Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hetanshi Naik is active.

Publication


Featured researches published by Hetanshi Naik.


The New England Journal of Medicine | 2015

Afamelanotide for Erythropoietic Protoporphyria

Janneke G. Langendonk; Manisha Balwani; Karl E. Anderson; Herbert L. Bonkovsky; Alexander Vincent Anstey; D. M. Bissell; Joseph R. Bloomer; C. Edwards; N. J. Neumann; Corette B. Parker; John D. Phillips; Hw Lim; I. Hamzavi; J. C. Deybach; R. Kauppinen; Lesley E. Rhodes; J. Frank; G.M. Murphy; F. P J Karstens; Eric J.G. Sijbrands; F.W.M. de Rooij; Mark Lebwohl; Hetanshi Naik; Colin R. Goding; J. H. P. Wilson; Robert J. Desnick

BACKGROUND Erythropoietic protoporphyria is a severe photodermatosis that is associated with acute phototoxicity. Patients with this condition have excruciating pain and a markedly reduced quality of life. We evaluated the safety and efficacy of an α-melanocyte-stimulating hormone analogue, afamelanotide, to decrease pain and improve quality of life. METHODS We conducted two multicenter, randomized, double-blind, placebo-controlled trials of subcutaneous implants containing 16 mg of afamelanotide. Patients in the European Union (74 patients) and the United States (94 patients) were randomly assigned, in a 1:1 ratio, to receive a subcutaneous implant containing either afamelanotide or placebo every 60 days (a total of five implants in the European Union study and three in the U.S study). The type and duration of sun exposure, number and severity of phototoxic reactions, and adverse events were recorded over the respective 180-day and 270-day study periods. Quality of life was assessed with the use of validated questionnaires. A subgroup of U.S. patients underwent photoprovocation testing. The primary efficacy end point was the number of hours of direct exposure to sunlight without pain. RESULTS In the U.S. study, the duration of pain-free time after 6 months was longer in the afamelanotide group (median, 69.4 hours, vs. 40.8 hours in the placebo group; P=0.04). In the European Union study, the duration of pain-free time after 9 months was also longer in the afamelanotide group than in the placebo group (median, 6.0 hours vs. 0.8 hours; P=0.005), and the number of phototoxic reactions was lower in the the afamelanotide group (77 vs. 146, P=0.04). In both trials, quality of life improved with afamelanotide therapy. Adverse events were mostly mild; serious adverse events were not thought to be related to the study drug. CONCLUSIONS Afamelanotide had an acceptable side-effect and adverse-event profile and was associated with an increased duration of sun exposure without pain and improved quality of life in patients with erythropoietic protoporphyria. (Funded by Clinuvel Pharmaceuticals and others; ClinicalTrials.gov numbers, NCT01605136 and NCT00979745.).


The American Journal of Medicine | 2014

Acute Porphyrias in the USA: Features of 108 Subjects from Porphyrias Consortium

Herbert L. Bonkovsky; Vinaya Maddukuri; Cemal Yazici; Karl E. Anderson; D. Montgomery Bissell; Joseph R. Bloomer; John D. Phillips; Hetanshi Naik; Inga Peter; Gwen Baillargeon; Krista Bossi; Laura Gandolfo; Carrie Light; David F. Bishop; Robert J. Desnick

BACKGROUND Recent descriptions of the clinical and laboratory features of subjects with acute porphyrias in the US are lacking. Our aim was to describe clinical, biochemical, and genetic features of 108 subjects. METHODS Between September 2010 and December 2012, 108 subjects with acute porphyrias (90 acute intermittent porphyrias, 9 hereditary coproporphyrias, 9 variegate porphyrias) were enrolled into an observational study. Genetic testing was performed at a central genetic testing laboratory and clinical information entered into a central database. Selected features were compared with data for adults in the US. RESULTS Most subjects (88/108, 81%) were female, with self-reported onset of symptoms in the second through fourth decades of life. The most common symptom was abdominal pain. Appendectomies and cholecystectomies were common before a diagnosis of porphyria. The diagnosis was delayed by a mean of 15 years. Anxiety and depression were common, and 18% complained of chronic symptoms, especially neuropathic and other pains. The incidences of systemic arterial hypertension, chronic kidney disease, seizure disorders, and psychiatric conditions were markedly increased. Mutations of the known causative genes were found in 102/105 of those tested, with novel mutations being found in 37, including in 7/8 subjects with hereditary coproporphyria. Therapy with intravenous hematin was the most effective therapy both for treatment of acute attacks and for prevention of recurrent attacks. CONCLUSIONS Acute porphyrias often remain undiagnosed for more than a decade after first symptoms develop. Intravenous hematin is the treatment of choice, both for treatment of acute attacks and for prevention of recurrent attacks.


Molecular Medicine | 2015

Liver transplantation for acute intermittent porphyria: Biochemical and pathologic studies of the explanted liver

Makiko Yasuda; Angelika Ludtke Erwin; Lawrence U Liu; Manisha Balwani; Brenden Chen; Senkottuvelan Kadirvel; Lin Gan; M. Isabel Fie; Ronald E. Gordon; Chunli Yu; Sonia Clavero; Antonios Arvelakis; Hetanshi Naik; L. David Martin; John D. Phillips; Karl E. Anderson; Vaithamanithi M. Sadagoparamanujam; Sander Florman; Robert J. Desnick

Acute intermittent porphyria (AIP) is an autosomal-dominant hepatic disorder caused by the half-normal activity of hydroxymethylbilane (HMB) synthase. Symptomatic individuals experience life-threatening acute neurovisceral attacks that are precipitated by factors that induce the hepatic expression of 5-aminolevulinic acid synthase 1 (ALAS1), resulting in the marked accumulation of the putative neurotoxic porphyrin precursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG). Here, we provide the first detailed description of the biochemical and pathologic alterations in the explanted liver of an AIP patient who underwent orthotopic liver transplantation (OLT) due to untreatable and debilitating chronic attacks. After OLT, the recipient’s plasma and urinary ALA and PBG rapidly normalized, and her attacks immediately stopped. In the explanted liver, (a) ALAS1 mRNA and activity were elevated approximately ∼3- and 5-fold, and ALA and PBG concentrations were increased ∼3- and 1,760-fold, respectively; (b) uroporphyrin III concentration was elevated; (c) microsomal heme content was sufficient, and representative cytochrome P450 activities were essentially normal; (d) HMB synthase activity was approximately half-normal (∼42%); (e) iron concentration was slightly elevated; and (f) heme oxygenase I mRNA was increased approximately three-fold. Notable pathologic findings included nodular regenerative hyperplasia, previously not reported in AIP livers, and minimal iron deposition, despite the large number of hemin infusions received before OLT. These findings suggest that the neurovisceral symptoms of AIP are not associated with generalized hepatic heme deficiency and support the neurotoxicity of ALA and/or PBG. Additionally, they indicate that substrate inhibition of hepatic HMB synthase activity by PBG is not a pathogenic mechanism in acute attacks.


JAMA Dermatology | 2017

Clinical, Biochemical, and Genetic Characterization of North American Patients With Erythropoietic Protoporphyria and X-linked Protoporphyria

Manisha Balwani; Hetanshi Naik; Karl E. Anderson; D. Montgomery Bissell; Joseph R. Bloomer; Herbert L. Bonkovsky; John D. Phillips; Jessica R. Overbey; Bruce Wang; Ashwani K. Singal; Lawrence U Liu; Robert J. Desnick

Importance Autosomal recessive erythropoietic protoporphyria (EPP) and X-linked protoporphyria (XLP) are rare photodermatoses presenting with variable degrees of painful phototoxicity that markedly affects quality of life. The clinical variability, determinants of severity, and genotype/phenotype correlations of these diseases are not well characterized. Objective To describe the baseline clinical characteristics, genotypes, and determinants of disease severity in a large patient cohort with EPP or XLP. Design, Setting, and Participants A prospective observational study was conducted among patients with confirmed diagnoses of EPP or XLP from November 1, 2010, to December 6, 2015, at 6 academic medical centers of the Porphyrias Consortium of the National Institutes of Health Rare Diseases Clinical Research Network. Detailed medical histories, including history of phototoxicity and treatment, were collected on standardized case report forms. Patients underwent baseline laboratory testing, total erythrocyte protoporphyrin (ePPIX) testing, and molecular genetic testing. Data were entered into a centralized database. Main Outcomes and Measures Results of biochemical and genetic tests were explored for association with clinical phenotype in patients with EPP or XLP. Results Of the 226 patients in the study (113 female and 113 male patients; mean [SD] age, 36.7 [17.0] years), 186 (82.3%) had EPP with a FECH (OMIM 612386) mutation and the common low-expression FECH allele IVS3–48T>C, and only 1 patient had 2 FECH mutations. Twenty-two patients had XLP (9.7%; 10 male and 12 female patients), and 9 patients (4.0%) had elevated ePPIX levels and symptoms consistent with protoporphyria but no detectable mutation in the FECH or ALAS2 (OMIM 301300) gene. Samples of DNA could not be obtained from 8 patients. Patients’ mean (SD) age at symptom onset was 4.4 (4.4) years. Anemia (107 [47.3%]), history of liver dysfunction (62 [27.4%]), and gallstones (53 [23.5%]) were commonly reported. Higher ePPIX levels were associated with earlier age of symptom onset (median ePPIX levels for those who developed symptoms before vs after 1 year of age, 1744 vs 1567 µg/dL; P = .02), less sun tolerance (median ePPIX levels for those reporting symptoms before vs after 10 minutes of sun exposure, 2233 vs 1524 µg/dL; P ⩽ .001), and increased risk of liver dysfunction (median ePPIX levels for those with liver dysfunction vs normal liver function, 2016 vs 1510 µg/dL; P = .003). Patients with EPP and FECH missense mutations had significantly lower ePPIX levels than those with other mutations (1462 vs 1702 µg/dL; P = .01). Male patients with XLP had significantly higher ePPIX levels, on average, than did patients with EPP (3574 vs 1669 µg/dL; P < .001). Marked clinical variability was seen in female patients with XLP owing to random X-chromosomal inactivation. Conclusions and Relevance These data suggest that higher ePPIX levels are a major determinant of disease severity and risk of liver dysfunction in patients with EPP or XLP. These findings provide a framework for clinical monitoring and management of these disorders.


Molecular Genetics and Metabolism | 2016

Acute Intermittent Porphyria in children: A case report and review of the literature

Manisha Balwani; Preeti Singh; Anju Seth; Ekta Malik Debnath; Hetanshi Naik; Dana Doheny; Brenden Chen; Makiko Yasuda; Robert J. Desnick

Acute Intermittent Porphyria (AIP), an autosomal dominant inborn error of heme metabolism, typically presents in adulthood, most often in women in the reproductive age group. There are limited reports on the clinical presentation in children, and in contrast to the adults, most of the reported pediatric cases are male. While acute abdominal pain is the most common presenting symptom in children, seizures are commonly seen and may precede the diagnosis of AIP. As an example, we report a 9year old developmentally normal pre-pubertal boy who presented with acute abdominal pain, vomiting and constipation followed by hyponatremia, seizures, weakness and neuropathy. After a diagnostic odyssey, his urine porphobilinogen was found to be significantly elevated and genetic testing showed a previously unreported consensus splice-site mutation IVS4-1G>A in the HMBS gene confirming the diagnosis of AIP. Here, we discuss the clinical presentation in this case, and 15 reported pediatric cases since the last review 30years ago and discuss the differential diagnosis and challenges in making the diagnosis in children. We review the childhood-onset cases reported in the Longitudinal Study of the Porphyrias Consortium. Of these, genetically and biochemically confirmed patients, 11 of 204 (5%) reported onset of attacks in childhood. Most of these patients (91%) reported recurrent attacks following the initial presentation. Thus, AIP should be considered in the differential diagnosis of children presenting with unexplained abdominal pain, seizures, weakness and neuropathy.


Molecular genetics and metabolism reports | 2018

Parkinson's disease prevalence in Fabry disease: A survey study

Adina Wise; Amy Yang; Hetanshi Naik; Chanan Stauffer; Natasha Zeid; Christopher Liong; Manisha Balwani; Robert J. Desnick; Roy N. Alcalay

Recent research has suggested a possible link between Parkinsons disease (PD) and Fabry disease. To test this relationship, we administered a self-report and family history questionnaire to determine the prevalence of PD in Fabry disease patients and family members with likely pathogenic alpha-galactosidase A (GLA) mutations. A total of 90 Fabry patients (77 from the online survey and 13 from the Icahn School of Medicine at Mount Sinai (ISMMS)) were included in the analysis. Two of the Fabry disease patients who completed the online survey were diagnosed with PD (2/90, 2.2%). Among probands older than 60, 8.3% (2/24) were diagnosed with PD. Using Kaplan Meier survival analysis, the age-specific risk of PD by age 70 was 11.1%. Family history was available on 72 Fabry families from the online study and 9 Fabry families from ISMMS. Among these 81 families, 6 (7.4%) had one first degree relative who fit the criteria for a conservative diagnosis of PD. The results of this study suggest that there may be an increased risk of developing PD in individuals with GLA mutations, but these findings should be interpreted with caution given the limitations of the study design.


PLOS ONE | 2016

The D519G Polymorphism of Glyceronephosphate O-Acyltransferase Is a Risk Factor for Familial Porphyria Cutanea Tarda

Colin P. Farrell; Jessica R. Overbey; Hetanshi Naik; Danielle Nance; Gordon D. McLaren; Christine E. McLaren; Luming Zhou; Robert J. Desnick; Charles J. Parker; John D. Phillips

Both familial and sporadic porphyria cutanea tarda (PCT) are iron dependent diseases. Symptoms of PCT resolve when iron stores are depleted by phlebotomy, and a sequence variant of HFE (C282Y, c.843G>A, rs1800562) that enhances iron aborption by reducing hepcidin expression is a risk factor for PCT. Recently, a polymorphic variant (D519G, c.1556A>G, rs11558492) of glyceronephosphate O-acyltransferase (GNPAT) was shown to be enriched in male patients with type I hereditary hemochromatosis (HFE C282Y homozygotes) who presented with a high iron phenotype, suggesting that GNPAT D519G, like HFE C282Y, is a modifier of iron homeostasis that favors iron absorption. To challenge this hypothesis, we investigated the frequency of GNPAT D519G in patients with both familial and sporadic PCT. Patients were screened for GNPAT D519G and allelic variants of HFE (both C282Y and H63D). Nucleotide sequencing of uroporphyrinogen decarboxylase (URO-D) identified mutant alleles. Patients with low erythrocyte URO-D activity or a damaging URO-D variant were classified as familial PCT (fPCT) and those with wild-type URO-D were classified as sporadic PCT (sPCT). GNPAT D519G was significantly enriched in the fPCT patient population (p = 0.0014) but not in the sPCT population (p = 0.4477). Both HFE C282Y and H63D (c.187C>G, rs1799945) were enriched in both PCT patient populations (p<0.0001) but showed no greater association with fPCT than with sPCT. Conclusion: GNPAT D519G is a risk factor for fPCT, but not for sPCT.


67th Annual Meeting of the American Association for the Study of Liver Diseases: The Liver Meeting 2016 | 2016

Sinusoidal and pericellular fibrosis in adult post-transplant liver biopsies: Association with histological changes, hepatic stellate cell activation and impact on patient outcome

Karl E. Anderson; Manisha Balwani; Paolo Ventura; Aneta Ivanova; Joseph R. Bloomer; D. Montgomery Bissell; Ulrich Stölzel; Charles J. Parker; David C. Rees; Penny Stein; Jerzy Windyga; Raili Kaupinnen; Janneke Langendonk; Michael Norman Badminton; Elizabeth Minder; Pavel Martásek; Felix Alegre; Maria Domenica Cappellini; Herbert L. Bonkovsky; Sverre Sandberg; Aasne K. Aarsand Kaarsand; Robert J. Desnick; Jean Charles Deybach; John Phillips; Hetanshi Naik; Eliane Sardh; Pauline Harper; Amy Chan; Chang Heok Soh; Kirsten Mccarthy

Introduction Although the accumulation of hepatic iron occurs during chronic liver injury, its role in fibrogenesis remains poorly understood. To date, studies show that excess iron in hepatocytes promotes formation of reactive oxygen species, which then activate hepatic stellate cells (HSC) to secrete collagen matrix. It is unclear however, whether iron directly affects HSC function. Aim Our aims were to investigate the effects of exogenous iron on core fibrogenic and iron-related genes and proteins in HSCs. Method The GRX murine HSC cell line was treated with the physiologically relevant form of transferrin-bound iron, holotransferrin (0.005, 0.05, 0.5, 2 and 5 g/L) for 24 h, with or without the iron chelator deferoxamine (10 μM). Expression of fibrogenic markers (α-SMA, TGF-β and Serpine-1) were analyzed by qRT-PCR, while ferritin was measured by ELISA. Levels of transferrin receptor (TfR1), TGF-β receptors and p-Smad 2 and 4 were analyzed by western blot, and secreted collagen was measured using the Sircol assay. Results HSCs express the iron-uptake and iron-exporter proteins TfR1 and ferroportin, respectively. Treatment with holotransferrin upregulated the expression of TfR1 by 1.8-fold (p<0.05), led to the accumulation of storage iron (ferritin) by up to 2-fold (p<0.01), and activated HSCs: α-SMA mRNA increased by up to 2-fold (p<0.03), TGF-β mRNA was elevated by 1.6-fold (p=0.05) and Serpine-1 mRNA was raised by 2.5-fold (p<0.05). These were accompanied by a 2-fold increase in secreted collagen (p<0.03), and activation of the TGF-β pathway: increased protein expression of TGF-β R1, TGF-b RII, and p-Smad 2 and 4 (p<0.05). Conversely, the addition of deferoxamine significantly lowered ferritin levels by 30% (p<0.03), repressed fibrogenic genes, α-SMA (0.2-fold; p<0.03), and TGF-β (0.4-fold; <0.03), and reduced collagen secretion by up to 60% (p<0.01). Deferoxamine also inhibited TGF-β signaling, with decreased levels of TGF-β RII and p-Smad 2. Conclusion HSCs express the iron transport proteins and are regulated by levels of exogenous iron. Excess iron activates HSCs and the TGF-β pathway, while iron depletion using deferoxamine attenuates the fibrogenic response, suggesting that iron depletion could be a useful adjunctive therapy in the treatment of individuals with advanced liver disease.


The Journal of Pediatrics | 2018

Diagnostic Delay in Erythropoietic Protoporphyria

Sajel M. Lala; Hetanshi Naik; Manisha Balwani

&NA; Erythropoietic protoporphyria is a photodermatosis presenting in childhood with severe pain on sun exposure. The diagnosis is often delayed because of the lack of awareness among pediatricians. We describe the diagnostic odyssey of 2 children presenting with symptoms of erythropoietic protoporphyria and report results of a survey of 129 affected individuals.


Clinical Chemistry | 2015

Pitfalls in Erythrocyte Protoporphyrin Measurement for Diagnosis and Monitoring of Protoporphyrias

Eric Gou; Manisha Balwani; D. Montgomery Bissell; Joseph R. Bloomer; Herbert L. Bonkovsky; Robert J. Desnick; Hetanshi Naik; John D. Phillips; Ashwani K. Singal; Bruce Wang; Siobán B. Keel; Karl E. Anderson

Collaboration


Dive into the Hetanshi Naik's collaboration.

Top Co-Authors

Avatar

Manisha Balwani

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Robert J. Desnick

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl E. Anderson

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph R. Bloomer

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jessica R. Overbey

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ashwani K. Singal

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge