Skip to main content

PAI1 inhibits the pathogenesis of primary focal hyperhidrosis by targeting CHRNA1

Abstract

Background

Primary focal hyperhidrosis (PFH) may be attributed to the up-regulation of the cholinergic receptor nicotinic alpha 1 subunit (CHRNA1) in eccrine glands. Plasminogen activator inhibitor-1 (PAI1, encoded by SERPINE1) is reported to inhibit the expression of CHRNA1, while the role of PAI1 in hyperhidrosis is unknown.

Methods

Serpine1 KO mice, Serpine1-Tg mice, and wild type BALB/c mice were intraperitoneally injected with pilocarpine hydrochloride to induce PFH. Cisatracurium (CIS, antagonist of CHRNA1) or PAI-039 (small-molecule inhibitor of PAI1) was pre-administrated before the induction of hyperhidrosis. On the other hand, Chrna1-expressing AAV was constructed and administered to Serpine1-Tg mice with hydrochloride stimulation. Hydrochloride-related biomarkers, such as acetylcholine (ACH) in the serum, calcium voltage-gated channel subunit alpha1 C (CACNA1C), and aquaporin 5 (AQP5) in sweat glands of mice were assayed with ELISA, RT-PCR, and Western blot.

Results

The administration of PAI-039 or Pai1 knock-out increased Chrna1 expression, sweat secretion, and hydrochloride-related biomarkers (ACH, CACNA1C, and AQP5) expression. On the other hand, CIS administration diminished the strengthened hyperhidrosis phenotype induced by Pai1 knock-out with decreased sweat gland secretion.

Conclusion

PAI1 inhibits CHRNA1-mediated hydrochloride-induced hyperhidrosis, with decreased sweat gland secretion and diminished ACH, AQP5, and CACNA1C expression. These results indicate the potential to utilize PAI1 to alleviate PFH.

Background

Hyperhidrosis is a dermatosis pathologically characterized by excessive generalized or localized (focal) sweating. Focal hyperhidrosis (PFH) is the most common form, and exocrine glands are overactive due to sympathetic nervous system dysfunction not triggered by temperature or physical activity [1, 2]. The prevalence of PFH has been estimated to be 2-3% [3]. PFH patients are more prone to cutaneous infection, and clammy palms, soles, and axillae can seriously affect the life quality of PFH patients [4]. Thoracoscopic excision of the T2 and T3 sympathetic ganglia and radiofrequency ablation are suggested for both palmar and axillary hyperhidrosis-affected patients, while the long-term outcome is not satisfactory in considering compensatory hyperhidrosis at the back [5, 6]. Therefore, it is urgent to decipher more relevant mechanisms to find a curable treatment target.

Up-regulated choline acetyltransferase, acetylcholine (ACH) receptor subunits, and alpha-7 neuronal nicotinic receptor subunits are detected in the thoracic sympathetic ganglion, which indicates that cholinergic neuron dysregulation might contribute to the development of PFH [7]. Significantly up-regulated cholinergic receptor nicotinic alpha 1 subunit (CHRNA1) is observed in thoracic sympathetic ganglia, which can bind and gate the ACH neurotransmitter [8]. In our previous research, small-interfering RNA targeting Chrna1 could attenuate the pathogenesis of pilocarpine-induced hyperhidrosis in mice [9]. On the other hand, we also found that cisatracurium pretreatment could block the function of CHRNA1 as an ion channel to prevent the pathogenesis of PFH in mice [10]. These studies demonstrate that CHRNA1 is a potential target to treat PFH.

Plasminogen activator inhibitor-1 (PAI1), encoded by SERPINE1, is a multi-functional protein, which can function as a principal inhibitor of plasminogen activation to inactivate both urokinase-type and tissue-type plasminogen activators [11]. On the other hand, PAI1 is a mechanistic contributor to several elements of the syndrome, including hepatic steatosis, and dyslipidemia, obesity, insulin resistance, and hypertension [12]. It is worth noting that PAI1 inhibitors (TM5484) could induce dose-dependent up-regulated CHRNA1 expression in myoblastic cells to prevent sarcopenia progression [13]. Whether PAI1 could be the upstream molecule to modulate the expression of CHRNA1 is still unknown. In this study, the potential role of PAI1 in targeting CHRNA1 to alleviate PFH is deciphered.

Methods

Hyperhidrosis mice model and treatment

Serpine1 (PAI1) KO mice (JAX: 002507) and Serpine1 (PAI1) transgenic mice (JAX: 007237) were ordered from Jackson Lab, and BALB/c mice (6–7 weeks old) were purchased from Peking Vital River Laboratory Animal Ltd. (Beijing, China). Six hours before hyperhidrosis induction, 1 mg/kg cisatracurium (CIS, Jiangsu Hengrui Pharmaceutical Company, Lianyungang, China) was injected intraperitoneally, 1 mg/kg PAI-039 (Sigma-Aldrich, St. Louis, MO) was administered orally, and 5 × 1010 µg Chrna1-expressing AAV (GenScript, Nanjing, China) were administrated as indicated. All experimental procedures were approved by the Animal Ethics Committee of Fujian Medical University (2020[328]).

2% iodine solution was smeared on the hind paws, and 5 mg/kg pilocarpine hydrochloride (Sigma-Aldrich) was immediately intraperitoneal injected to induce hyperhidrosis. When iodine solution was drying, 0.5 g/mL starch was utilized to cover the surface of the hind paws. Five min after perspiration stimulation, black spots on hind paws were imaged to assay hyperhidrosis with ImageJ software (National Institutes of Health). Two hours after hyperhidrosis induction, tissues and serum were harvested for further analysis.

Western blotting

Lysed sweat gland tissues (50 µg), collected from three mice in each group after the examination of ACH secretion, were loaded on 12% SDS-PAGE and transferred to PVDF membranes. Nonspecific binding was blocked with 1% BSA for 1 h. The membranes were incubated with the primary antibodies specific for PAI1 (Abcam, cat#: ab28207), AQP5 (Abcam, cat#: ab78486), and CACNA1C (Abcam, cat#: ab84814) at a 1:1000 dilution at 4 °C overnight. The PVDF membranes were then incubated in peroxidase-conjugated secondary antibody (Invitrogen, Waltham, MA USA) at a 1:1000 dilution for 1 h and developed with an ECL system (GE Healthcare Life Sciences, Chalfont, UK). GAPDH was used as an internal control.

qRT-PCR

Total RNAs were isolated from sweat gland tissues, collected from another three mice in each group after the examination of ACH secretion, with Trizol (ThermoFisher, Waltham, MA USA), which were further reversely transcribed into cDNA, and SYBR green kit (ThermoFisher) was utilized to detect the amplification with the following procedure: 95 °C for 10 min to denature, 40 cycles of 95 °C for 15 s to amplification, and 60 °C for 1 min to prolongate on a Mastercycler® X50 Eppendorf thermocycler. Gapdh was used as an internal control. Primers were designed by Genscript Biotech Corporation (Nanjing, China) and validated by melt curve analysis, listed as follows:

Chrna1,

forward, 5’- TCATCATTCCCTGCCTGCTCTTCT-3’,

reverse, 5’-TCTCTGCAATGTACTTCACGCCCT-3’;

Pai1,

forward, 5’- TTCAGCCCTTGCTTGCCTC-3’,

reverse, 5’-ACACTTTTACTCCGAAGTCGGT-3’;

Aqp5,

forward, 5’-AGAAGGAGGTGTGTTCAGTTGC-3’,

reverse, 5’- GCCAGAGTAATGGCCGGAT-3’;

Cacna1c,

forward, 5’- ATGAAAACACGAGGATGTACGTT-3’,

reverse, 5’- ACTGACGGTAGAGATGGTTGC-3’;

Gapdh,

forward, 5’- AGGTCGGTGTGAACGGATTTG-3’,

reverse, 5’- TGTAGACCATGTAGTTGAGGTCA-3’.

ELISA

Acetylcholine (ACH) level in the mice serum (six mice in each group) was assayed with the relevant ELISA kit (Abcam, ab287812, Cambridge, MA, USA) following manufacturer instructions. All standards and samples were measured with a Multiskan Mk3 Microplate Reader (Thermo Fisher) at a wavelength of 450 nm.

Immunohistochemistry

Footpad skin tissues embedded in paraffin were sectioned (2 μm thickness) and processed with gradient alcohol and Antigen Retrieval Buffer (Abcam, ab93684). After blocking with 1% BSA, the sections were incubated with primary antibody against PAI1 (Abcam, Cat#: ab28207, 1:500), biotinylated secondary antibody, avidin: biotinylated enzyme complex, and 3,3′-diaminobenzidine substrates (Zhongshan Goldenbridge, Guangzhou, China). The sections were finally counterstained with hematoxylin.

Statistical analysis

Data points represented the biological replicates. The differences between two groups were analyzed with Student’s t test. The differences between multiple groups were analyzed with one-way ANOVA followed by a post hoc test. All statistical analyses were performed using GraphPad Prism.

Results

Down-regulated PAI1 in sweat glands of hydrochloride-induced hyperhidrosis mice

In order to investigate the involvement of PAI1 in hyperhidrosis development, PAI1 expression in the sweat glands of hydrochloride-induced hyperhidrosis mice was detected with RT-PCR and Western blot. Diminished PAI1 expression in both mRNA (Fig. 1A) and protein (Fig. 1B) levels were detected.

Fig. 1
figure 1

PAI1 was downregulated in sweat glands of hydrochloride-induced hyperhidrosis mice. The relative expression of PAI1 in both relative mRNA level (A) and relative protein level (B) in sweat glands of hydrochloride-induced hyperhidrosis mice were determined by RT-qPCR and Western blot, respectively. n = 3 mice. H, hyperhidrosis. The relative mRNA and protein expression level were normalized to control group. The significant difference was analyzed using two-tailed t-test. Error bar represents the Mean ± SD. **p < 0.01

PAI1 inhibition increases CHRNA1 expression and sweat secretion in hydrochloride-induced hyperhidrosis mice

PAI1 inhibitor (PAI-039) was utilized to decipher the role of PAI1 in hyperhidrosis. PAI-039 could up-regulate the relative expression of CHRNA1 in both mRNA (Fig. 2A) and protein levels (Fig. 2B C) in hydrochloride-induced hyperhidrosis mice, while PAI-039 did not alter the relevant CHRNA1 expression in normal mice without hydrochloride stimulation. On the other hand, PAI1 inhibition could functionally increase the extent of hyperhidrosis, as indicated by the increased number of black dots (Fig. 2D). These results indicated that PAI1 inhibition could promote hyperhidrosis phenotype and CHRNA1 expression.

Fig. 2
figure 2

Inhibition of PAI1 increased the relative expression of CHRNA1 and sweat secretion in hydrochloride-induced hyperhidrosis mice. Mice were treated with 1 mg/kg PAI-039 or vehicle 6 h before the induction of hyperhidrosis. After hyperhidrosis induction, the relative CHRNA1 mRNA (A) and protein level (B-C) in sweat glands were detected by RT-qPCR and Western blot, respectively. n = 3. (D) The number of black dots was calculated with ImageJ. n = 6 mice. H, hyperhidrosis. The relative mRNA and protein expression level were normalized to control-vehicle group. The difference was analyzed by one-way ANOVA. Error bar represents Mean ± SD. *p < 0.05, ***p < 0.001. ns, not significant

PAI1 deficiency increases CHRNA1 expression in hydrochloride-induced hyperhidrosis mice

A well-validated Pai1 KO mice were used and confirmed by no detection of Pai1 expression in foot pad skin (Fig. 3A) [14]. Pai1 knock-out mice demonstrated up-regulated expression of CHRNA1 in both mRNA (Fig. 3A) and protein levels (Fig. 3B C) after hyperhidrosis induction. In contrast, Pai1 knock-out mice without hyperhidrosis induction did not show any significant difference in the expression of CHRNA1 compared with normal control mice. These results indicated that Pai1 knock-out could promote CHRNA1 expression in hydrochloride-induced hyperhidrosis mice.

Fig. 3
figure 3

The deficiency of Pai1 increased CHRNA1 expression in sweat glands of hydrochloride-induced hyperhidrosis mice. (A-C) The relative CHRNA1 mRNA (A) and protein level (B-C) in sweat glands of indicated groups were determined by RT-qPCR and Western blot, respectively. n = 3 mice. H, hyperhidrosis. The relative mRNA and protein expression level were normalized to control-WT group. The difference was analyzed using one-way ANOVA. Error bar represents Mean ± SD. ***p < 0.001. ns, not significant

Pai1 deficiency promotes CHRNA1-mediated hydrochloride-induced hyperhidrosis

As an intermediate-acting, non-depolarizing neuromuscular blocking, CIS was utilized to down-regulation the relative expression of Chrna1 as indicated by previous research [10]. Pai1 knock-out mice demonstrated increased numbers of black dots, while such phenomenon could be prohibited by the administration of CIS (Fig. 4A), which indicated that PAI1 could negatively regulate CHRNA1-mediated hyperhidrosis. On the other hand, PFH-relevant molecules, such as ACH, AQP5, and CACNA1C, were detected. Pai1 knock-out mice demonstrated up-regulated secretion of ACH in the serum (Fig. 4B), increased mRNA and protein levels of AQP5 (Fig. 4C and E, and 4 F), and CACNA1C (Fig. 4D and E, and 4G) after hydrochloride exposure. These results demonstrate that PAI1 could inhibit CACNA1C expression, which may contribute to the alleviation of hydrochloride-induced hyperhidrosis.

Fig. 4
figure 4

The deficiency of PAI1 promoted sweat secretion in hyperhidrosis mice. WT and Pai1 KO mice were treated with vehicle (H-WT and H-KO groups) or cisatracurium (H-WT + CIS and H-KO + CIS groups) for 6 h, then were induced hyperhidrosis. After hyperhidrosis induction, the sweat secretion, serum acetylcholine level, and relative expression level of AQP5 and CACNA1 were analyzed. (A) After pilocarpine injection, black dots were calculated with ImageJ. n = 6 mice. (B) Acetylcholine concentration in serum was analyzed by ELISA. n = 6 mice. (C-D) The relative mRNA level of Aqp5(C) and Cacna1c(D) in sweat gland tissues were determined by RT-qPCR. n = 3 mice. (E-G) The relative protein level of AQP5 (E and F) and CACNA1C (E and G) in sweat gland tissues were determined by Western blot. n = 3 mice. H, hyperhidrosis. The relative mRNA and protein expression level were normalized to control-WT group. The difference was analyzed using one-way ANOVA. Error bar represents Mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001. ns, not significant

Transgenic Pai1 decreases CHRNA1-mediated hydrochloride-induced hyperhidrosis

To further reveal the role of PAI1, Pai1-Tg mice were used, in which Pai1 expression was driven by a CMV promoter. Up-regulated Pai1 expression was observed in Pai1-Tg mice (Fig. 5A and C, and 5D). As expected, Pai1-Tg mice showed diminished expression of CHRNA1 in both mRNA (Fig. 5B) and protein levels (Fig. 5C and E) after hydrochloride exposure in sweat glands.

Fig. 5
figure 5

Transgenic Pai1 decreased CHRNA1 expression in sweat glands of hydrochloride-induced hyperhidrosis mice (A-B) The relative mRNA level of Pai1(A) and Chrna1(B) in sweat glands of indicated groups were determined by RT-qPCR. (C-E) The relative protein level of PAI1 (C and D) and CHRNA1 (C and E) in sweat glands of indicated groups were determined by Western blot. n = 3 mice. H, hyperhidrosis. The relative mRNA and protein expression level were normalized to control-WT group. The difference was analyzed using one-way ANOVA. Error bar represents Mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001

Pai1-Tg mice showed a diminished number of black dots, which could be restored by the administration of Chrna1-expressing AAV (Fig. 6A). Pai1-Tg mice demonstrated down-regulated secretion of ACH in the serum (Fig. 6B) and down-regulated mRNA and protein levels of AQP5 (Fig. 6C and E, and 6 F) and CACNA1C (Fig. 6D and E, and 6G) in sweat glands of hydrochloride-induced hyperhidrosis when compared with normal mice exposure to hydrochloride. The above results further confirmed that transgenic Pai1 could inhibit the development of CHRNA1-mediated hydrochloride-induced hyperhidrosis.

Fig. 6
figure 6

Pai1 overexpression inhibited sweat secretion in hyperhidrosis mice. WT and Pai1 transgenic (Tg) mice were treated with empty vector (H-WT and H-Tg groups) or Chrna1-expressing AAV (H-WT + Chrna1 and H-Tg + Chrna1 groups) for 72 h, then were induced hyperhidrosis. After hyperhidrosis induction, the sweat secretion, serum acetylcholine level, and relative expression level of Aqp5 and Cacna1 were analyzed. (A) Black dots were counted with ImageJ after pilocarpine injection. n = 6 mice. (B) Acetylcholine concentration in the serum was analyzed by ELISA. n = 6 mice. (C-D) The relative mRNA level of Aqp5(C) and Cacna1c(D) in sweat gland tissues were determined by RT-qPCR. n = 3 mice. (E-G) The relative AQP5 (E and F) and CACNA1C (E and G) protein level in sweat gland tissues were determined by Western blot. n = 3 mice. H, hyperhidrosis. The relative mRNA and protein expression level were normalized to control-WT group. The difference was analyzed using one-way ANOVA. Error bar represents Mean ± SD. **p < 0.01, ***p < 0.001. ns, not significant

Discussion

Down-regulated PAI1 is observed in sweat glands of hydrochloride-induced hyperhidrosis mice. PAI1 inhibition with PAI-039 or Pai1 deficiency could induce up-regulated CHRNA1, ACH, AQP5, and CACNA1C expression and sweat secretion in hydrochloride-induced hyperhidrosis mice. On the other hand, transgenic Pai1 could inhibit the development of hyperhidrosis with decreased CHRNA1 expression, and the alleviation of hyperhidrosis phenotype could be restored by the administration of Chrna1-expressing AAV. Our results indicate that PAI1 could prohibit CHRNA1-mediated hydrochloride-induced hyperhidrosis.

Up-regulated CHRNA1 could be considered as a characteristic biomarker in PFH patients sweat glands [9]. CHRNA1 homeostasis is vital for postsynaptic signal regulation, and altered CHRN endocytosis mediated by carbonic anhydrase 3 might contribute to the pathogenesis of myasthenia gravis [15]. Postsynaptic CHRNA1 ablation could preserve muscle innervation in type III neuregulin 1 mutant mice [16]. On the other hand, pharmacological inhibition of PAI1 with tiplaxtinin could block lysosomal degradation to up-regulate surface PDL1 expression in melanoma to synergize PDL1 immune checkpoint blockade [17], which indicates that PAI1-mediated endocytosis is a novel mechanism to regulate the homeostasis of signal molecules. Although the detailed or precise mechanism of PAI1-mediated CHRNA1 inhibition is not deciphered in this study, our investigation further indicates that PAI1-mediated endocytosis may affect CHRNA1 homeostasis in hyperhidrosis. Further understanding of CHRNA1 homeostasis-mediated by PAI1 will shed light on the pathogenesis of hyperhidrosis. In addition, PAI1 may also bind to low-density lipoprotein receptor-related protein 1 to trigger JAK/STAT1 signaling [18]. Whether such a mechanism is also involved in hyperhidrosis needs further analysis.

PAI1 regulation could alter the relative expression of CACNA1C and AQP5, which are vital components involved in the process of sweat secretion and pathological hyperhidrosis. AQP5, an exocrine-type water-channel protein, may mediate the excretion of water from sweat gland [19, 20]. In Aqp5 knock-out mice, decreased number of active sweat glands is observed upon pilocarpine stimulation [21]. CACNA1C, as a subunit of voltage-dependent calcium channel, may mediate calcium ions influx into the cell upon membrane polarization [22, 23]. In addition to the fact that serum ACH level can be regulated by PAI1, it is indicated that PAI1 has an inhibitory effect on neurosecretion.

The precise mechanism related to PAI1 and CHRNA1 is not deciphered in this study. PAI1 may contribute to the development of thrombosis, fibrosis, obesity, and insulin resistance [24, 25]. PAI1 not only functions as a plasminogen activator inhibitor, but also interacts with vitronectin to promote the development of multiple cancers, thrombotic and vascular disorders [26, 27]. In addition, the function of PAI1 in the intravascular space differ from its role in the brain parenchyma under both pathological and physiological conditions [28]. Further research on the utilization of PAI1 in hyperhidrosis will depend on a detailed understanding of PAI1 interaction with vitronectin or the administration sites. For the indicated neurosecretion mechanism and potential plasminogen activator inhibitor, the clinic safety or route of administration may be a major consideration in the future.

There are two limitations should be noted. First, to obtain a more convincing conclusion, more samples in each group should be included. Meanwhile, it may introduce bias using Student’s t test for comparisons in a small number of samples. Second, studies on detailed mechanisms are warranted to understand the function of PAI1 in PFH.

Conclusion

PAI1 inhibition and Pai1 deficiency will promote CHRNA1 expression and pathogenesis of PFH. On the other hand, Pai1 transgenic could inhibit the expression of CHRNA1 and the pathogenesis of PFH. Such effect could be restored by the administration of Chrna1-expressing AAV. Our findings demonstrate that PAI1 could target CHRNA1-mediated hydrochloride-induced hyperhidrosis in mice.

Data Availability

The datasets used and/or analyzed during the current study are available from https://pan.baidu.com/s/1t5hRIaU7AoX3cv_P3BXbBA?pwd=3v9z.

Abbreviations

PFH:

Primary focal hyperhidrosis

CHRNA1:

Cholinergic receptor nicotinic alpha 1 subunit

PAI1:

Plasminogen activator inhibitor-1

ACH:

Acetylcholine

CACNA1C:

Calcium voltage-gated channel subunit alpha1 C

AQP5:

Aquaporin 5

References

  1. Khurana RK. Sweating Disorders; in (Aminoff MJ, and Daroff RB, eds) Encyclopedia of the Neurological Sciences (Second Edition). Oxford, Academic Press, 2014, vol. https://doi.org/10.1016/B978-0-12-385157-4.00513-3p.^pp. 359–362.

  2. Liu V, Farshchian M, Potts GA. Management of primary focal hyperhidrosis: an Algorithmic Approach. J Drugs Dermatol. 2021;20:523–8.

    PubMed  Google Scholar 

  3. Owens AP, Low DA, Iodice V, et al. Emotion and the autonomic nervous System—A Two-Way Street: insights from affective, autonomic and dissociative Disorders; in Reference Module in Neuroscience and Biobehavioral psychology. Elsevier; 2017. https://doi.org/10.1016/B978-0-12-809324-5.01799-5p.^pp.

  4. Henning MAS, Thorlacius L, Ibler KS, et al. How to diagnose and measure primary hyperhidrosis: a systematic review of the literature. Clin Auton Res. 2021;31:511–28.

    Article  PubMed  Google Scholar 

  5. Elhalaby IE, Mansour MA, Tawfik AMI, et al. Thoracoscopic T2-T3 versus T4 sympathectomy for primary palmar hyperhidrosis in children and adolescents: a randomized comparative study. Annals of Pediatric Surgery. 2021;17:26.

    Article  Google Scholar 

  6. Mostafa MS, Elsherbeny M, Abdelbarr A, et al. Thoracoscopic excision versus radiofrequency ablation of the sympathetic chain as a treatment for palmar hyperhidrosis: comparative study. Annals of Pediatric Surgery. 2020;16:21.

    Article  Google Scholar 

  7. de Moura Júnior NB, das-Neves-Pereira JC, de Oliveira FR, et al. Expression of acetylcholine and its receptor in human sympathetic ganglia in primary hyperhidrosis. Ann Thorac Surg. 2013;95:465–70.

    Article  PubMed  Google Scholar 

  8. Roux I, Wu JS, McIntosh JM, et al. Assessment of the expression and role of the α1-nAChR subunit in efferent cholinergic function during the development of the mammalian cochlea. J Neurophysiol. 2016;116:479–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lin JB, Kang MQ, Huang LP, et al. CHRNA1 promotes the pathogenesis of primary focal hyperhidrosis. Mol Cell Neurosci. 2021;111:103598.

    Article  CAS  PubMed  Google Scholar 

  10. Lin JB, Lin NL, Li X, et al. Antagonist of Chrna1 prevents the pathogenesis of primary focal hyperhidrosis. Ann Clin Transl Neurol. 2022;9:786–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Aihemaiti A, Yamamoto N, Piao J, et al. A novel PAI-1 inhibitor prevents ageing-related muscle fiber atrophy. Biochem Biophys Res Commun. 2021;534:849–56.

    Article  CAS  PubMed  Google Scholar 

  12. Levine JA, Oleaga C, Eren M, et al. Role of PAI-1 in hepatic steatosis and dyslipidemia. Sci Rep. 2021;11:430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chandler WL. Chap. 146 - Laboratory Techniques in Fibrinolysis Testing; in (Shaz BH, Hillyer CD, and Reyes Gil M, eds) Transfusion Medicine and Hemostasis (Third Edition). Elsevier, 2019, vol. https://doi.org/10.1016/B978-0-12-813726-0.00146-Xp.^pp. 865–868.

  14. Carmeliet P, Kieckens L, Schoonjans L, et al. Plasminogen activator inhibitor-1 gene-deficient mice. I. Generation by homologous recombination and characterization. J Clin Invest. 1993;92:2746–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Du A, Huang S, Zhao X, et al. Suppression of CHRN endocytosis by carbonic anhydrase CAR3 in the pathogenesis of myasthenia gravis. Autophagy. 2017;13:1981–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Liu Y, Sugiura Y, Chen F, et al. Blocking skeletal muscle DHPRs/Ryr1 prevents neuromuscular synapse loss in mutant mice deficient in type III neuregulin 1 (CRD-Nrg1). PLoS Genet. 2019;15:e1007857.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tseng YJ, Lee CH, Chen WY, et al. Inhibition of PAI-1 Blocks PD-L1 endocytosis and improves the response of Melanoma cells to Immune Checkpoint Blockade. J Invest Dermatol. 2021;141:2690–2698e2696.

    Article  CAS  PubMed  Google Scholar 

  18. Czekay R-P, Wilkins-Port CE, Higgins SP, et al. PAI-1: an integrator of Cell Signaling and Migration. Int J Cell Biol. 2011;2011:562481.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Moon CS, Moon D, Kang SK. Aquaporins in Cancer Biology Front Oncol. 2022;12:782829.

    CAS  PubMed  Google Scholar 

  20. Piraino LR, Benoit DSW, DeLouise LA. Salivary gland tissue Engineering Approaches: state of the art and future directions. Cells. 2021;10.

  21. Song Y, Sonawane N, Verkman AS. Localization of aquaporin-5 in sweat glands and functional analysis using knockout mice. J Physiol. 2002;541:561–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Moon AL, Haan N, Wilkinson LS, et al. CACNA1C: Association with Psychiatric Disorders, Behavior, and neurogenesis. Schizophr Bull. 2018;44:958–65.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Li M, Li T, Xiao X, et al. Phenotypes, mechanisms and therapeutics: insights from bipolar disorder GWAS findings. Mol Psychiatry. 2022;27:2927–39.

    Article  PubMed  Google Scholar 

  24. Miyashita K, Nishikawa S, Hosokawa M. Chap. 29 - therapeutic effect of Fucoxanthin on metabolic syndrome and type 2 diabetes. In: Bagchi D, Sreejayan N, editors. Nutritional and therapeutic interventions for diabetes and metabolic syndrome. San Diego: Academic Press; 2012. pp. 367-79. https://doi.org/10.1016/B978-0-12-385083-6.00029-2

    Chapter  Google Scholar 

  25. Ismail AA, Shaker BT, Bajou K. The plasminogen-activator plasmin system in physiological and pathophysiological angiogenesis. Int J Mol Sci. 2021;23.

  26. Andreasen PA. PAI-1 - a potential therapeutic target in cancer. Curr Drug Targets. 2007;8:1030–41.

    Article  CAS  PubMed  Google Scholar 

  27. Mahmood N, Rabbani SA. Fibrinolytic system and Cancer: diagnostic and therapeutic applications. Int J Mol Sci. 2021;22.

  28. Tjärnlund-Wolf A, Brogren H, Lo EH, et al. Plasminogen activator inhibitor-1 and thrombotic cerebrovascular diseases. Stroke. 2012;43:2833–9.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by Natural Science Foundation of Fujian Province (2022J02034); Joint Funds for the Innovation of Science and Technology of Fujian Province (2019Y9119), Fujian Provincial Health Technology Project (2020CXA037) and Fujian Provincial Finance Project (No.BPB-2022LJB).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by [JFC], [ML], [XL] and [JBL]. The first draft of the manuscript was written by [JBL] and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jian-Bo Lin.

Ethics declarations

Ethics approval and consent to participate

All experimental procedures were approved by the Animal Ethics Committee of Fujian Medical University (2020[328]). This study was performed in strict accordance with the NIH guidelines for the care and use of laboratory animals (8th edition, NIH).

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, JF., Lin, M., Li, X. et al. PAI1 inhibits the pathogenesis of primary focal hyperhidrosis by targeting CHRNA1. Orphanet J Rare Dis 18, 205 (2023). https://doi.org/10.1186/s13023-023-02808-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13023-023-02808-0

Keywords