Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Life Sci ; 333: 122172, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37832632

ABSTRACT

AIMS: Cachexia, a metabolic syndrome, affects 21 % of patients suffering from ischemic encephalopathy. However, the specific mechanism and prevention measures are still unclear. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been proven to reduce inflammatory cytokine levels during ischemic events, but whether they have a protective effect against cachexia after hypoxic-ischemic brain damage (HIBD) remains unclear. MAIN METHODS: C57BL/6J wild-type and mfat-1 transgenic male mice were treated with and without HIBD. One day after HIBD, the epididymal white fat, gastrocnemius muscle and hypothalamus were weighed and analyzed the phenotypic changes. RNA sequencing was applied to gastrocnemius muscle to identify differential genes and pathways in HIBD groups. The effect of HPA axis on cachexia post-HIBD was examined via adrenalectomy, dexamethasone (0.1 mg/kg), and corticosterone injection (100 mg/kg). KEY FINDINGS: The results showed that the incidence of cachexia in mfat-1 mice, which produce high proportion of n-3 PUFAs, was significantly lower than that in wild-type mice post-HIBD. Cachexia-related factors, such as inflammation, muscle atrophy and lipid metabolism were significantly improved in mfat-1 HIBD. RNA sequencing revealed that catabolic and proteasome pathways were significantly downregulated. In hypothalamus, inflammatory cytokines, lipid peroxidation levels were reduced. Corticosterone, glucocorticoid receptor, and dexamethasone suppression test all showed that mfat-1 improved the dysfunction of the HPA axis post-HIBD. The present study elucidated for the first time that mfat-1 reduced HIBD-induced hyperactivation of the HPA axis in mice by reducing inflammation and oxidative stress and contributed to the reduction of metabolic imbalance in peripheral tissues. SIGNIFICANCE: Our study provides mechanistic information for the development of intervention strategies to prevent cachexia.


Subject(s)
Hypothalamo-Hypophyseal System , Hypoxia-Ischemia, Brain , Humans , Mice , Animals , Male , Hypothalamo-Hypophyseal System/metabolism , Cachexia/etiology , Cachexia/prevention & control , Cachexia/metabolism , Corticosterone/metabolism , Mice, Inbred C57BL , Pituitary-Adrenal System/metabolism , Mice, Transgenic , Hypothalamus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Inflammation/metabolism , Dexamethasone/metabolism , Animals, Newborn , Brain/metabolism
2.
Nature ; 621(7979): 568-576, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37704722

ABSTRACT

Growth faltering in children (low length for age or low weight for length) during the first 1,000 days of life (from conception to 2 years of age) influences short-term and long-term health and survival1,2. Interventions such as nutritional supplementation during pregnancy and the postnatal period could help prevent growth faltering, but programmatic action has been insufficient to eliminate the high burden of stunting and wasting in low- and middle-income countries. Identification of age windows and population subgroups on which to focus will benefit future preventive efforts. Here we use a population intervention effects analysis of 33 longitudinal cohorts (83,671 children, 662,763 measurements) and 30 separate exposures to show that improving maternal anthropometry and child condition at birth accounted for population increases in length-for-age z-scores of up to 0.40 and weight-for-length z-scores of up to 0.15 by 24 months of age. Boys had consistently higher risk of all forms of growth faltering than girls. Early postnatal growth faltering predisposed children to subsequent and persistent growth faltering. Children with multiple growth deficits exhibited higher mortality rates from birth to 2 years of age than children without growth deficits (hazard ratios 1.9 to 8.7). The importance of prenatal causes and severe consequences for children who experienced early growth faltering support a focus on pre-conception and pregnancy as a key opportunity for new preventive interventions.


Subject(s)
Cachexia , Developing Countries , Growth Disorders , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Pregnancy , Cachexia/economics , Cachexia/epidemiology , Cachexia/etiology , Cachexia/prevention & control , Cohort Studies , Developing Countries/economics , Developing Countries/statistics & numerical data , Dietary Supplements , Growth Disorders/epidemiology , Growth Disorders/prevention & control , Longitudinal Studies , Mothers , Sex Factors , Malnutrition/economics , Malnutrition/epidemiology , Malnutrition/etiology , Malnutrition/prevention & control , Anthropometry
3.
Genes (Basel) ; 14(9)2023 09 19.
Article in English | MEDLINE | ID: mdl-37761958

ABSTRACT

Previous studies have shown that inhibition of TNF family member FN14 (gene: TNFRSF12A) in colon tumors decreases inflammatory cytokine expression and mitigates cancer-induced cachexia. However, the molecular mechanisms underlying the regulation of FN14 expression remain unclear. Tumor microenvironments are often devoid of nutrients and oxygen, yet how the cachexic response relates to the tumor microenvironment and, importantly, nutrient stress is unknown. Here, we looked at the connections between metabolic stress and FN14 expression. We found that TNFRSF12A expression was transcriptionally induced during glutamine deprivation in cancer cell lines. We also show that the downstream glutaminolysis metabolite, alpha-ketoglutarate (aKG), is sufficient to rescue glutamine-deprivation-promoted TNFRSF12A induction. As aKG is a co-factor for histone de-methylase, we looked at histone methylation and found that histone H3K4me3 at the Tnfrsf12a promoter is increased under glutamine-deprived conditions and rescued via DM-aKG supplementation. Finally, expression of Tnfrsf12a and cachexia-induced weight loss can be inhibited in vivo by DM-aKG in a mouse cancer cachexia model. These findings highlight a connection between metabolic stress and cancer cachexia development.


Subject(s)
Cachexia , Colonic Neoplasms , TWEAK Receptor , Animals , Mice , Cachexia/genetics , Cachexia/prevention & control , Disease Models, Animal , Glutamine/pharmacology , Histone Code , Histone Methyltransferases , Histones/genetics , Ketoglutaric Acids/pharmacology , Tumor Microenvironment , Humans , Cell Line, Tumor/metabolism , TWEAK Receptor/genetics , TWEAK Receptor/metabolism
4.
Nutrients ; 15(15)2023 Jul 30.
Article in English | MEDLINE | ID: mdl-37571328

ABSTRACT

Cancer cachexia is a multi-organ syndrome with unintentional weight loss, sarcopenia, and systemic inflammation. Gastrointestinal (GI) cancer patients are more susceptible to cachexia development due to impaired nutrient absorption and digestion. Given the widespread availability and relatively low cost of dietary supplements, we examined the evidence and effects of fish oil (omega-3 fatty acids), melatonin, probiotics, and green tea for managing symptoms of GI cancer cachexia. A literature review of four specific supplements was conducted using PubMed, Google Scholar, and CINAHL without a date restriction. Of 4621 available literature references, 26 articles were eligible for review. Fish oil decreased C-reactive protein and maintained CD4+ cell count, while melatonin indicated inconsistent findings on managing cachexia, but was well-tolerated. Probiotics decreased serum pro-inflammatory biomarkers and increased the tolerability of chemotherapy by reducing side effects. Green tea preparations and extracts showed a decreased risk of developing various cancers and did not impact tumor growth, survival, or adverse effects. Among these four supplements, probiotics are most promising for further research in preventing systemic inflammation and maintaining adequate absorption of nutrients to prevent the progression of cancer cachexia. Supplements may benefit treatment outcomes in cancer cachexia without side effects while supporting nutritional and therapeutic needs.


Subject(s)
Fatty Acids, Omega-3 , Gastrointestinal Neoplasms , Melatonin , Neoplasms , Humans , Cachexia/etiology , Cachexia/prevention & control , Cachexia/drug therapy , Fatty Acids, Omega-3/therapeutic use , Melatonin/therapeutic use , Fish Oils/therapeutic use , Dietary Supplements , Gastrointestinal Neoplasms/complications , Gastrointestinal Neoplasms/drug therapy , Neoplasms/drug therapy , Inflammation/drug therapy
5.
J Nutr Biochem ; 114: 109248, 2023 04.
Article in English | MEDLINE | ID: mdl-36503110

ABSTRACT

Cancer cachexia is a metabolic disease affecting multiple organs and characterized by loss adipose and muscle tissues. Metabolic dysregulated of adipose tissue has a crucial role in cancer cachexia. ß-Carotene (BC) is stored in adipose tissues and increases muscle mass and differentiation. However, its regulatory effects on adipose tissues in cancer cachexia have not been investigated yet. In this study, we found that BC supplementations could inhibit several cancer cachexia-related changes, including decreased carcass-tumor (carcass weight after tumor removal), adipose weights, and muscle weights in CT26-induced cancer cachexia mice. Moreover, BC supplementations suppressed cancer cachexia-induced lipolysis, fat browning, hepatic gluconeogenesis, and systemic inflammation. Altered diversity and composition of gut microbiota in cancer cachexia were restored by the BC supplementations. BC treatments could reverse the down-regulated adipogenesis and dysregulated mitochondrial respiration and glycolysis in adipocytes and colon cancer organoid co-culture systems. Taken together, these results suggest that BC can be a potential therapeutic strategy for cancer cachexia.


Subject(s)
Colonic Neoplasms , Gastrointestinal Microbiome , Neoplasms , Animals , Mice , Cachexia/etiology , Cachexia/prevention & control , Cachexia/metabolism , beta Carotene/metabolism , Adipose Tissue/metabolism , Neoplasms/metabolism , Colonic Neoplasms/complications , Colonic Neoplasms/metabolism , Muscle, Skeletal/metabolism
6.
Clin Nutr ; 41(12): 2843-2857, 2022 12.
Article in English | MEDLINE | ID: mdl-36403384

ABSTRACT

Cachexia, a multifactorial and often irreversible wasting syndrome, is often associated with the final phase of several chronic disorders. Although cachexia is characterized by skeletal muscle wasting and adipose tissue loss, it is a syndrome affecting different organs, which ultimately results in systemic complications and impaired quality of life. The pathogenesis and underlying molecular mechanisms of cachexia are not fully understood, and currently there are no effective standard treatments or approved drug therapies to completely reverse cachexia. Moreover, adequate nutritional interventions alone cannot significantly improve cachexia. Other approaches to ameliorate cachexia are urgently needed, and thus, the role of medicinal plants has received considerable importance in this respect due to their beneficial health properties. Increasing evidence indicates great potential of medicinal plants and their phytochemicals as an alternative and promising treatment strategy to reduce the symptoms of many diseases including cachexia. This article reviews the current status of cachexia, the molecular mechanisms of primary events driving cachexia, and state-of-the-art knowledge that reports the preventive and therapeutic activities of multiple families of phytochemical compounds and their pharmacological mode of action, which may hold promise as an alternative treatment modality for the management of cachexia. Based on our review of various in vitro and in vivo models of cachexia, we would conclude that phytochemicals may have therapeutic potential to attenuate cachexia, although clinical trials are required to unequivocally confirm this premise.


Subject(s)
Cachexia , Wasting Syndrome , Humans , Cachexia/drug therapy , Cachexia/prevention & control , Quality of Life , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Immunotherapy
7.
Medicina (Kaunas) ; 58(7)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35888685

ABSTRACT

Background and Objectives: The prevalence of cachexia has increased across all of the cancer types and accounts for up to 20% of cancer-related deaths. This paper is a systematic review of nutritional interventions aiming to improve cachexia outcomes in cancer, focusing on weight gain. Materials and Methods: A search in Medline and Elsevier databases for articles up until the 23 January 2022, was conducted. Results: Out of 5732 screened records, 26 publications were included in the final analysis. Four randomized clinical trials showed a significant body weight (BW) increase in patients treated with eicosapentaenoic acid (EPA), ß-hydroxy-beta-methyl butyrate (ß-HMB), arginine, and glutamine or marine phospholipids (MPL). An upward BW trend was observed in patients treated with L-carnitine, an Ethanwell/Ethanzyme (EE) regimen enriched with ω-3 fatty acids, micronutrients, probiotics, fish oil, a leucine-rich supplement, or total parental nutrition (TPN) with a high dose of a branched-chain amino acid (BCAA). Conclusions: Although clinical trials relating to large numbers of nutritional supplements present promising data, many trials provided negative results. Further studies investigating the underlying mechanisms of action of these nutritional supplements in cancer cachexia are needed. Early screening for cancer cachexia risk and nutritional intervention in cancer patients before aggravating weight loss may stabilize their weight, preventing cachexia syndrome. According to the GRADE methodology, no positive recommendation for these nutritional supplements may be expressed.


Subject(s)
Cachexia , Neoplasms , Cachexia/drug therapy , Cachexia/etiology , Cachexia/prevention & control , Dietary Supplements , Eicosapentaenoic Acid/therapeutic use , Humans , Micronutrients/therapeutic use , Neoplasms/complications , Neoplasms/drug therapy
8.
Nutrients ; 14(7)2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35406121

ABSTRACT

Cachexia, which is characterised by the wasting of fat and skeletal muscles, is the most common risk factor for increased mortality rates among patients with advanced lung cancer. PTHLH (parathyroid hormone-like hormone) is reported to be involved in the pathogenesis of cancer cachexia. However, the molecular mechanisms underlying the regulation of PTHLH expression and the inhibitors of PTHLH have not yet been identified. The PTHLH mRNA levels were measured using quantitative real-time polymerase chain reaction, while the PTHrP (parathyroid hormone-related protein) expression levels were measured using Western blotting and enzyme-linked immunosorbent assay. The interaction between TCF4 (Transcription Factor 4) and TWIST1 and the binding of the TCF4-TWIST1 complex to the PTHLH promoter were analysed using co-immunoprecipitation and chromatin immunoprecipitation. The results of the mammalian two-hybrid luciferase assay revealed that emodin inhibited TCF4-TWIST1 interaction. The effects of Polygonum cuspidatum extract (Pc-Ex), which contains emodin, on cachexia were investigated in vivo using A549 tumour-bearing mice. Ectopic expression of TCF4 upregulated PTHLH expression. Conversely, TCF4 knockdown downregulated PTHLH expression in lung cancer cells. The expression of PTHLH was upregulated in cells ectopically co-expressing TCF4 and TWIST1 when compared with that in cells expressing TCF4 or TWIST1 alone. Emodin inhibited the interaction between TCF4 and TWIST1 and consequently suppressed the TCF4/TWIST1 complex-induced upregulated mRNA and protein levels of PTHLH and PTHrP. Meanwhile, emodin-containing Pc-Ex significantly alleviated skeletal muscle atrophy and downregulated fat browning-related genes in A549 tumour-bearing mice. Emodin-containing Pc-Ex exerted therapeutic effects on lung cancer-associated cachexia by inhibiting TCF4/TWIST1 complex-induced PTHrP expression.


Subject(s)
Emodin , Fallopia japonica , Lung Neoplasms , Animals , Cachexia/drug therapy , Cachexia/etiology , Cachexia/prevention & control , Emodin/pharmacology , Emodin/therapeutic use , Humans , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Mammals/genetics , Mammals/metabolism , Mice , Nuclear Proteins/genetics , Parathyroid Hormone-Related Protein/genetics , Plant Extracts , RNA, Messenger/metabolism , Transcription Factor 4/genetics , Twist-Related Protein 1/genetics
9.
Mol Nutr Food Res ; 65(9): e2000863, 2021 05.
Article in English | MEDLINE | ID: mdl-33651486

ABSTRACT

SCOPE: Nutritional supplementation of the maternal diet can modify the cancer susceptibility in adult offspring. Therefore, the authors evaluate the effects of a fish-oil diet administered to a long-term, during pre-mating, gestation, and lactation, in reducing cancer-cachexia damages in adult Walker-256 tumor-bearing offspring. METHODS AND RESULTS: Female rats receive control or fish oil diet during pre-mating, gestation, and lactation. After weaning, male offspring are fed the control diet until adulthood and distributed in (C) control adult-offspring; (W) adult tumor-bearing offspring; (OC) adult-offspring of maternal fish oil diet; (WOC) adult tumor-bearing offspring of maternal fish oil diet groups. Fat body mass is preserved, muscle expression of mechanistic target of rapamicin (mTOR) and eukariotic binding protein of eukariotic factor 4E (4E-BP1) is modified, being associated with lower 20S proteasome protein expression, and the liver alanine aminotransferase (ALT) enzyme content maintained in the WOC group. Also, the OC group shows reduced triglyceridemia. CONCLUSION: In this experimental model of cachexia, the long-term maternal supplementation is a positive strategy to improve liver function and lipid metabolism, as well as to modify muscle proteins expression in the mTOR pathway and also reduce the 20S muscle proteasome protein, without altering the tumor development and muscle wasting in adult tumor-bearing offspring.


Subject(s)
Cachexia/prevention & control , Carcinoma 256, Walker/complications , Fish Oils/administration & dosage , Alanine Transaminase/metabolism , Animals , Body Composition , Carcinoma 256, Walker/metabolism , Dietary Supplements , Female , Lactation , Male , Muscle Proteins/metabolism , Rats , Rats, Wistar , TOR Serine-Threonine Kinases/physiology , Triglycerides/blood
10.
BMC Complement Med Ther ; 21(1): 20, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33413302

ABSTRACT

BACKGROUND: Cancer is a multifactorial disease caused by uncontrolled proliferation of cells. About 50-80% of cancer patients develop cachexia, a complex metabolic syndrome associated with an increase of mortality and morbidity. However, there are no effective therapies in medical clinic for cancer cachexia. Vochysia tucanorum Mart. is a common three of the Brazilian "Cerrado". The butanolic fraction of V. tucanorum (Fr-BuVt), very rich in triterpenes with various biological activities, might be interesting in being tested in cancer cachexia syndrome. Hence, the present study was undertaken to investigate the antitumoral activity of Fr-BuVt and its potential against cachexia development. METHODS: Ehrlich tumor was used as model of cancer cachexia. Ascitic Ehrlich tumor cells were collected, processed and inoculated subcutaneously in saline solution (1 × 107/100 µl; ≥95% viability) for the obtention of solid Ehrlich carcinoma. After inoculation, solid Ehrlich carcinoma-bearing mice were treated by 14 consecutive days by gavage with Fr-BuVt (200 mg/kg). Body weight and tumor volume were measure during the treatment period. Tumors were removed, weighed and properly processed to measure the content and phosphorylation levels of key-proteins involved to apoptotic and proliferation process by Western Blot. Muscles and adipose tissues were removed for weighed. Serum was collected to cytokines levels and energetic blood markers measurements. RESULTS: The treatment with the Fr-BuVt (200 mg/kg, 14 days) decreased the solid Ehrlich tumor volume and weight besides increased the expression of the pro-apoptotic proteins caspase-3 and BAX, but also decreased the expression of the proteins involved in proliferation NFκB, mTOR and ERK. In addition, our data shows that the administration of Fr-BuVt was able to prevent the installation of cancer cachexia in Ehrlich carcinoma-bearing mice, since prevented the loss of body weight, as well as the loss of muscle and adipose tissue. Moreover, an improvement in some blood parameters such as decrease in cytokines TNF-α and IL-6 levels is observed. CONCLUSIONS: The study revealed that Fr-BuVt has antitumoral activity and prevent installation of cancer cachexia in Ehrlich model. Therefore, Fr-BuVt may represent an alternative treatment for cancer cachexia.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cachexia/prevention & control , Carcinoma, Ehrlich Tumor/drug therapy , Myrtales/chemistry , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Body Weight/drug effects , Brazil , Butanols , Cachexia/etiology , Carcinoma, Ehrlich Tumor/complications , Cell Proliferation/drug effects , Cytokines/blood , Male , Mice , Xenograft Model Antitumor Assays
11.
J Ethnopharmacol ; 267: 113470, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33068652

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus membranaceus (Fisch.) and Bunge and Paeonia japonica (Makino)Miyabe & H.Takeda have been traditionally used to improve the poor quality of life such as weakness, lack of appetite, fatigue, and malaise which is considered with cachexia condition. AIM OF THE STUDY: We investigated anti-cachectic effects of a herbal formula composed of Astragalus membranaceus and Paeonia japonica (APX) and the molecular mechanisms of APX in C26 cancer-induced cachexia mice and TNF-a-treated C2C12 myotubes. Additionally synergistic anti-cachectic effects of APX were compared to those of individual herbal extracts and megestrol acetate. METHODS AND MATERIALS: The forty-two BALB/c mice were randomly divided into 6 groups: normal (nontreatment), control (C26 injection), AM (C26 injection with Astragalus membranaceus), PJ (C26 injection with Paeonia japonica), APX (C26 injection with combination of Astragalus membranaceus and Paeonia japonica and MA (C26 injection with megestrol acetate). All mice were orally administered DW (normal and control groups) or 100 mg/kg AM, PJ, APX or MA for 10 days. In the animal model, several tissues were weighed, and muscle tissue and blood were used to measure pro-inflammatory cytokines. C2C12 myotubes were exposed to 100 ng/mL TNF- α with or without 10 µg/mL of AM, PJ, APX or MA for 48 h. The cells were used to immunofluorescence staining and western blot analyses. RESULTS: C26 injection induced notable body and muscle weight loss while APX administration significantly attenuated these alterations and the decrease of muscle weights and strength. APX also significantly attenuated the abnormal elevations in the concentration of three muscle atrophy-inducible cytokines; serum and muscle TNF-α,muscle TWEAK and IL-6 in C26 tumor-bearing mice. In the TNF-α-treated C2C12 myotube model, TNF-α treatment notably decreased MyH but activated atrophic proteins (MuRF and Fbx32) along with p38 and NFκB while these molecular alterations were significantly ameliorated by APX treatment. These pharmacological actions of APX were supported by the results of immunofluorescence staining to MyH expression and the translocation of NFκB into the nucleus in C2C12 myotubes. CONCLUSIONS: Our data indicate the potential of an herbal formula, APX as an anti-cachexia agent; the effect of APX was superior to that of megestrol acetate overall especially for muscle atrophy. The underlying mechanisms of this herbal formula may involve the modulation of muscle atrophy-promoting molecules including p38, NFκB, TNF-α and TWEAK.


Subject(s)
Astragalus propinquus , Cachexia/prevention & control , Colonic Neoplasms/drug therapy , Muscle, Skeletal/drug effects , Muscular Atrophy/prevention & control , Paeonia , Plant Extracts/pharmacology , Animals , Astragalus propinquus/chemistry , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Cell Line, Tumor , Colonic Neoplasms/complications , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokine TWEAK/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred BALB C , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , NF-kappa B/metabolism , Paeonia/chemistry , Plant Extracts/isolation & purification , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Cancer Res Ther ; 16(6): 1210-1214, 2020.
Article in English | MEDLINE | ID: mdl-33342775

ABSTRACT

Anorexia and cachexia are major clinical problems seen in a large proportion of patients with advanced cancer. Weight loss has also been identified as an indicator of poor prognosis in cancer patients. Around 20% of patients with advanced cancer present mortality from the effects of malnutrition rather than from cancer itself. Early nutrition intervention has seen to improve outcomes in cancer patients such as weight gain, treatment tolerance, and improved quality of life (QoL). Effective therapies for addressing these threatening conditions are lacking. Pharmacotherapeutic agents such as corticosteroids, megestrol acetate, and cyproheptadine have several adverse reactions and also lack satisfactory results. Rasayana therapy is known to prevent loss of body mass and at the same time help to improve appetite and increase patient's QoL. The Rasayana compound used by us to prevent cachexia mainly includes swarna sindoor, Hirak bhasma, and suvarna bhasma. To evaluate benefits of Rasayana therapy on these variables, we maintain complete documentation of different clinical variables in all cancer patients. Here, in this observational study, we analyzed the data collected from a group of stage IV breast cancer patients (n = 30) receiving Rasayana therapy. Patients were followed at an interval of every 15 days from baseline for 3 months. Furthermore, at each visit, there weight was recorded on calibrated digital weight balance. QoL in these patients was recorded at quarterly interval using functional assessment of cancer therapies questionnaire. It was seen that in the duration of 3 months patients appetite increased significantly (P = 0.03). Significant weight gain was seen in patients (P = 0.04). Significant improvement was also seen in QoL especially related to QoL subdomains of physical wellbeing (P = 0.01), emotional wellbeing (P < 0.04), and functional wellbeing (P < 0.001). Rasayana therapy was seen to be well tolerated by all patients.


Subject(s)
Anorexia/prevention & control , Breast Neoplasms/metabolism , Breast Neoplasms/microbiology , Cachexia/prevention & control , Medicine, Ayurvedic/methods , Anorexia/etiology , Appetite/drug effects , Breast Neoplasms/pathology , Cachexia/etiology , Female , Humans , Middle Aged , Quality of Life , Retrospective Studies , Surveys and Questionnaires , Weight Gain/drug effects
13.
Nutrients ; 12(10)2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33053632

ABSTRACT

In cancer patients, loss of muscle mass is significantly associated with low tolerability of chemotherapy and poor survival. Despite the great strides in the treatment of cancer, targeted therapies such as tyrosine kinase inhibitors (TKIs) could exacerbate muscle wasting. Over recent years, the impact of skeletal muscle loss during TKI therapy on clinical outcomes has been in the spotlight. In this review, we focus on the different molecular pathways of TKIs potentially involved in muscle wasting. Then, we report the results of the studies assessing the effects of different TKI therapies-such as sorafenib, regorafenib, sunitinib, and lenvatinib-on muscle mass, and highlight their potential clinical implications. Finally, we discuss an integrative nutritional approach to be adopted during TKI treatment. The assessment of muscle mass from computerized tomography imaging could be helpful in predicting toxicity and prognosis in patients treated with TKI such as sorafenib. Early recognition of low muscle mass and effective personalized nutritional support could prevent or attenuate muscle mass wasting. However, the role of nutrition is still overlooked, and future clinical trials are needed to find the optimal nutritional support to countermeasure muscle mass depletion during TKI therapy.


Subject(s)
Cachexia/diet therapy , Cachexia/prevention & control , Muscle, Skeletal/drug effects , Nutrition Assessment , Protein Kinase Inhibitors/therapeutic use , Humans , Molecular Targeted Therapy , Multicenter Studies as Topic , Muscle, Skeletal/metabolism , Neoplasms/drug therapy , Nutritional Status , Phenylurea Compounds/therapeutic use , Pyridines/therapeutic use , Quinolines/therapeutic use , Randomized Controlled Trials as Topic , Recommended Dietary Allowances , Sorafenib/therapeutic use , Sunitinib/therapeutic use , Treatment Outcome
14.
Biomed Res ; 41(4): 179-186, 2020.
Article in English | MEDLINE | ID: mdl-32801267

ABSTRACT

Cachexia is a multifactorial condition characterized by muscle mass loss and induces metabolic dysfunction of the skeletal muscles. The preventive effects of medium-chain triglycerides (MCT) supplementation on the oxidative capacity in skeletal muscle under cachectic condition were investigated in the present study. ICR mice were randomly divided into four groups; control, lipopolysaccharide (LPS), LPS plus long-chain triglycerides (LCT) and LPS plus MCT supplementation. LCT and MCT oil were administered to the LPS + LCT and LPS + MCT groups orally (5.0 g/kg body weight/day) by a catheter for one week. Cachexia was induced in the LPS, LPS + LCT, and LPS + MCT groups via LPS injection (7.5 mg/kg body weight, i.p.) after the supplementation. LPS induced a reduction of ketone bodies concentration in blood plasma. LPS also induced a decrease in succinate dehydrogenase activity and PGC-1α expression level in tibialis anterior muscles. Meanwhile, MCT supplementation suppressed a decrease in ketone bodies concentration and succinate dehydrogenase activity. In addition, MCT supplementation increased the level of citrate synthase activity in the muscles. These results suggested the preventive effect of MCT supplementation on oxidative capacity in skeletal muscle and the involvements of ketone bodies regulation under cachectic condition.


Subject(s)
Cachexia/prevention & control , Dietary Supplements , Lipopolysaccharides/antagonists & inhibitors , Muscle, Skeletal/drug effects , Triglycerides/pharmacology , Animals , Body Weight/drug effects , Cachexia/genetics , Cachexia/metabolism , Cachexia/pathology , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/metabolism , Gene Expression Regulation , Ketone Bodies/blood , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred ICR , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Triglycerides/chemistry
15.
J Ethnopharmacol ; 260: 113066, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32505837

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Salvia miltiorrhiza bunge (Danshen) has been extensively used to treat a wide variety of diseases including cancers. Cryptotanshinone is a major lipophilic compound extracted from the root of Danshen and has been reported to exert various pharmacological effects, however, its anti-cachectic remains unknown. AIM OF THE STUDY: The present study aims to investigate the anti-cachectic efficacy of cryptotanshinone and elucidate the underlying mechanism. MATERIALS AND METHODS: Prevention of muscle wasting by cryptotanshinone in colon adenocarcinoma CT26-induced cachexia and CT26 conditioned medium (TCM)-induced myotubes were investigated. Main features of cancer cachexia were determined after cryptotanshinone administration. The therapeutic effect of cryptotanshinone on myotube atrophy was assessed by morphological observation and myotube fiber width determination. E3 ubiquitin ligases muscle RING-finger containing protein 1 (MuRF1) and muscle atrophy Fbox protein (MAFbx/Atrogin-1) expression and STAT3 activation were examined using western blot, real-time qPCR and dual-luciferase reporter gene assays both in vitro and in vivo. The myotubes were infected with lentiviruses expressing STAT3 or GFP. RESULTS: In CT26 tumor-bearing mice, cryptotanshinone (20 and 60 mg/kg) administration drastically prevented systemic cancer cachexia from whole body weight loss and wasting of multiple tissues including heart, fat and skeletal muscle, with a negligible effect on cancer growth at dose of 20 mg/kg cryptotanshinone administration prevented the induction of MuRF1 and MAFbx/Atrogin-1 in cachectic muscles. Moreover, cryptotanshinone (2.5-10 µM) dose-dependently reduced the elevated expression of MuRF1 and MAFbx/Atrogin-1 in C2C12 myotubes, and improved myotube atrophy. We showed that cryptotanshinone significantly suppressed the hyper-activated STAT3 in cachectic muscles and C2C12 myotubes and inhibited STAT3 transcriptional activity, but it did not repress the activation of STAT1. The inhibitory effect of cryptotanshinone on TCM-induced myotube atrophy was blocked by STAT3 overexpression. CONCLUSIONS: These data suggest that cryptotanshinone prevents muscle wasting in cancer cachexia through STAT3 inhibition, and it may be a promising candidate drug for the treatment of cancer cachexia.


Subject(s)
Adenocarcinoma/prevention & control , Cachexia/prevention & control , Colonic Neoplasms/drug therapy , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy/prevention & control , Phenanthrenes/pharmacology , STAT3 Transcription Factor/metabolism , Adenocarcinoma/complications , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Colonic Neoplasms/complications , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Inbred BALB C , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Phosphorylation , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Weight Loss/drug effects
16.
J Ren Nutr ; 30(5): 440-451, 2020 09.
Article in English | MEDLINE | ID: mdl-32303413

ABSTRACT

OBJECTIVE: The objective of the study was to quantify the loss and arterial blood concentration of the three main classes of amino acids (AAs)-nonessential amino acids (NEAAs), essential amino acids (EAAs), and branched-chain amino acids-as resulting from high-efficiency hemodialysis (HED) and hemodiafiltration (HDF). We moreover aimed to identify the different fates and metabolic effects manifested in patients undergoing hemodialysis and the consequences on body composition and influence of nutritional decline into protein energy wasting. DESIGN AND METHODS: Identical dialysis monitors, membranes, and dialysate/infusate were used to ensure consistency. Ten patients were recruited and randomized to receive treatment with on-line modern HED and HDF. Arterial plasma concentrations of individual AAs were compared in healthy volunteers and patients undergoing hemodialysis, and AA levels outflowing from the dialyzer were evaluated. Baseline AA plasma levels of patients undergoing hemodialysis were compared with findings obtained 1 year later. RESULTS: A severe loss of AA with HED/HDF was confirmed: a marked loss of total AAs (5 g/session) was detected, corresponding to more than 65% of all AAs. With regard to individual AAs, glutamine displayed a consistent increase (+150%), whereas all other AAs decreased after 12 months of HD/HDF. Only a few AAs, such as proline, cysteine, and histidine maintained normal levels. The most severe metabolic consequences may result from losses of EAAs such as valine, leucine, and histidine and from NEAAs including proline, cysteine, and glutamic acid eliciting the onset of hypercatabolism threatening muscle mass loss. CONCLUSION: Dialysis losses, together with the effect of chronic uremia, resulted in a reduction of fundamental EAAs and NEAAs, which progressively led our patients after 12 months to a deterioration of lean mass toward sarcopenia. Therefore, the reintroduction of a correctly balanced AA supplementation in patients undergoing HD to prevent or halt decline of hypercatabolism into cachexia is recommended.


Subject(s)
Amino Acids/blood , Cachexia/prevention & control , Hemodiafiltration/adverse effects , Nutritional Status , Renal Dialysis/adverse effects , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Time Factors
17.
Laryngorhinootologie ; 99(3): 149-154, 2020 03.
Article in German | MEDLINE | ID: mdl-32120438

ABSTRACT

Nutritional effects on cancer occurrence and on treatment outcome in cancer patients may depend on food preferences and on the quantity of foods supplied. However, it has been difficult to reliably show beneficial effects of specific dietary concepts on cancer incidence. On the other hand, obesity as a result of chronic overfeeding has been linked firmly to an increased risk of a number of cancers as well as on cancer recurrence after treatment. Metabolic consequences of obesity and other components of the metabolic syndrome may be responsible for inducing and/or promoting cancer growth and should be antagonized by regular moderate physical activity in healthy subjects and in cancer survivors. During cancer treatment and in patients with advanced disease, inadequate food intake and physical inactivity may lead to malnutrition, while recurrent and chronic systemic inflammatory reactions induce chronic catabolism with a preferential loss of muscle and cell mass, condition referred to as cachexia.


Subject(s)
Cachexia/etiology , Cachexia/prevention & control , Cachexia/therapy , Neoplasms/therapy , Humans , Obesity/epidemiology , Obesity/prevention & control , Obesity/therapy
18.
J Ethnopharmacol ; 255: 112722, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32114165

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Euphorbia tirucalli L. is an African plant that grows well in Brazil. Individuals diagnosed with cancer frequently consume latex from E. tirucalli, dissolved in drinking water. In vitro studies confirm the antitumor potential of E. tirucalli latex, but in vivo evaluations are scarce. AIM OF THE STUDY: To evaluate the effect of intake of an aqueous solution of E. tirucalli latex on tumor growth, cachexia, and immune response in Walker 256 tumor-bearing rats. MATERIALS AND METHODS: Latex from E. tirucalli was collected and analyzed by LC-MS. Sixty male Wistar rats (age, 90 days) were randomly divided into four groups: C, control group (without tumor); W, Walker 256 tumor-bearing group; SW1, W animals but treated with 25 µL latex/mL water; and SW2, W animals but treated with 50 µL latex/mL water. Animals received 1 mL of latex solution once a day by gavage. After 15 d, animals were euthanized, tumor mass was determined, and glucose and triacylglycerol serum levels were measured by using commercial kits. Change in the body weight during tumor development was calculated, and proliferation capacity of tumor cells was assessed by the Alamar Blue assay. Phagocytosis and superoxide anion production by peritoneal macrophages and circulating neutrophils were analyzed by enzymatic and colorimetric assays. Data are analyzed by one-way ANOVA followed by Tukey's post-hoc test, with the significance level set at 5%. RESULTS: The analysis of the latex revealed the presence of triterpenes. The ingestion of the latex aqueous solution promoted 40% and 60% reduction of the tumor mass in SW1 and SW2 groups, respectively (p < 0.05). The proliferative capacity of tumor cells from SW2 group was 76% lower than that of cells from W group (p < 0.0001). Animals treated with latex gained, on average, 20 g (SW1) and 8 g (SW2) weight. Glucose and triacylglycerol serum levels in SW1 and SW2 animals were similar to those in C group rats. Peritoneal macrophages and blood neutrophils from SW1 and SW2 animals produced 30-40% less superoxide anions than those from W group animals (p < 0.05), but neutrophils from SW2 group showed an increased phagocytic capacity (20%, vs. W group). CONCLUSIONS: E. tirucalli latex, administered orally for 15 d, efficiently reduced tumor growth and cachexia in Walker 256 tumor-bearing rats. Decreased tumor cell proliferative capacity was one of the mechanisms involved in this effect. Further, the data suggest immunomodulatory properties of E. tirucalli latex. The results agree with folk data on the antitumor effect of latex ingestion, indicating that it may be useful as an adjunct in the treatment of cancer patients. For this, further in vivo studies in animal and human models need to be conducted.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cachexia/prevention & control , Carcinoma 256, Walker/drug therapy , Euphorbia , Latex/pharmacology , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Cachexia/blood , Cachexia/immunology , Cachexia/physiopathology , Carcinoma 256, Walker/pathology , Cell Proliferation/drug effects , Cells, Cultured , Euphorbia/chemistry , Latex/isolation & purification , Macrophages/drug effects , Macrophages/immunology , Male , Neutrophils/drug effects , Neutrophils/immunology , Plant Extracts/isolation & purification , Rats, Wistar , Triglycerides/blood , Tumor Burden/drug effects , Weight Loss/drug effects
19.
Nutrition ; 67-68: 110541, 2019.
Article in English | MEDLINE | ID: mdl-31470259

ABSTRACT

Weight loss and functional decline is a common and detrimental consequence of cancer. The interventions that are offered to patients with weight loss and functional decline often seem haphazard and varying from center to center. The lack of stringent management is probably based both on lack of knowledge of existing treatment guidelines and the current weak level of evidence of clinical effects of different nutritional and exercise interventions. Some studies evaluated multimodal interventions with various treatment combinations, including nutrition and exercise, that report clinically significant effects on cachexia outcomes. As of today, however, there is a paucity of large randomized controlled trials that incorporate both a fully structured exercise program and a well-described nutritional intervention. Studies investigating combinations of several interventions in patients with active cancer and risk for losing weight are too few and too heterogeneous to enable firm conclusions about effect, optimal dose, or timing of interventions. However, data presented in this review suggest an overall benefit, especially if interventions are started before weight loss and loss of function become too severe. Thus, the aim of this review was to examine the evidence for combined treatments targeting weight loss in cancer patients.


Subject(s)
Cachexia/prevention & control , Exercise Therapy/methods , Neoplasms/complications , Nutrition Therapy/methods , Cachexia/etiology , Combined Modality Therapy , Exercise , Humans , Neoplasms/physiopathology , Nutritional Status , Risk Factors , Treatment Outcome , Weight Loss
20.
Med Hypotheses ; 132: 109326, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31421423

ABSTRACT

The mechanisms underlying cancer cachexia - the proximate cause of at least 20% of cancer-related deaths - have until recently remained rather obscure. New research, however, clarifies that cancers evoking cachexia release microvesicles rich in heat shock proteins 70 and 90, and that these extracellular heat shock proteins induce cachexia by serving as agonists for toll-like receptor 4 (TLR4) in skeletal muscle, macrophages, and adipocytes. Hence, safe nutraceutical measures which can down-regulate TLR4 signaling can be expected to aid prevention and control of cancer cachexia. There is reason to suspect that phycocyanobilin, ferulic acid, glycine, long-chain omega-3s, green tea catechins, ß-hydroxy-ß-methylbutyrate, carnitine, and high-dose biotin may have some utility in this regard.


Subject(s)
Adipocytes/metabolism , Cachexia/prevention & control , Dietary Supplements , Neoplasms/pathology , Signal Transduction , Toll-Like Receptor 4/metabolism , 3-Hydroxybutyric Acid/metabolism , Biotin/metabolism , Cachexia/metabolism , Carnitine/metabolism , Catechin/metabolism , Coumaric Acids/metabolism , Fatty Acids, Omega-3/metabolism , Glycine/metabolism , Humans , Macrophages/metabolism , Muscle, Skeletal/metabolism , Phycobilins/metabolism , Phycocyanin/metabolism , Tea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL