Skip to main content

Diverse pharmacological actions of potential carbazole derivatives by influencing various pathways of molecular signaling

Abstract

Background

Carbazoles are an important class of heterocyclic aromatic compounds that contain nitrogen atom in the ring. They have a large-conjugated system, attractive “electrical and charge-transport properties”, and the ability to efficiently incorporate different functional groups into the structurally inflexible carbazolyl ring.

Main text

Carbazole derivative ECCA acts as an anticancer agent by reactivating the P53 molecular signaling pathway; similarly, some other derivatives of carbazole show antifungal activity by acting on the RAS-MAPK pathway. Carbazole derivatives also show their effect on inflammation by inhibiting the p38 mitogen-activated protein kinase signaling pathway by stopping the conversion of DAXX protein into ASK-1. By modifying the AKT molecular signaling pathway through boosting protein phosphatase activity in the brain, they show anti-Alzheimer’s activity and also by translocating the GLUT4 these are effective against diabetes.

Conclusion

After exploring the literature on carbazole, it was found that carbazole has an immeasurably great potential for the treatment of various diseases as the carbazole nucleus leads to various synthesized derivatives which are used for their pharmacological activities. So there is a need to explore carbazole for some newer drugs.

Background

Heterocyclic compounds are composed of cyclic rings containing two or more different types of atoms [1, 2]. Throughout nature, heterocyclic compounds are very common and essential. In carbazole, two benzene rings with 6 carbon atoms are fused on either side of a five-membered heterocyclic ring with nitrogen atom showing vast delocalization of electrons [3].

Carbazole and its compounds are useful types of heterocyclic aromatic compounds that show desirable properties of transporting electrons and charges throughout the ring [4]. Also, many polyfunctional groups can be easily added into the carbazolyl ring for the formation of some other novel derivatives [5]. These properties of carbazole-based derivatives result in a wide range of potential applications in the chemistry field (dyes, photoelectrical materials, supramolecular recognition, etc.), and these derivatives also result in various pharmacological activities like antidiabetic [6, 7], antitumor [8,9,10], anticonvulsant [11], antimicrobial [12,13,14], antioxidative [15, 16], antifungal [17, 18] antihistaminic [19], anti-inflammatory [20, 21], antitubercular [22], antiviral [23], carbonic anhydrase enzyme activity [24], neuroprotective activity [25] and antidiarrhoel [26], etc.

figure a

C-3 and C-6 positions in carbazoles have the highest electron density so in recent years the synthesis of some novel carbazoles has been done by modification in carbazole at C-3 and C-6, and 9th positions of N [27]. If there is no steric hindrance that occurs, the carbazoles along with unblocked 3rd and 6th positions of Carbon in the ring form at least a dimer upon oxidation. Carbazoles undergo electrophilic substitution reaction like sulfonation, nitration, Friedel–Crafts acylation and Friedel–Crafts alkylation forming different derivatives of carbazole. The problem with this substitution is that a mixture of ortho and para positions is obtained but due to high electron density at the 3rd and 6th carbon positions, 75% of the para product formed concerning C-1 and C-8 (ortho position). Formation of mono along with di-, tri-, and tetra-substituted carbazoles is possible due to the reactivity of carbon atoms to the imine group in carbazole. By using appropriate alkylating or acylating agents at room temperature, the N-9 substituted carbazole derivatives are also formed [4].

Main text

Diseases with neurodegeneration

Neurodegenerative diseases (NDs) are a group of disorders (multiple sclerosis, Parkinson's disease, Alzheimer’s disease, Huntington’s disease, amyotrophic lateral sclerosis, etc.) that cause loss of neuron’s structure or function [28,29,30,31,32,33]. As a result of these changes in the brain anatomy, the logical or functional decline may result in protein misfolding, diminished mitochondrial functions, oxidative stress, impaired neuro-transmission, and loss of neurons which are just a few physiological indicators of NDs [34, 35].

Anticancer potential

Cancer is the major health problem in individuals. Every year approx 18 million peoples are diagnosed with cancer, and it is the prime cause of death worldwide. In developed countries, the age-standardized rate for diagnosis is 296 cases per 1000,000 peoples [36,37,38,39,40]. Cancer is a pathological condition of the body which is due to various genetic mutations which are characterized by uncontrolled proliferation and growth of cells [41,42,43,44]. Every cell of the body can become a cancer cell when a genetic mutation occurs [45]. Carcinogenesis process occurs in three stages; in the first stage alteration has already been identified so it is called the initiation stage, in the second promotion stage these altered cells are mutated and form malignant cells, and in the last stage of carcinogenesis which is continuation stage once the tumor has increased in size and cancer cells begin to split in an accelerated irreversible mode. And in the last stage the cells instead of growing locally start to disseminate to other body parts (metastasis) [46,47,48].

In human melanoma cells, the p53 pathway is reactivated by the anticancer effect of carbazole derivative ECCA, (9-ethyl-9H-carbazole-3-carbaldehyde). In melanoma therapy, about 84% of human melanomas harbor wild-type p53 is thought to be a supreme target for treating melanoma. Carbazole derivatives enhanced the phosphorylation of c-Jun N-terminal kinase (JNK) and p38-MAPK, and either a p38 mitogen-activated protein kinase or c-Jun N-terminal kinase inhibitor reassured cell proliferation inhibition produced by 9-ethyl-9-H-carbazole-3-carbaldehyde (ECCA), whose expression was dependent on p53 gene. Carbazole derivatives selectively and significantly depress the expansion of melanoma cells by persuading the programmed cell death of melanoma cells and senescence through p53 activation [49].

figure b

Yonghua et al. designated water-soluble carbazole and sulfonamide derivatives which are tested in vivo against human HepG2 (hepatoblastoma liver cancer cell line) xenograft mouse tumor expansion and found that compound 4c (Sodium 6-(N-(2,6-dimethoxypyridin-3-yl)sulfamoyl)-9-methyl-9H-carbazol-2-yl phosphate) is the most effective tumor inhibitor [50,51,52].

figure c

Huang shows the in vitro cytotoxic activity of different synthesized carbazole derivatives in which compound 7 g (N′-(Benzo[d](1,3) dioxol-5-ylmethylene)-1-methyl-9H-carbazole-2-carbohydrazide) and compound 7p (N′-(4-Chlorobenzylidene)-1,9-dimethyl-9H-carbazole-2-carbohydrazide) is the potent inhibitor of cancer cells but do not affect normal cells. 7g Carbazole derivative with acylhydrazone substituted with 1,3-benzodiooxozole displayed significant selective proliferation inhibition activity in vitro with (IC50 < 12.24 μM). For the in vitro investigation of cytotoxic activity three cell lines, human melanoma (A875), human hepatocellular liver carcinoma (HepG2), and a subclone of the African green monkey cell line (MARC145) were used [53].

figure d
figure e

Capan, I et al. synthesized a sequence of carbazole derivatives. Result among all the synthesized derivatives showed that compound 9 [(E)-2-(9H-carbazol-9-yl)-N′-(3-(4-chlorophenyl)-4-oxothiazolidin-2-ylidene)acetohydrazide)] and compound 10 [5-((9H-carbazol-9-yl)methyl)-1,3,4-oxadiazol-2(3H)-one] are found most potent anticancer agent against HepG2, HeLa, and MCF-7 cancer cell lines with IC50 values of 7.68, 10.09 μM. Against HeLa, cancer cell lines found that compound 9 is the best antiproliferative agent [54].

figure f
figure g

Leu et al. synthesized a novel series of carbazole derivatives that are synthesized by imidazole derivatives along with N-alkyl bromide substituted carbazole. Compound 61[1-(5-(9H-carbazol-9-yl)pentyl)-3-(2-bromobenzyl)-5,6-dimethyl-1H-benzo[d]imidazol-3-ium bromide], which was bearing a 2-bromobenzyl substituent at position-3 of the 5,6-dimethyl-benzimidazole, showed powerful inhibitory activities and found to be more selective to human promyelocytic leukemia cell line (HL-60), human hepatocellular carcinoma cell line (SMMC-7721), breast cancer cell line (MCF-7) and human colon adenocarcinoma (SW480) cell lines with IC50 values 0.51–2.48 μM. The results of the research indicated that the presence of 5,6-dimethyl-benzimidazole ring and substitution of the imidazolyl-3-position with a 2-bromobenzyl group, as well as for the antitumor activity the length of alkyl chain between carbazole and imidazole ring were important [55] (Fig. 1).

Fig. 1
figure 1

Anticancer mechanism of carbazole derivatives. 9-Ethyl-9-H-carbazole-3-carbaldehyde (ECCA) re-activates P53 pathway and causes phosphorylation of P38MAPK

figure h

Breast and uterine cancer

The natural and synthetic carbazole derivatives represent an exciting heterocycle class that has shown several pharmaceutical properties and is also an excellent antitumor tool in preclinical experiments. The antibodies target several cellular key points, such as Topoisomerases I, II and DNA. Several efforts have been made to plan and synthesize some novel carbazole derivatives that have lesser side effects and good potency. The carbazole derivatives without affecting non-tumor cell lines possess antiproliferative activities against uterine and breast cancer cell lines. Carbazole derivatives through the inhibition of Topoisomerase II trigger cancer cells' intrinsic apoptotic pathway.

Antifungal potential

Fungal infections (mycosis) are increasing throughout the world. Although there are several reasons for this higher increase in mycosis, immune modulation of the host is one of the foremost risk factors for intrusive mycosis [56]. Humans suffer from most of the fungal infections due to the setting of iatrogenic immunosuppression. When these factors are absent fungi cause mild, self-limited mucocutaneous surface infections. When this infection develops in a normal host, it causes adaptive immune dysfunction because of genetic defects [56,57,58].

Tang et al. tested the antifungal activity of all the synthesized compounds against different fungal strains and found that compound 3f (2-(9H-carbazol-9-yl)-N-(4,5-dihydro-5-(thiophen-2-yl)-1,3,4-thiadiazol-2-yl)acetamide) and 3i (2-(1-chloro-9H-carbazol-9-yl)-N-(4,5-dihydro-5-p-tolyl-1,3,4-thiadiazol-2-yl)acetamide) exhibited the most potent inhibitory activity against C. wilt having inhibition rates 72.40% and 67.65% [59].

figure i
figure j

Shaikh et al. synthesized two series of carbazole analogues. The first one is 8-methoxy-N-substituted-9H-carbazole-3-carboxamides, and the second one is carbazolyl-substituted rhodanines. The final compounds were prepared from these two series and were tested for antibacterial activity against Bacillus subtilis, Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli [60,61,62] and antifungal activity against Cryptococcus tropicalis, Aspergillus niger, Cryptococcus neoformans, and Candida albicans [63,64,65]. Various substituents, nitrogen-containing heterocyclic systems like pyrimidinyl and piperidinyl on carbazole nucleus exerted a significant antifungal activity in the first series of compounds. 2-Methyl piperidinyl carboxamide derivative compound 6f (8-Methoxy-9H-carbazol-3-yl)(2-methylpiperidin-1-yl)methanone found as potent antifungal and antibacterial agent. In second series substitution at 3 positions of rhodanines which is conjugated to -methyl carbazole through an acrylidine linkage, the result showed the effect of bioisosteres coumarin present in compound 15i [(5Z)-3-(4-methyl-2-oxo-2H-chromen-7-yl)-5-((9-methyl-9H-carbazol-6-yl)methylene)-2-thioxothiazolidin-4-one]were shown potent inhibitory activity. It was concluded that electron donating groups such as hydroxy, alkoxy, and alkyl groups on the aromatic ring greatly contributed toward the antifungal and antibacterial activity [64].

figure k
figure l

The morphogenic process came up with the virulency of Candida albicans, an opportunistic human fungal pathogen. Ras1-MAPK pathways are crucial to Candida albicans virulence through controlling morphogenesis, cell growth, and biofilm formation.

Several carbazole-containing drugs have been prescribed for infections of Candida albicans, leading to the expansion of drug-resistant strains. For that reason, it is needed to properly treat fungal infections, and new compounds must be developed. By suppressing Ras/MAPK-related gene protein and RNA levels, carbazole derivatives prevent morphogenesis [66, 67] (Fig. 2).

Fig. 2
figure 2

Antifungal mechanism of carbazole derivatives. N-(4-methoxybenzyl)-3(9H-carbazol-9yl) propan 1-amine inhibits morphogenesis by suppressing protein and RNA levels of Ras/MAPK-related genes

Anti-psoriasis potential

Psoriasis is a chronic autoimmune disease that is associated with systemic manifestation and characterized by keratinocytes and hyperproliferation mediated by T-cells [68,69,70]. It is an autoimmune disease with a genetic predisposition. In the psoriatic lesions presence of dendritic cells, cytokines and T lymphocytes has prompted the development of biologic therapies [71, 72].

Carbazole present in coal tar is an active antiangiogenic compound along with that antiangiogenic effect carbazole also withdraws the formation of inflammatory IL-15 by human mononuclear cells. It is believed that IL-15, which is increased in psoriasis, contributes to the inflammation of psoriasis [29, 73]. Moreover, carbazole treatment reduced the activity of nitric oxide synthase (iNOS) (a pro-inflammatory enzyme), which is elevated in psoriasis patients. In a study on human psoriasis, carbazole was found to inhibit the activator of stat 3-mediated transcription, which is relevant to psoriasis. iNOS, IL-15, and stat3 activation rely on the small GTPase Rac for optimum activity. As a mechanism for inhibiting downstream inflammatory and angiogenic pathways, carbazole inhibits Rac activation [74]. The mechanism of action of carbazole could involve the inhibition of pro-inflammatory cytokine synthesis, inhibition of activator protein-1 (AP-1) activity, and inhibition of EGFR activation. Production of IL-15 by mononuclear cells is also inhibited by carbazole, a key cytokine in psoriasis [75,76,77] (Fig. 3).

Fig. 3
figure 3

Anti-psoriasis mechanism of carbazole derivatives. Carbazole present in coal tar inhibits the P38 signaling pathway by inactivation of Rac

Anti-Alzheimer potential

Generally, dementia is caused by Alzheimer’s disease, the primary or cellular phase of Alzheimer's disease happens in parallel with accumulating amyloid β, inducing the spread of tau pathology [78]. Pathological characteristics in Alzheimer's disease (AD) are deposition of β-amyloid (Aβ) peptide and hyperphosphorylated tau in the brain, carbazole derivative (9c) N(6-(9H-carbazol-9-yl)-hexyl)-1benzylpiperidin-4-amine modulated AKT pathway and boosting protein phosphatase 2A activity in the brain. The compound 9c with (IC50 = 26.5 µM for AChE and IC50 = 0.18 µM for BuChE) results in the highest inhibitory activity against butyrylcholinesterase and acetylcholinesterase. Structure–activity relationship suggested that attachment to the 4-amino-N-benzylpiperidine fragment with C5 and C6 alkyl linkers in compound 9c results in the most potent compound among all [79].

figure m

Choubdar et al. designated several carbazole derivatives by using some heterocyclic nucleus like quinoline, pyridine, piperidine, benzyl piperidine, and benzyl piperazine, etc., in which compound 3s (9-(5-(quinoline-1 (2H)-yl)pentyl)-9H-carbazole) linked with quinoline found as the potent drug against BuChE and AChE and also showed the inhibitory activity for AChE induced β-amyloid (Aβ) aggregation. The most potent compound 3s with IC50 = 0.11 µM and 0.02 µM showed the best activity against AChE and BuChE, respectively. The presence of quinoline moiety in compound 3s more actively inhibited Aβ self-aggregation. Docking studies suggested that compound 3s binds effectively to the PAS and anionic binding site of the enzyme with the assistance of π stacking and hydrophobic interactions [80].

figure n

Carbazole derivatives regulating Ca2+/CaMKII/CREB signaling pathway. The brain contains CaMKII (Calcium/Calmodulin-dependent Protein Kinase II) which is a specific protein kinase of serine/threonine, which is regulated by Ca2+/calmodulin [81].

Shaikh et al. synthesized a series of α-amino phosphonate-based carbazole derivatives that were in silico and in vitro tested for their cholinesterase activity. All the compounds present better AchE activity [0.475–7.781 μM] than BuChE (3.306–21.32 μM). Against AChE as well as BuChE compound 4j [Diethyl(9-ethyl-9H-carbazol-3-yl)amino) (3nhydroxyphenyl)methyl)phosphonate] was the most potent derivative with IC50¼0.475 ± 0.12 mM and IC50¼3.306 ± 0.21 μM, respectively [82] (Fig. 4).

Fig. 4
figure 4

Anti-Alzheimer mechanism of carbazole derivatives. AKT pathway regulation by carbazole derivative N-(6-(9-H-carbazol-9yl) hexyl)-1-benzylpiperidine-4-amine by boosting protein phosphatase activity in brain

figure o

Anti-inflammatory potential

Inflammation is a result of a complex biological response that includes inflammatory mediators, sensors, inflammation-inducing factors, and affected target tissues [83, 84]. Physical injuries, toxic chemicals, viruses, fungi, and bacteria are some harmful stimuli under inflammation-inducing factors. Pathogen-associated molecular patterns (PAMs) triggered the inflammatory response [85, 86]. Inflammatory pathways are mediated by some inflammatory mediators like cyclooxygenase (COX), chemokines, vasoactive amines nitric oxide (NO), etc. [87]. The inflammatory responses finally lead to symptoms like heat, pain, redness, and swelling, etc. [88, 89].

Bandgar et al. designated a novel series of carbazole(5a–o) which was tested against the anti-inflammatory activity. Compound 5c via hydrophobic interactions potently binds at the site of COX-II. The oxygen atom of the methoxy group on compound 5c formed two hydrogen bonds with the Asp B:225 and Nag C:671 and the nitrogen atom present in the pyrazoline ring on compound 5c formed two hydrogen bonds with the Leu A:131 [90].

figure p

Carbazole derivatives inhibit the lipopolysaccharide-induced inflammatory mediator production in macrophages via suppression of p38 MAPK (mitogen-activated protein kinase signaling pathway). Carbazole derivatives inhibit the formation of TNF-α (tumor necrosis factor α), PGE2 (prostaglandin E2), and nitric oxide (NO) induced by Lipopolysaccharide (LPS) [91,92,93] (Fig. 5).

Fig. 5
figure 5

Anti-inflammatory mechanism of carbazole derivatives. 3-(3-(2-Methoxy-5-methylphenoxy)-4 5-dihydro-1H pyrazole-5yl)-9H-Carbazole inhibits the p38 MAPK signaling pathway by stopping the conversion of DAXX protein into ASK-1

Antidiabetic potential

Diabetes is a condition in which due to autoimmune destruction pancreatic β cells slow down the production of insulin results hyperglycemia due to excess availability of glucose [94,95,96,97,98]. α-glucosidase inhibitory activity of compound 7k (1-(5,6-di(furan-2-yl)-1,2,4-triazin-3-ylthio)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol)was found most effective and the result revealed that compound7k is the potent compound with h IC50 values of 4.27 ± 0.07 μM among all the synthesized compound by Wang et al. The potent compound with high-density van der Waals contact binds at the bottom of the α-glucosidase pocket, whereas near the entrance of the pocket, furan ring was positioned which made only a few contacts. Detailed analysis showed that the carbazole ring of the potent compound formed arene-cation interactions with Arg-439 and Arg-312 residues, respectively. Furan ring of a potent compound located at the hydrophobic pocket, surrounded by Leu-176, Phe-177, Phe-157, and Pro-240 residues. All these interactions helped 7k to anchor in the binding site of α-glucosidase [99].

figure q

Carbazole derivative koenidine translocates the GLUT 4 which is mediated through the AKT-dependent signaling pathway (4) in L6-GLUT-4 myc myotubes [100].

figure r

The in vitro testing of separated carbazole alkaloids on uptake of glucose and translocation of GLUT 4 in L6-GLUT-4 myc myotubes was done on streptozotocin-induced diabetic rats for their activities. Therefore, the in vitro study of the koenidine suggested that koenidine, a carbazole derivative, possesses a promising antidiabetic activity through managing diabetes and insulin resistance [6, 101] (Fig. 6).

Fig. 6
figure 6

Antidiabetic mechanism of carbazole derivatives. Koenidine 4 translocates GLUT 4 for antidiabetic activity

Conclusion

Carbazole moiety itself is responsible for various types of pharmacological activities; due to high electron delocalization, carbazole shows better physicochemical properties. Derivatives having carbazole pharmacophore are responsible for various therapeutic activities like anti-Alzheimer, antioxidant, antidiabetic, anticancer, anticonvulsant, antimicrobial, and anti-inflammatory, etc., by specifically acting on potential molecular level proteins and factors such as RAS-MAPK, DAXX, ASK-1, AKT, and JNK, either by inhibiting or by activating them by de-phosphorylation or phosphorylation (Fig. 7). Due to all these important activities, the carbazole nucleus has attracted the attention of researchers in the discovery of other novel derivatives of carbazole.

Fig. 7
figure 7

Molecular mechanisms of carbazole derivatives for different pharmacological activities

figure s

Complete SAR of carbazole derivatives

Availability of data and materials

Present article is a review article, and the authors have not used any material and data.

Abbreviations

GLUT 4:

Glucose transporter type 4

ASK-1:

Apoptosis signal-regulating kinase 1

ECCA:

9-Ethyl-9H-carbazole-3-carbaldehyde

RAS-MAPK:

Ras/mitogen-activated protein kinase

DAXX:

Death-domain-associated protein

AKT:

Protein kinase B

JNK:

Jun N-terminal kinase

MCF-7:

Michigan Cancer Foundation

DNA:

Deoxyribonucleic acid

RNA:

Ribonucleic acid

iNOS:

Inducible nitric oxide synthase

IL-5:

Interleukin-5

EGFR:

Epidermal growth factor receptor

AChE:

Acetylcholinesterase

BuChE:

Butyrylcholinesterase

A875:

Human melanoma

HepG2:

Human hepatocellular liver carcinoma

MARC145:

A subclone of the African green monkey cell line

HL-60:

Human promyelocytic leukemia cell line

SMMC-7721:

Human hepatocellular carcinoma cell line

MCF-7:

Breast cancer cell line

SW480:

Human colon adenocarcinoma cell lines

References

  1. Al-Mulla A (2017) A review: biological importance of heterocyclic compounds. Pharm Chem 9(13):141–147

    CAS  Google Scholar 

  2. Saini MS, Kumar A, Dwivedi J, Singh R (2013) A review: biological significances of heterocyclic compounds. Int J Pharm Sci Res 4(3):66–77

    CAS  Google Scholar 

  3. Ziarani GM, Moradi R, Lashgari N, Kruger HG (2018) Chapter 6—Carbazole dyes. In: Ziarani GM, Moradi R, Lashgari N, Kruger HG (eds) Metal-free synthetic organic dyes. Elsevier, Amsterdam, pp 109–116

    Chapter  Google Scholar 

  4. Campbell N, Barclay BM (1947) Recent advances in the chemistry of carbazole. Chem Rev 40(3):359–380

    Article  CAS  PubMed  Google Scholar 

  5. Karon K, Lapkowski MJ (2015) Carbazole electrochemistry: a short review. J Solid State Electrochem 19:2601–2610

    Article  CAS  Google Scholar 

  6. Eseyin OA, Edem E, Johnson E, Ahmad A, Afzal S (2018) Synthesis and in vitro antidiabetic activity of some alkyl carbazole compounds. Trop J Pharm Res 17(3):537–541

    Article  CAS  Google Scholar 

  7. Mitra A, Mahadevappa M (2010) Antidiabetic and hypolipidemic effects of mahanimbine (carbazole alkaloid) from Murraya koenigii (rutaceae) leaves. Int J Phytomed 2:22–30

    Google Scholar 

  8. Chen Z, Yang T, Wang W, Yao J, Han S, Tao Y et al (2018) Synthesis and biological evaluation of carbazole aminoalcohols as antitumor agents. ChemistrySelect 3(44):12630–12638

    Article  CAS  Google Scholar 

  9. Issa S, Prandina A, Bedel N, Rongved P, Yous S, Le Borgne M et al (2019) Carbazole scaffolds in cancer therapy: a review from 2012 to 2018. J Enzyme Inhib Med Chem 34(1):1321–1346

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bondock S, Alqahtani S, Fouda AM (2019) Convenient synthesis and antitumor evaluation of some new 9-ethyl-3-(hetaryl) carbazoles. Synth Commun 49(17):2188–2202

    Article  CAS  Google Scholar 

  11. Emmanuel BD, Beevi J, Dharan SS (2020) A concise review on carbazole derivatives and its biological activities. J Pharm Sci Res 12(10):1271–1277

    Google Scholar 

  12. Ding Y-Y, Zhou H, Zhang B-Q, Zhang Z-J, Wang G-H, Zhang S-Y et al (2023) Antimicrobial activity of natural and semi-synthetic carbazole alkaloids. Eur J Med Chem 259:115627

    Article  CAS  PubMed  Google Scholar 

  13. Al-Sultani KTJA (2010) Synthesis and evaluation of the biological activity for some of carbazole derivatives. Al-Nahrain J Sci 13(4):31–38

    Google Scholar 

  14. Xue Y-J, Li M-Y, Jin X-J, Zheng C-J, Piao H-R (2021) Design, synthesis and evaluation of carbazole derivatives as potential antimicrobial agents. J Enzyme Inhib Med Chem 36(1):296–307

    Article  PubMed Central  Google Scholar 

  15. Serdaroğlu G, Uludağ N, Ercag E, Sugumar P, Rajkumar PJ (2021) Carbazole derivatives: synthesis, spectroscopic characterization, antioxidant activity, molecular docking study, and the quantum chemical calculations. J Mol Liq 330:115651

    Article  Google Scholar 

  16. Hieda Y, Anraku M, Choshi T, Tomida H, Fujioka H, Hatae N et al (2014) Antioxidant effects of the highly-substituted carbazole alkaloids and their related carbazoles. Bioorg Med Chem Lett 24(15):3530–3533

    Article  CAS  PubMed  Google Scholar 

  17. Thevissen K, Marchand A, Chaltin P, Meert EM, Cammue B (2009) Antifungal carbazoles. Curr Med Chem 16(17):2205–2211

    Article  CAS  PubMed  Google Scholar 

  18. Zhu S-P, Wang W-Y, Fang K, Li Z-G, Dong G-Q, Miao Z-Y et al (2014) Design, synthesis and antifungal activity of carbazole derivatives. Chin Chem Lett 25(2):229–233

    Article  CAS  Google Scholar 

  19. Aftrid ZHVI, Cahyana AH (eds) (2022) Synthesis of carbazole derivative compound with the Mannich reaction and antioxidant activity. AIP conference proceedings. AIP Publishing

  20. Nandy BC, Gupta A, Mittal A, Vyas VJ (2014) Carbazole: it’s biological activity. J Biomed Pharm Res 3(1):42–8

    CAS  Google Scholar 

  21. Głuszyńska A (2015) Biological potential of carbazole derivatives. Eur J Med Chem 94:405–426

    Article  PubMed  Google Scholar 

  22. Sellamuthu S, Gutti G, Kumar D, Kumar Singh S (2018) Carbazole: a potent scaffold for antitubercular drugs. Mini Rev Org Chem 15(6):498–507

    Article  CAS  Google Scholar 

  23. Caruso A, Ceramella J, Iacopetta D, Saturnino C, Mauro MV, Bruno R et al (2019) Carbazole derivatives as antiviral agents: an overview. Molecules 24(10):1912

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rifati-Nixha A, Arslan M, Gencer N, Çıkrıkıçı K, Gökçe B, Arslan O et al (2019) Synthesis of carbazole bearing pyridopyrimidine-substituted sulfonamide derivatives and studies their carbonic anhydrase enzyme activity. J Biochem Mol Toxicol 33(6):e22306

    Article  PubMed  Google Scholar 

  25. Liu Y-P, Guo J-M, Liu Y-Y, Hu S, Yan G, Qiang L et al (2019) Carbazole alkaloids with potential neuroprotective activities from the fruits of Clausena lansium. J Agric Food Chem 67(20):5764–5771

    Article  CAS  PubMed  Google Scholar 

  26. Mandal S, Nayak A, Kar M, Banerjee SK, Das A, Upadhyay S et al (2010) Antidiarrhoeal activity of carbazole alkaloids from Murraya koenigii Spreng (Rutaceae) seeds. Fitoterapia 81(1):72–74

    Article  CAS  PubMed  Google Scholar 

  27. Luthra PM, Kumar N (2021) Progress and development of C-3, C-6, and N-9 positions substituted carbazole integrated molecular hybrid molecules as potential anticancer agents. Mini Rev Med Chem 21(19):2929–2956

    Article  CAS  PubMed  Google Scholar 

  28. Liu Q, Xie F, Alvarado-Diaz A, Smith MA, Moreira PI, Zhu X et al (2011) Neurofilamentopathy in neurodegenerative diseases. Open Neurol J 5:58

    Article  PubMed  PubMed Central  Google Scholar 

  29. Martins AM, Ascenso A, Ribeiro HM, Marto J (2020) Current and future therapies for psoriasis with a focus on serotonergic drugs. Mol Neurobiol 57(5):2391–2419

    Article  CAS  PubMed  Google Scholar 

  30. Mandemakers W, Morais VA, De Strooper B (2007) A cell biological perspective on mitochondrial dysfunction in Parkinson disease and other neurodegenerative diseases. J Cell Sci 120(10):1707–1716

    Article  CAS  PubMed  Google Scholar 

  31. Lamptey RN, Chaulagain B, Trivedi R, Gothwal A, Layek B, Singh J (2022) A review of the common neurodegenerative disorders: current therapeutic approaches and the potential role of nanotherapeutics. Int J Mol Sci 23(3):1851

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Erekat NS (2022) Apoptosis and its therapeutic implications in neurodegenerative diseases. Clin Anat 35(1):65–78

    Article  PubMed  Google Scholar 

  33. Margiotta A (2021) Role of SNAREs in neurodegenerative diseases. Cells 10(5):991

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Voet S, Srinivasan S, Lamkanfi M, van Loo G (2019) Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol Med 11(6):e10248

    Article  PubMed  PubMed Central  Google Scholar 

  35. Dugger BN, Dickson DW (2017) Pathology of neurodegenerative diseases. Cold Spring Harbor Perspect Biol 9(7):a028035

    Article  Google Scholar 

  36. Zhang C-L, Huang T, Wu B-L, He W-X, Liu D (2017) Stem cells in cancer therapy: opportunities and challenges. Oncotarget 8(43):75756

    Article  PubMed  PubMed Central  Google Scholar 

  37. Ayob AZ, Ramasamy TS (2018) Cancer stem cells as key drivers of tumour progression. J Biomed Sci 25:1–18

    Article  Google Scholar 

  38. Hesketh R (2023) Introduction to cancer biology. Cambridge University Press, Cambridge

    Book  Google Scholar 

  39. Siegel RL, Miller KD, Wagle NS, Jemal AJ (2023) Cancer statistics, 2023. CA Cancer J Clin 73(1):17–48

    Article  PubMed  Google Scholar 

  40. Islami F, Guerra CE, Minihan A, Yabroff KR, Fedewa SA, Sloan K et al (2021) American Cancer Society’s report on the status of cancer disparities in the United States. CA Cancer J Clin 72(2):112–143

    Article  PubMed  Google Scholar 

  41. Choudhuri S, Chanderbhan R, Mattia A (2018) Chapter 20-Carcinogenesis: mechanisms and models*. In: Veterinary toxicology, vol 3, p 354

  42. Hashem S, Ali TA, Akhtar S, Nisar S, Sageena G, Ali S et al (2022) Targeting cancer signaling pathways by natural products: exploring promising anti-cancer agents. Biomed Pharmacother 150:113054

    Article  CAS  PubMed  Google Scholar 

  43. Yadav AR, Mohite SK (2020) Cancer-a silent killer: an overview. Asian J Pharm Res 10(3):213–216

    Article  Google Scholar 

  44. Alzahrani SM, Al Doghaither HA, Al-Ghafari AB (2021) General insight into cancer: An overview of colorectal cancer. Mol Clin Oncol 15(6):1–8

    Article  Google Scholar 

  45. Rahman MM, Sarker MT, Alam Tumpa MA, Yamin M, Islam T, Park MN et al (2022) Exploring the recent trends in perturbing the cellular signaling pathways in cancer by natural products. Front Pharmacol 13:950109

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W et al (2022) Natural products as anticancer agents: Current status and future perspectives. Molecules 27(23):8367

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Xiong S, Dong L, Cheng L (2021) Neutrophils in cancer carcinogenesis and metastasis. J Hematol Oncol 14(1):1–17

    Article  Google Scholar 

  48. Hatta MNA, Mohamad Hanif EA, Chin S-F, Neoh H (2021) Pathogens and carcinogenesis: a review. Biology 10(6):533

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wen J, Chen W, Zhao B, Xu Q, Liu C, Zhang Q et al (2021) A carbazole compound, 9-ethyl-9H-carbazole-3-carbaldehyde, plays an antitumor function through reactivation of the p53 pathway in human melanoma cells. Cell Death Dis 12(6):591

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu Y, Wu Y, Gu Y, Hu L (2020) Synthesis and structure-activity relationship study of water-soluble carbazole sulfonamide derivatives as new anticancer agents. Eur J Med Chem 191:112181

    Article  CAS  PubMed  Google Scholar 

  51. Lin W, Wang Y, Lin S, Li C, Zhou C, Wang S et al (2012) Induction of cell cycle arrest by the carbazole alkaloid Clauszoline-I from Clausena vestita DD Tao via inhibition of the PKCδ phosphorylation. Eur J Med Chem 47:214–220

    Article  CAS  PubMed  Google Scholar 

  52. Wang G, Sun S, Guo H (2022) Current status of carbazole hybrids as anticancer agents. Eur J Med Chem 229:113999

    Article  CAS  PubMed  Google Scholar 

  53. Huang W, Gao Z, Zhang Z, Fang W, Wang Z, Wan Z et al (2021) Selective and effective anticancer agents: Synthesis, biological evaluation and structure–activity relationships of novel carbazole derivatives. Bioorgan Chem 113:104991

    Article  CAS  Google Scholar 

  54. Çapan İ, Hawash M, Jaradat N, Sert Y, Servi R, Koca İ (2023) Design, synthesis, molecular docking and biological evaluation of new carbazole derivatives as anticancer, and antioxidant agents. BMC Chem 17(1):1–17

    Article  Google Scholar 

  55. Liu L-X, Wang X-Q, Zhou B, Yang L-J, Li Y, Zhang H-B et al (2015) Synthesis and antitumor activity of novel N-substituted carbazole imidazolium salt derivatives. Sci Rep 5(1):13101

    Article  PubMed  PubMed Central  Google Scholar 

  56. Lockhart SR, Guarner J (2019) Emerging and reemerging fungal infections. Semin Diagn Pathol 36(3):177–181

    Article  PubMed  Google Scholar 

  57. Lionakis MS (2012) Genetic susceptibility to fungal infections in humans. Curr Fung Infect Rep 6:11–22

    Article  Google Scholar 

  58. Kainz K, Bauer MA, Madeo F, Carmona-Gutierrez D (2020) Fungal infections in humans: the silent crisis. Microb Cell 7(6):143

    Article  PubMed  PubMed Central  Google Scholar 

  59. Tang C, Chen X, Yang S, Guo W, Yang X, Li P et al (2023) Discovery of novel carbazole derivatives containing a 1, 3, 4-thiadiazole moiety as antifungal candidates. Phosphorus Sulfur Silicon Relat Elem 198:1–5

    Article  Google Scholar 

  60. Kaplancikli ZA, Yurttaş L, Turan-Zitouni G, Özdemir A, Özic R, Ulusoylar-Yıldırım Ş et al (2012) Synthesis, antimicrobial activity and cytotoxicity of some new carbazole derivatives. Arab J Chem 27(6):868–874

    CAS  Google Scholar 

  61. Salih N, Salimon J, Yousif E (2016) Synthesis and antimicrobial activities of 9H-carbazole derivatives. Arab J Chem 9:S781–S786

    Article  CAS  Google Scholar 

  62. Ruan B, Tian Y, Zhou H, Wu J, Liu Z, Zhu C et al (2009) Synthesis, crystal structure and in vitro antibacterial activity of two novel silver (I) complexes. J Organometall Chem 694(18):2883–2887

    Article  CAS  Google Scholar 

  63. Cruz KS, Lima ES, Silva MJA, Souza ES, Montoia A, Pohlit AM et al (2019) Screening and antifungal activity of a β-carboline derivative against Cryptococcus neoformans and C. gattii. Int J Microbiol 2019:7157845

    Article  PubMed  PubMed Central  Google Scholar 

  64. Shaikh MS, Chandrasekaran B, Palkar MB, Kanhed AM, Kajee A, Mlisana KP et al (2020) Synthesis and biological evaluation of novel carbazole hybrids as promising antimicrobial agents. Chem Biodivers 17(5):e1900550

    Article  CAS  PubMed  Google Scholar 

  65. Afeltra J, Verweij P (2003) Antifungal activity of nonantifungal drugs. Eur J Clin Microbiol Infect Dis 22:397–407

    Article  CAS  PubMed  Google Scholar 

  66. Park Y-K, Shin J, Lee H-Y, Kim H-D, Kim J (2021) Development of carbazole derivatives compounds against Candida albicans: candidates to prevent hyphal formation via the Ras1-MAPK pathway. J Fungi 7(9):688

    Article  CAS  Google Scholar 

  67. Park YK, Shin J, Lee H-Y, Kim HD, Kim J (2021) Inhibition of Ras1-MAPK pathways for hypha formation by novel drug candidates in Candida albicans. 2021.07. 06.451239.

  68. Gudjonsson JE, Johnston A, Sigmundsdottir H, Valdimarsson H (2003) Immunopathogenic mechanisms in psoriasis. Clin Exp Immunol 135(1):1–8

    Article  Google Scholar 

  69. Wu M, Dai C, Zeng F (2023) Cellular mechanisms of psoriasis pathogenesis: a systemic review. Clin Cosmet Investig Dermatol 2023:2503–2515

    Article  Google Scholar 

  70. Türkben H, Ayaz F (2023) Different types, pathogenesis and cytokine network of Psoriasis. Adv Eng Days 7:9–11

    Google Scholar 

  71. Rendon A, Schäkel K (2019) Psoriasis pathogenesis and treatment. Int J Mol Sci 20(6):1475

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Li L, Liu P, Chen C, Yan B, Chen X, Li J et al (2022) Advancements in the characterization of tissue resident memory T cells in skin disease. Clin Immunol 245:109183

    Article  CAS  PubMed  Google Scholar 

  73. Dinesh D, Nanjappa DP, Babu N, Kalladka K, Chakraborty G, Chakraborty AJ et al (2020) Evaluation of toxicity and antiangiogenic activity of Murraya koenigii leaf extracts in zebrafish. J Health Allied Sci 10(02):79–85

    Article  Google Scholar 

  74. Xin Y, Roh K, Cho E, Park D, Whang W, Jung E (2021) Isookanin inhibits pge2-mediated angiogenesis by inducing cell arrest through inhibiting the phosphorylation of ERK1/2 and creb in hmec-1 cells. Int J Mol Sci 22(12):6466

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Arbiser JL, Govindarajan B, Battle TE, Lynch R, Frank DA, Ushio-Fukai M et al (2006) Carbazole Is a naturally occurring inhibitor of angiogenesis and inflammation isolated from antipsoriatic coal tar. J Investig Dermatol 126(6):1396–1402

    Article  CAS  PubMed  Google Scholar 

  76. Caruso A, Barbarossa A, Carocci A, Salzano G, Sinicropi MS, Saturnino C (2021) Carbazole derivatives as STAT inhibitors: an overview. Appl Sci 11(13):6192

    Article  CAS  Google Scholar 

  77. Ávalos-Viveros M, Esquivel-García R, García-Pérez M, Torres-García E, Bartolomé-Camacho MC, Santes V et al (2023) Updated view of tars for psoriasis: what have we learned over the last decade? Int J Dermatol 62(3):290–301

    Article  PubMed  Google Scholar 

  78. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE et al (2021) Alzheimer’s disease. Lancet 397(10284):1577–1590

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Sadeghian B, Sakhteman A, Faghih Z, Nadri H, Edraki N, Iraji A et al (2020) Design, synthesis and biological activity evaluation of novel carbazole-benzylpiperidine hybrids as potential anti Alzheimer agents. J Mol Struct 1221:128793

    Article  CAS  Google Scholar 

  80. Choubdar N, Golshani M, Jalili-Baleh L, Nadri H, Küçükkilinç TT, Ayazgök B et al (2019) New classes of carbazoles as potential multi-functional anti-Alzheimer’s agents. Bioorg Chem 91:103164

    Article  CAS  PubMed  Google Scholar 

  81. Chen C, Xu D, Zhang Z-H, Jia S-Z, Cao X-C, Chen Y-B et al (2020) Cognitive improvement and synaptic deficit attenuation by a multifunctional carbazole-based cyanine in AD mice model through regulation of Ca2+/CaMKII/CREB signaling pathway. Exp Neurol 327:113210

    Article  CAS  PubMed  Google Scholar 

  82. Shaikh S, Dhavan P, Singh P, Uparkar J, Vaidya SP, Jadhav BL et al (2022) Synthesis of carbazole based α-aminophosphonate derivatives: design, molecular docking and in vitro cholinesterase activity. J Biomol Struct Dyn 40(11):4801–4814

    Article  CAS  PubMed  Google Scholar 

  83. Bindu S, Mazumder S, Bandyopadhyay U (2020) Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: a current perspective. Biochem Pharmacol 180:114147

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Wang X, Jin M, Jin C, Sun J, Zhou W, Li G (2018) A new sesquiterpene, a new monoterpene and other constituents with anti-inflammatory activities from the roots of Aristolochia debilis. Nat Prod Resh 34(3):351–358

    Article  Google Scholar 

  85. Dang T, Zheng G, Zhang Q, Jin P, Zhang H, Su L et al (2019) Sesquiterpenoids with diverse carbon skeletons from the roots of Cichorium glandulosum and their anti-inflammatory activities. Fitoterapia 136:104170

    Article  CAS  PubMed  Google Scholar 

  86. Cheng X, Zeng Q, Ren J, Qin J, Zhang S, Shen Y et al (2011) Sesquiterpene lactones from Inula falconeri, a plant endemic to the Himalayas, as potential anti-inflammatory agents. Eur J Med Chem 46(11):5408–5415

    Article  CAS  PubMed  Google Scholar 

  87. Tan J-Y, Liu Y, Cheng Y-G, Sun Y-P, Pan J, Yang S-H et al (2020) Anti-inflammatory sesquiterpenoids from the leaves of Datura metel L. Fitoterapia 142:104531

    Article  CAS  PubMed  Google Scholar 

  88. Zou Y-H, Zhao L, Xu Y-K, Bao J-M, Liu X, Zhang J-S et al (2018) Anti-inflammatory sesquiterpenoids from the traditional Chinese medicine Salvia plebeia: regulates pro-inflammatory mediators through inhibition of NF-κB and Erk1/2 signaling pathways in LPS-induced Raw264.7 cells. J Ethnopharmacol 210:95–106

    Article  CAS  PubMed  Google Scholar 

  89. Ge J, Liu Z, Zhong Z, Wang L, Zhuo X, Li J et al (2022) Natural terpenoids with anti-inflammatory activities: potential leads for anti-inflammatory drug discovery. Bioorg Chem 124:105817

    Article  CAS  PubMed  Google Scholar 

  90. Bandgar BP, Adsul LK, Chavan HV, Jalde SS, Shringare SN, Shaikh R et al (2012) Synthesis, biological evaluation, and docking studies of 3-(substituted)-aryl-5-(9-methyl-3-carbazole)-1H-2-pyrazolines as potent anti-inflammatory and antioxidant agents. Bioorg Med Chem Lett 22(18):5839–5844

    Article  CAS  PubMed  Google Scholar 

  91. Nalli Y, Khajuria V, Gupta S, Arora P, Riyaz-Ul-Hassan S, Ahmed Z et al (2016) Four new carbazole alkaloids from Murraya koenigii that display anti-inflammatory and anti-microbial activities. Org Biomol Chem 14(12):3322–3332

    Article  CAS  PubMed  Google Scholar 

  92. Jahan H, Siddiqui NN, Iqbal S, Basha FZ, Shaikh S, Pizzi M et al (2022) Suppression of COX-2/PGE2 levels by carbazole-linked triazoles via modulating methylglyoxal-AGEs and glucose-AGEs–induced ROS/NF-κB signaling in monocytes. Cell Signal 97:110372

    Article  CAS  PubMed  Google Scholar 

  93. Tan MA, Sharma N, An SSA (2022) Phyto-carbazole alkaloids from the rutaceae family as potential protective agents against neurodegenerative diseases. Antioxidants 11(3):493

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Syed FZ (2022) Type 1 diabetes mellitus. Ann Intern Med 175(3):ITC33–ITC48

    Article  PubMed  Google Scholar 

  95. Rai U, Senapati D, Arora MK (2023) Insights on the role of anti-inflammatory and immunosuppressive agents in the amelioration of diabetes. Diabetol Int 14(2):134–144

    Article  PubMed  Google Scholar 

  96. Ojo OA, Ibrahim HS, Rotimi DE, Ogunlakin AD, Ojo AB (2023) Diabetes mellitus: From molecular mechanism to pathophysiology and pharmacology. Med Novel Technol Devices 19:100247

    Article  Google Scholar 

  97. Mariadoss AVA, Sivakumar AS, Lee C-H, Kim SJ (2022) Diabetes mellitus and diabetic foot ulcer: Etiology, biochemical and molecular based treatment strategies via gene and nanotherapy. Biomed Pharmacother 151:113134

    Article  CAS  PubMed  Google Scholar 

  98. Antar SA, Ashour NA, Sharaky M, Khattab M, Ashour NA, Zaid RT et al (2023) Diabetes mellitus: classification, mediators, and complications; a gate to identify potential targets for the development of new effective treatments. Biomed Pharmacother 168:115734

    Article  CAS  PubMed  Google Scholar 

  99. Wang G, Wang J, He D, Li X, Li J, Peng Z (2016) Synthesis and biological evaluation of novel 1,2,4-triazine derivatives bearing carbazole moiety as potent α-glucosidase inhibitors. Bioorg Med Chem Lett 26(12):2806–2809

    Article  CAS  PubMed  Google Scholar 

  100. Patel OP, Mishra A, Maurya R, Saini D, Pandey J, Taneja I et al (2016) Naturally occurring carbazole alkaloids from Murraya koenigii as potential antidiabetic agents. J Nat Prod 79(5):1276–1284

    Article  CAS  PubMed  Google Scholar 

  101. Kasim SM, Al-Dabbagh BM, Mustafa YF (2022) A review on the biological potentials of carbazole and its derived products. Eur Chem Commun 4(6):495–512

    CAS  Google Scholar 

Download references

Acknowledgements

Authors are thankful the Khwaja Moinuddin Chishti Language University, Lucknow, Uttar Pradesh, India, and Dr. Shakuntala Mishra National Rehabilitation University, Lucknow, Uttar Pradesh, India, for providing the necessary facilities to complete this manuscript.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

AT has done various works on synthetic chemistry on carbazole derivatives and collected important information from her research and various literature for this review. BM has conceptualized the idea for this review, prepared the outlines and did the refinement of the paper.

Corresponding author

Correspondence to Bharat Mishra.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All the authors have read the manuscript and given their consent for publication.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tiwari, A., Mishra, B. Diverse pharmacological actions of potential carbazole derivatives by influencing various pathways of molecular signaling. Futur J Pharm Sci 10, 77 (2024). https://doi.org/10.1186/s43094-024-00650-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-024-00650-0

Keywords