Evans blue dye
[77]
GingivitisBCS oil (diluted with water 50%)
Rinsing
Gingivitis patients↓ IL-6
↓ Colony-forming units
↓ Pathogenic bacteria
[78]
PsoriasisBCS (ointment, powder, capsule, combination of ointment and capsule)Psoriatic patients↓ Psoriasis Area and Severity Index score
↓ MDA
- Cure of psoriatic lesions
[79]
Atopic dermatitisTQ (10 mg/kg dissolved in ethanol/distilled water) orally,
TQ (5 μmol in 0.2 mL acetone) topical
Mice↓ IgE level
↓ IL-4, IL-5
↓ IFN-γ
[80]
↑ increase; ↓ decrease; i.p.: Intraperitoneal injection; IL: interleukin; CCL12/MCP-5: chemokine (C-C motif) ligand 12/monocyte chemoattractant protein-5; CCL2/MCP-1: chemokine (C-C motif) ligand 2/monocyte chemoattractant protein-1; MDA: malondialdehyde; SOD: superoxide dismutase; CAT: catalase; AChE: acetylcholinesterase; hs-CRP: high-sensitivity C-reactive protein; HDL-C: high-density lipoprotein cholesterol; LDL-C: low-density lipoprotein cholesterol; BGL: blood glucose level; TC: total cholesterol; TNF-α: tumor necrosis factor-alpha; GSH: reduced glutathione; GPx: glutathione peroxidase; TAC: total antioxidant capacity; TOS: total oxidative stress; NF-κB: nuclear factor-kappa B; LDH: lactate dehydrogenase; Bax: bcl-2-associated X protein; BCL-2: B-cell lymphoma 2 protein; α-SMA: alpha-smooth muscle actin; TIMP-1: tissue inhibitor of metalloproteinase 1; MMP-13: matrix metalloproteinase-13; SIRT1: sirtuin 1; LKB1: liver kinase B1; PGE2: prostaglandin E2; TGF-β: transforming growth factor-beta; BALF: bronchoalveolar lavage fluid; IgE: immunoglobulin E; IFN-γ: interferon-gamma; AMPK: AMP-activated protein kinase.
Table 3. Updated studies of using BCS constituents in various types of cancer.
Table 3. Updated studies of using BCS constituents in various types of cancer.
Cancer
Type
TQ/BCS FormulaModelImproved ParametersRef.
Breast cancerBCS protein extractMCF-7 cells↑ BAX
↓ BCL-2
↑ Caspase-3
↓ Survivin
[139]
TQ (50μM)MDA-MB-231 triple negative breast cancer (TNBC)↓ CXCR4
↓ NF-κB
[140]
TQ (2 and 4 mg/kg)Mice↓ CXCR4
↓ Metastases and metastatic biomarkers
↓ Osteolytic lesions
Brain cancerTQ (10–100 μM)Glioma cells
(U87MG,
U118MG,
A172)
↑ Cell cycle arrest
↑ Par-4
↑ p53, p21, Rb
↓ Lamin B1, CDK-2 and Cyclin E
[141]
Blood cancerTQ
(1, 2, 3 μM)
HL60 cells↑ Cell cycle arrest
↓ Cell proliferation
↑ Apoptotic activity
↑ Negative Regulators of JAK/STAT: SOCS-1, SOCS-3 and SHP-1
[142]
Lung cancerTQ (25, 50, 100μM)A549 cells↑ BAX
↓ BCL-2
↑ p53
↑ Caspases-3 and -9
[143]
Hepatic cancerTQ (10, 30, 50 μM)HepG2↑ Apoptosis
↓ Angiogenesis-related genes:
VCAN, Grb2 and EZH2 expressions
[144]
Kidney cancerTQ
(20, 40 μM)
(25, 50 μM)
A498 cells and
Caki-1 cells
↑ Cell cycle arrest
↑ BAX
↓ BCL-2
↓ Akt phosphorylation
[145]
Bladder cancerTQ
(25, 50 μM)
5637 and T24 cells↑ ROS
↑ Bax, cleaved caspase 3, PARP, cleaved poly (ADP-ribose).
↓ BCL-2, BCL-XL
↑ Beclin-1, ATG7 and LC3B proteins
↑ miR-877–5p
[146]
Prostate cancerTQ
(11.11, 22.22, 44.44 μM)
Panc-1 cells↑ ROS
↓ MMP-9
[147]
Ovarian cancerTQ oxime derivative (2.5–100 μM)SKOV-3 cells
CHO-K1 cells
↑ Intracellular ROS and Ca2+
↓ GSH
[148]
↑ increase; ↓ decrease; BAX: Bcl-2-associated X protein; BCL-2: B-cell lymphoma 2 protein; CXCR4: C-X-C chemokine receptor type 4; NF-κB: nuclear factor-kappa B; Par-4: prostate apoptosis response 4; p53: tumor protein 53; p21: cyclin-dependent kinase inhibitor 1; Rb: retinoblastoma protein; CDK-2: cyclin-dependent kinase 2; SOCS-1: suppressors of cytokine signaling 1; SOCS-3: suppressors of cytokine signaling 3; SHP-1: Src homology region 2 domain-containing phosphatase-1; VCAN: versican; Grb2: growth factor receptor-bound protein 2; EZH2: enhancer of zeste homolog 2 (histone methyltransferase for lysine 27 of histone 3); PARP: poly-ADP ribose polymerase; ROS: reactive oxygen species; Akt: protein kinase B (PKB); ATG7: autophagy-related protein 7; LC3B: microtubule-associated protein light chain 3 beta; MMP-9: matrix metalloproteinase-9; GSH: reduced glutathione.
Table 4. Nano-formulations of BCS in different medical applications.
Table 4. Nano-formulations of BCS in different medical applications.
DisorderNano FormulaModelImproved ParametersRef.
Alzheimer’s diseaseBCS oil-
pDNA-chitosan-PLGA nanoparticles
N2a cell- Promoted neurite outgrowth (that is important for the regrowth or repair of nervous tissues or cells[149]
Huntington’s diseaseEncapsulated ThymoquinoneRats- Enhanced the behavioral tests
↓ MDA, Protein carbonyls, and Nitrite
↑ SOD, CAT,
↑ GST, GPx, GR, GSH
↑ Vitamin C and E
↑ SDH
↓ AChE activity
[150]
Diabetic
neuropathy
Silver nanoparticles of aqueous BCS extractRats↓ Glucose level
↑ Serum insulin
↓ Advanced glycation
↓ TNF-α, NF-κB
↓ Serum Aldose reductase
↓ MDA, NO
↑ GSH
↑ Nitrotyrosine
[151]
Anti-bacterialGO-PEG Nanoparticles of BCS ExtractAgar well diffusion for
Staphylococcus aureus,
Escherichia coli
- Destroying the bacteria by:
interfering with the cell wall integrity,
damaging nucleic acid
elevating cell wall permeability.
[152]
Hepato-toxicity,
Renal toxicity
Silver nanoparticles of aqueous BCS extractMice↓ The hepatosomatic index
↓ Serum levels of ALT, AST, ALP, MDA
↑ Total protein
↓ Creatinine
- No change in renal somatic index
[153]
Neuro-endocrine tumorsCopper nanoparticles of BCS aqueous extract) in 2 concentrationsAdrenal phaeochro-mocytoma cells↑ Cell viability
↓ Apoptosis index
↑ Mitochondrial membrane potential
↓ IL1α, IL1β, IL6, TNF-α
↓ Caspase-3
[154]
Chronic Lung InjuryPLGA Nanoparticles with loaded ThymoquinoneRats↓ Serum IL-10
↓ TGF-β1
- Amelioration of histopathological changes and ultrastructure findings
[155]
Pulmonary
Fibrosis
PLGA-PVA Nanoparticles with loaded ThymoquinoneRats↓ Serum IL-10
↓ TGF-β1
↓ iNOS
- Amelioration of histopathological changes and Ultrastructure findings
[156]
PsoriasisEthosomal vesicles loaded with TQRats↑ % Drug activity
↑ % Orthokeratosis
[157]
Breast CancerSilver nanoparticles of aqueous BCS extractMCF-7 cells Ab↓ COX-2
↑ BAX
↓ BCL-2
[158]
↑ increase; ↓ decrease; TNF-α: tumor necrosis factor-alpha; MDA: malondialdehyde; NF-κB: nuclear factor-kappa B; NO: nitric oxide; GSH: reduced glutathione; GST: glutathione S-transferase; GPx: glutathione peroxidase; GR: glutathione reductase; ALT: alanine aminotransferase; AST: aspartate aminotransferase; ALP: alkaline phosphatase; IL-: interleukin; TGF-β1: transforming growth factor-beta 1; iNOS: inducible nitric oxide synthase; SDH: succinate dehydrogenase; AChE: acetylcholinesterase; COX-2: cyclooxygenase-2; BCL-2: B-cell lymphoma 2 protein. BAX: BCL-2-associated X protein; GO: graphene oxide; PEG: polyethylene glycol.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rashwan, H.K.; Mahgoub, S.; Abuelezz, N.Z.; Amin, H.K. Black Cumin Seed (Nigella sativa) in Inflammatory Disorders: Therapeutic Potential and Promising Molecular Mechanisms. Drugs Drug Candidates 2023, 2, 516-537. https://doi.org/10.3390/ddc2020027

AMA Style

Rashwan HK, Mahgoub S, Abuelezz NZ, Amin HK. Black Cumin Seed (Nigella sativa) in Inflammatory Disorders: Therapeutic Potential and Promising Molecular Mechanisms. Drugs and Drug Candidates. 2023; 2(2):516-537. https://doi.org/10.3390/ddc2020027

Chicago/Turabian Style

Rashwan, Hager K., Shahenda Mahgoub, Nermeen Z. Abuelezz, and Hatem K. Amin. 2023. "Black Cumin Seed (Nigella sativa) in Inflammatory Disorders: Therapeutic Potential and Promising Molecular Mechanisms" Drugs and Drug Candidates 2, no. 2: 516-537. https://doi.org/10.3390/ddc2020027

APA Style

Rashwan, H. K., Mahgoub, S., Abuelezz, N. Z., & Amin, H. K. (2023). Black Cumin Seed (Nigella sativa) in Inflammatory Disorders: Therapeutic Potential and Promising Molecular Mechanisms. Drugs and Drug Candidates, 2(2), 516-537. https://doi.org/10.3390/ddc2020027

Article Metrics

Back to TopTop