scispace - formally typeset
Search or ask a question

What's the latest science of reversing grey hair? 


Best insight from top research papers

The latest science of reversing grey hair involves several approaches. One approach is the use of specific compounds such as enzymes and essential amino acids taken as oral dietary supplements . Another approach is the use of combination therapeutics like RT1640, which consists of drugs that stimulate hair growth and a non-immunosuppressive immunophilin ligand implicated in tissue regeneration . Additionally, adipose-derived autologous mesenchymal stem cells have been tested for their ability to regenerate tissues . Recent research has also shown that hair greying is reversible in humans, with white hairs naturally regaining pigmentation within days to weeks . Proteomic analysis has revealed that graying is marked by the upregulation of proteins related to energy metabolism, mitochondria, and antioxidant defenses . Furthermore, stress has been found to accelerate hair greying, and the reduction of stress can lead to the reversal of grey hair .

Answers from top 4 papers

More filters
Papers (4)Insight
The paper provides evidence that human hair graying is naturally reversible, but it does not discuss the latest science or methods for reversing gray hair.
The paper discusses a new method for measuring small changes in hair coloring and suggests that some white hairs naturally regain their color. However, it does not provide information on the latest science of reversing grey hair.
The paper discusses the use of specific compounds such as enzymes and essential amino acids taken as oral dietary supplements to slow, stop, or reverse human greying.
The provided paper does not discuss the latest science of reversing grey hair. It focuses on the incidental finding of hair greying reversal following autologous adipose mesenchymal stem cell transplantations for neurodegenerative disorders.

Related Questions

What are the best treatments for gray hair?4 answersThe best treatments for gray hair include traditional Chinese medicine formulations and compositions. These treatments aim to improve hair quality, promote hair growth, and enhance melanin secretion to restore black hair color. Traditional Chinese medicine for treating gray hair consists of various herbal ingredients such as prepared rehmannia roots, herba ecliptae, and chrysanthemums. Another traditional Chinese medicine formulation includes Radix Polygoni Multiflori Preparata, Radix Angelicae Sinensis, and Radix Glycyrrhizae, which can supplement the required nutrition and gradually restore black hair. Additionally, traditional Chinese medicine wine composed of radix rehmannia preparata, lyceum sinensis, and radix glycyrrhizae has shown good treatment effects and efficiency. Apart from traditional Chinese medicine, a composition containing collagen, hydroxyethyl cellulose, and keratin has been developed to delay and dye gray hair, restoring its natural color and revitalizing the hair.
How to cure premature gray hair?5 answersPremature graying of hair can be treated using various methods. One approach is the use of topical formulations containing biomimetic biopeptides such as palmitoyl tetrapeptide 20, which stimulates melanogenesis and promotes the conversion of gray hair to black hair. Traditional Chinese medicine compositions have also been developed for treating premature graying of hair. These compositions consist of a combination of medicinal herbs such as cuscuta chinensis, medlar, radix rehmanniae, red paeonia, folium mori, cortex moutan, chrysanthemum, fleece-flower root, radix angelicae, fructus viticis, and ligusticum wallichii. Another traditional Chinese medicine prescription includes medicinal materials like cortex mori, galla chinensis, cortex lycii, semen juglandis, flos carthami, poria, radix polygoni multiflori preparata, herba gynostemmatis pentaphylli, morus nigra, and semen sesami nigrum. Additionally, a medicine preparation method involves the use of traditional Chinese medicines such as phoenix tree seed, black sesame, radix polygoni multiflori, radix rehmanniae preparata, Chinese angelica, radix astragali, and fructus lycii. These methods aim to nourish the body, supplement qi and blood, and promote the growth of black hair.
Can aging be reversed?5 answersAging can be reversed through various mechanisms. Recent studies have shown that mitochondrial dysfunction, a hallmark of aging, can be restored by reestablishing communication between mitochondria and the nucleus. Reprogramming strategies, such as converting somatic cells to induced pluripotent stem cells, have also demonstrated the rejuvenation of aged cells. Reactivating telomerase, an enzyme involved in telomere maintenance, has been shown to reverse multiple aging phenotypes in mice. Furthermore, aging can be postponed or reversed through simple physiological interventions that target fundamental age-related changes, such as reduced transcription, altered gene expression, and reduced DNA repair. These observations suggest that aging is not an inevitable process and that interventions can potentially reverse or delay its effects.
How to revert to original skin color?3 answersTo revert to the original skin color, several methods and devices have been developed. One approach involves using a skin color rectification method based on color conceptualization. This method includes face identification, brightness assessment, brightness enhancement, tone statistics, and color conversion. Another method involves skin color adjustment using skin color models. This method recognizes the skin color region, calculates original color data, selects a skin color model, determines target color data, and adjusts the skin color region accordingly. Additionally, a skin changing method can be used, which involves analyzing a skin resource package, obtaining a picture color value, and carrying out dyeing processing on the original picture. These methods aim to reduce the difference between the adjusted skin color and the actual skin color, improve the beautifying effect, and simplify the operation.
What is the latest research on age reversing via NMA in humans?5 answersEpigenetic "clocks" have shown that epigenetic aging can be reversed in humans. A study conducted by Fahy et al. found that a protocol intended to regenerate the thymus resulted in protective immunological changes, improved risk indices for age-related diseases, and a mean epigenetic age approximately 1.5 years less than baseline after 1 year of treatment. The rate of epigenetic aging reversal accelerated over time, with a greater decrease in epigenetic age relative to chronological age observed from 9-12 months of treatment. The study also showed a decrease in the GrimAge predictor of human morbidity and mortality, indicating an increase in predicted human lifespan. This study is the first to report an increase in predicted lifespan based on an epigenetic age estimator through an accessible aging intervention.
How can you reverse aging?5 answersReverse aging can be achieved through various methods. One approach is the use of stem cell therapies, which have the potential to delay or even reverse aging by leveraging the plasticity, self-renewal, and multidirectional differentiation potential of stem cells. Another method involves identifying and targeting molecular biomarkers associated with aging through next-generation sequencing and gene regulatory network analysis. By understanding the regulatory mechanisms and functional repercussions of these biomarkers, therapeutic strategies can be developed to counteract the effects of aging. Additionally, facial reverse-aging techniques have been explored, which involve quantifying roughness in appearance and using wrinkle inpainting to rejuvenate the face and make it look younger. These approaches offer promising avenues for reversing the natural aging process and improving the quality of life for individuals.

See what other people are reading

How does the interface between an electroconductive scaffold and medium behave under applied electrical stimulation?
5 answers
The interface between an electroconductive scaffold and medium under applied electrical stimulation influences cell behavior and differentiation. Conductive scaffolds, such as chitosan-polyaniline substrates, when combined with electrical stimulation, promote neural-like phenotypes in stem cells, enhancing nerve tissue engineering outcomes. Carbon nanofiller-based scaffolds, synchronized with electrical stimulation, show promise in nerve tissue repair due to their excellent electrical properties and biocompatibility. In cardiac tissue engineering, external electrical stimulation in conductive scaffolds affects cardiomyocyte behavior positively, leading to improved function and tissue construct optimization. Additionally, electroconductive scaffolds combined with electrical stimulation guide adipose-derived stem cells towards neurogenic differentiation, indicating potential applications in nerve injury regeneration. These findings collectively highlight the significant impact of the electroconductive scaffold-medium interface under electrical stimulation on directing cell behavior and differentiation for tissue engineering applications.
What is a good study design for the effect of nutrients in a murine model?
5 answers
A well-designed study for investigating the effects of nutrients in a murine model involves random assignment of mice to different dietary intervention groups, measurement of relevant parameters, and comparison of outcomes. For example, studies like those by Yang et al.and Wang et al.utilized random assignment of mice to intervention groups with specific nutrient combinations to assess outcomes related to insulin resistance and Alzheimer's disease, respectively. Additionally, incorporating behavioral tests, biochemical analyses, and histological evaluations, as demonstrated in studies by Muranishi et al.and Cai et al., can provide a comprehensive understanding of the impact of nutrients on various aspects of health in murine models. Such a design allows for a thorough assessment of the effects of nutrients on physiological parameters and disease outcomes in murine models.
How dose high fat diet afect SCFA?
5 answers
High-fat diets (HFD) have varying effects on short-chain fatty acids (SCFAs). Research indicates that refined HFD can increase gut microbiota diversity and SCFAs, leading to enhanced energy metabolism. Additionally, supplementation of SCFAs in HFD-induced obese mice can reverse abnormal expressions of adiponectin and resistin, potentially through epigenetic regulation. Furthermore, a combination of HFD with sucrose can lead to morphological changes in the small intestine, affecting lipid metabolism and inflammation markers. Substituting saturated fatty acids (SFA) with monounsaturated fatty acids (MUFA) in a HFD can prevent hyperinsulinemia and pancreatic dysfunction, suggesting a role in mitigating metabolic inflammation. Overall, high-fat diets can impact SCFAs through alterations in gut microbiota diversity, gene expressions, and epigenetic modifications, influencing metabolic outcomes and inflammatory processes.
How does fatty diet affect immunity?
5 answers
A high-fat diet (HFD) impacts immunity by inducing inflammation and altering immune cell populations in adipose tissue. Studies show that HFD consumption leads to increased infiltration of pro-inflammatory Th17 cells, dendritic cells, and macrophages, while reducing regulatory T cells. This results in elevated levels of inflammatory cytokines and chemokines, along with changes in key signaling pathways like PPAR-γ. Additionally, dietary fatty acids, particularly saturated fatty acids like palmitic acid (PA), can act as inflammatory stimuli, inducing a hyper-inflammatory response and altering immune memory. PA exposure enhances inflammation in macrophages through ceramide synthesis, contributing to long-lived innate immune memory. Conversely, oleic acid reverses PA-induced inflammation. These findings highlight the significant impact of fatty diets on immunity, influencing immune cell function and inflammatory responses.
What are the benefits of strength training?
4 answers
Strength training offers a multitude of benefits across various aspects of health and wellness. It positively impacts cardiovascular health by reducing blood pressure, improving lipid profiles, and lowering the risk of associated diseases. Additionally, it enhances musculoskeletal health by increasing bone density, muscle strength, and flexibility, serving as an effective treatment for conditions like osteoporosis and arthritis. Strength training also contributes to neuropsychological well-being by reducing anxiety and depression while enhancing cognitive function. Moreover, it plays a role in metabolic health by managing weight, increasing fat-free mass, and boosting metabolic rate, aiding in weight management and overall health improvement. Overall, incorporating strength training into a routine can significantly impact various facets of health, making it a valuable component of a well-rounded fitness regimen.
What is the role of FXR in regulating lipid metabolism?
5 answers
The farnesoid X receptor (FXR) plays a crucial role in regulating lipid metabolism. FXR is involved in the transcriptional regulation of genes related to lipid homeostasis, glucose metabolism, and mitochondrial functions in adipose tissues. Studies have shown that FXR modulates the downstream genes involved in lipid metabolism, such as CYP7A1, impacting lipid accumulation in the liver and preventing liver fibrosis. Additionally, FXR is implicated in the absorption and transportation of lipids through interactions with bile acids, highlighting its significance in lipid metabolic processes. Overall, FXR's regulatory role in lipid metabolism extends to adipose tissue expansion, mitochondrial gene expression, energy expenditure, and systemic glucose tolerance, emphasizing its importance in maintaining whole-body energy homeostasis.
Inulin effects in mice?
5 answers
In mice, inulin supplementation has shown various beneficial effects. Studies have demonstrated that inulin can lead to weight loss, improved glucose tolerance, increased energy expenditure, and reduction in visceral adipose tissue. Inulin alters the gut microbiota composition, increasing the abundance of beneficial bacteria like Alistipes and promoting the production of short-chain fatty acids (SCFAs). Furthermore, inulin intake has been associated with anti-inflammatory properties, potentially through the inhibition of the NF-κB signaling pathway and the promotion of defense proteins like Hmox1. These findings suggest that inulin plays a significant role in modulating metabolic parameters, gut microbiota, and inflammatory responses in mice, highlighting its potential as a nutritional strategy for addressing metabolic diseases and obesity-related pathologies.
How does the disruption of the circadian clock affect the development and progression of osteoporosis?
5 answers
Disruption of the circadian clock significantly impacts osteoporosis development and progression. The circadian system plays a crucial role in bone health, with disturbed rhythms linked to osteoporosis. The circadian rhythm influences bone remodeling, and genetic disruptions in clock genes can lead to bone structure alterations. In aging, interrupted circadian rhythms contribute to decreased osteogenic differentiation of bone marrow stem cells, a key factor in senile osteoporosis. Moreover, the skeletal muscle circadian clock is vital for regulating lipid and glucose metabolism, further connecting circadian disturbances to osteoporosis pathogenesis. Addressing circadian disruptions through therapeutic strategies targeting clock genes like BMAL1 and TTK may hold promise in treating osteoporosis and delaying bone repair in aging individuals.
What are the potential long-term effects of [insert drug name] on human cells at a molecular level?
5 answers
Low-dose naltrexone (LDN) can have varying effects on human cells at a molecular level. LDN treatment alters the immunomodulatory properties of adipose-derived mesenchymal stem cells (ASCs), affecting genes like IDO, PD-L1, and cytokine levels. Asparaginase treatment for acute lymphoblastic leukemia can lead to pancreatitis, prompting the need for long-term follow-up due to potential health-related effects. Ankaferd blood stopper® (ABS) does not significantly impact drug-metabolizing enzymes in HepG2 cells, suggesting no risk of clinical drug toxicity or metabolism disorders. Celecoxib induces endoplasmic reticulum stress and affects mitochondrial function in cancer cells, potentially through the regulation of specific genes like TMEM117, TFAM, and UCP2.
Why weight gain non-significant in high-fat diet induced obesity-resistant mice?
4 answers
Weight gain is non-significant in high-fat diet-induced obesity-resistant mice due to various factors. Studies show that obesity-resistant animals may exhibit higher physical activity levels and lipid oxidation rates, leading to less fat deposition. Additionally, differences in gene expression related to fat metabolism, such as lower induction of PPARγ and CD36 in the liver of obesity-resistant mice compared to obesity-prone mice during high-fat diet feeding, contribute to their resistance to weight gain. Furthermore, the gut microbiota plays a crucial role in regulating energy homeostasis, as seen in mice fed with different polyunsaturated fatty acids-enriched diets, which improved metabolism and inhibited colonic inflammation in high-fat diet-induced obesity. These combined factors help explain why weight gain is non-significant in high-fat diet-induced obesity-resistant mice.
What is the link between senescence, fibrosis and cancer ?
5 answers
Senescence plays a crucial role in connecting fibrosis and cancer. Senescent cells develop a senescence-associated secretory phenotype (SASP) that includes the secretion of cytokines, growth factors, and exosomes, promoting fibrosis. This SASP can induce genetic instability and epithelial-mesenchymal transition, facilitating cancer progression. Additionally, senescent fibroblasts acquire a cancer-associated fibroblast (CAF) phenotype, promoting tumor growth and metastasis through the secretion of specific factors and increased collagen activity. The interplay between senescence, fibrosis, and cancer involves shared mechanisms such as TGF-β signaling, oxidative stress, and cellular senescence, contributing to the development and progression of both fibrotic diseases and cancers. Understanding these connections is crucial for developing innovative treatment strategies targeting common pathways in fibrosis and cancer.